151
|
Kakkad J, Deshmukh P, Gaurkar S. Cartilage's Contribution in Otology: A Comprehensive Review of Its Role in Ear Surgery. Cureus 2023; 15:e49800. [PMID: 38161551 PMCID: PMC10757830 DOI: 10.7759/cureus.49800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
This comprehensive review thoroughly examines the pivotal role of cartilage in otologic surgery, elucidating its multifaceted contributions to both cosmetic and functional outcomes. From reconstructing the external ear to reinforcing the tympanic membrane and restoring the ossicular chain, cartilage emerges as a versatile and resilient biological material with unique properties that make it an invaluable resource for otologic surgeons. The review explores the nuances of cartilage's applications in various surgical contexts, emphasizing its significance in promoting tissue regeneration and healing. The text delves into advancements in tissue engineering, biodegradable scaffolds, and 3D printing technology, pointing toward a future where more precise and personalized interventions may redefine the landscape of otologic surgery. The convergence of these innovations holds the promise of elevating the standard of care, minimizing complications, and improving the quality of life for patients undergoing cartilage-based otologic procedures. This synthesis of current knowledge and future possibilities provides a valuable resource for otologists, surgeons, and researchers in the dynamic field of otology.
Collapse
Affiliation(s)
- Jasleen Kakkad
- Otolaryngology, Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Prasad Deshmukh
- Otolaryngology, Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sagar Gaurkar
- Otolaryngology, Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
152
|
Hu Y, Kang M, Yin X, Cheng Y, Liu Z, Wei Y, Huang D. High biocompatible polyacrylamide hydrogels fabricated by surface mineralization for subchondral bone tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:2217-2231. [PMID: 37368489 DOI: 10.1080/09205063.2023.2230856] [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: 05/05/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023]
Abstract
The subchondral bone is an important part of cartilage which contains a large amount of hydroxyapatite. The mineral components of subchondral bone is the key factor which determines the biomechanical strength, and then affects the biological function of articular cartilage. Here, a mineralized polyacrylamide (PAM-Mineralized) hydrogel with good ALP activity, cell adhesion and biocompatibility was fabricated for subchondral bone tissue engineering. The micromorphology, composition and mechanical properties of PAM and PAM-Mineralized hydrogels were studied. The PAM hydrogels showed a porous structure, while the PAM-Mineralized hydrogels had well-distributed layers of hydroxyapatite mineralization on the surface. The XRD results show that the characteristic peak of hydroxyapatite (HA) was measured in PAM-Mineralized, indicating that the main component of the mineralized structure formed on the surface of the hydrogel after mineralization is HA. The formation of HA ectively decreased the rate of equilibrium swelling of the PAM hydrogel, with PAM-M reaching swelling equilibrium at 6 h. Meanwhile, compressive strength of PAM-Mineralized hydrogel (moisture state) reached 290 ± 30 kPa, compressive modulus reached 130 ± 4 kPa. PAM-Mineralized hydrogels did not affect the growth and proliferation of MC3T3-E1 cells. Surface mineralization of PAM hydrogel could significantly improve osteogenic differentiation of MC3T3-E1 cells. These results showed that PAM-Mineralized hydrogel could possess potential application in the field of subchondral bone tissue engineering.
Collapse
Affiliation(s)
- Yinchun Hu
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, P.R. China
| | - Min Kang
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
| | - Xiangfei Yin
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
| | - Yizhu Cheng
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
| | - Zexin Liu
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
| | - Yan Wei
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, P.R. China
| | - Di Huang
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, P.R. China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, P.R. China
| |
Collapse
|
153
|
Kumar M, Chauhan M, Verma SK, Biswas A, Ansari A, Mishra A, Sanap SN, Bisen AC, Sashidhara KV, Bhatta RS. Preclinical pharmacokinetic exploration of a novel osteoporotic quinazolinone-benzopyran-indole hybrid (S019-0385) using LC-MS/MS. Xenobiotica 2023; 53:484-497. [PMID: 37787761 DOI: 10.1080/00498254.2023.2265475] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/27/2023] [Indexed: 10/04/2023]
Abstract
1. The current investigation was to develop and validate the LC-MS/MS method in order to analyse the various pharmacokinetic parameters of S019-0385. A sensitive, selective, and robust LC-MS/MS approach was established and validated for measuring S019-0385 in female mice plasma and tissue, using optimal multiple reaction monitoring (MRM) transition m/z 488.25/329.12 on positive mode. On a Waters Symmetry Shield C18 column, the analyte was separated using acetonitrile and deionised water with formic acid within 6 min at 0.7 mL/min. Linearity (R2 ≥ 0.99) was observed across 0.195-100 ng/mL concentration range using linear least-squares regression.2. Blood-to-plasma ratio and plasma protein drug binding (%) in mice and human was assessed and found to be less than 1 and >83%, respectively. Absolute bioavailability (%F) of S019-0385 in female Swiss mice was exhibited to be 6.90%. Percent dose excreted S019-0385 in unchanged form through urine and faecal was found to be less than 2% and 0.5%, respectively.3. Following oral administration at 5 mg/kg, the concentration of S019-0385 in tissue distribution was found to be in the order of C small intestine > C bone > C lung > C spleen > C kidney > C liver > C heart > C brain.
Collapse
Affiliation(s)
- Mukesh Kumar
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Mridula Chauhan
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sarvesh Kumar Verma
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Arpon Biswas
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Alisha Ansari
- Academy of Scientific and Innovative Research, Ghaziabad, India
- Division of medicinal and process chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Anjali Mishra
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Sachin Nashik Sanap
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Amol Chhatrapati Bisen
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Koneni V Sashidhara
- Division of medicinal and process chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rabi Sankar Bhatta
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
154
|
Jirofti N, Hashemi M, Moradi A, Kalalinia F. Fabrication and characterization of 3D printing biocompatible crocin-loaded chitosan/collagen/hydroxyapatite-based scaffolds for bone tissue engineering applications. Int J Biol Macromol 2023; 252:126279. [PMID: 37572811 DOI: 10.1016/j.ijbiomac.2023.126279] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
INTRODUCTION Crocin (Cro) is a bioactive biomaterial with properties that promote osteoconduction, osteoinduction, and osteogenic differentiation, making it an ideal candidate for developing mechanically enhanced scaffolds for bone tissue engineering (BTE). Present study focused on a 3D printing matrix loaded with Cro and featuring a composite structure consisting of Chitosan (CH), collagen (Col), and hydroxyapatite (HA). METHOD The scaffolds' structural properties were analyzed using FESEM, and FTIR DSC, while the degradation rate, swelling ratio, cell viability were examined to determine their in vitro performance. Additionally, the scaffolds' mechanical properties were calculated by examining their force, stress, elongation, and Young's modulus. RESULTS The CH/Col/nHA scaffolds demonstrated interconnected porous structures. The cell study results indicated that the Cro-loaded in scaffolds cause to reduce the toxicity of Cro. Biocompatibility was confirmed through degradation rate, swelling ratio parameters, and contact angle measurements for all structures. The addition of Cro showed a significant impact on the strength of the fabricated scaffolds. By loading 25 and 50 μl of Cro, the Young's modulus improved by 71 % and 74 %, respectively, compared to the free drug scaffold. CONCLUSION The obtained results indicated that the 3D printing crocin-loaded scaffolds based chitosan/collagen/hydroxyapatite structure can be introduced as a promising candidate for BTE applications.
Collapse
Affiliation(s)
- Nafiseh Jirofti
- Orthopedic Research Center, Department of Orthopedic Surgery,Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran; Bone and Joint Research Laboratory, Ghaem Hospital, Mashhad University of Medical Science, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Departments of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ali Moradi
- Orthopedic Research Center, Department of Orthopedic Surgery,Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran; Bone and Joint Research Laboratory, Ghaem Hospital, Mashhad University of Medical Science, Mashhad, Iran.
| | - Fatemeh Kalalinia
- Bone and Joint Research Laboratory, Ghaem Hospital, Mashhad University of Medical Science, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
155
|
Dalavi PA, Prabhu A, M S, Murugan SS, Jayachandran V. Casein-assisted exfoliation of tungsten disulfide nanosheets for biomedical applications. Colloids Surf B Biointerfaces 2023; 232:113595. [PMID: 37913705 DOI: 10.1016/j.colsurfb.2023.113595] [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: 07/24/2023] [Revised: 10/14/2023] [Accepted: 10/14/2023] [Indexed: 11/03/2023]
Abstract
Our regular life can be more challenging by bone abnormalities. Bone tissue engineering is used for repairing, regenerating, or replacing bone tissue that has been injured or infected. It is effective in overcoming the drawbacks of conventional bone grafting methods like autograft and allograft by enhancing the effectiveness of bone regeneration. Recent discoveries have shown that the exfoliation of transition metal dichalcogenides (TMDs) with protein is in great demand for bone tissue engineering applications. WS2 nanosheets were developed using casein and subsequently characterized with different analytical techniques. Strong absorption peaks were observed in the UV-visible spectra at 520 nm and 630 nm. Alginate and alginate-casein WS2 microspheres were developed. Stereomicroscopic images of the microspheres are spherical in shape and have an average diameter of around 0.8 ± 0.2 mm. The alginate-casein WS2 microspheres show higher content of water absorption and retention properties than only alginate-containing microspheres. The apatite formation in the simulated bodily fluid solution was facilitated more effectively by the alginate-casein-WS2 microspheres. Additionally, alginate-casein-WS2 microspheres have a compressive strength is 58.01 ± 4 MPa. Finally, in vitro cell interaction studies reveals that both the microspheres are biocompatible with the C3H10T1/2 cells, and alginate-casein-WS2-based microspheres promote cell growth more significantly. Alginate-casein-WS2 microspheres promote alkaline phosphatase activity, and mineralization process. Additionally, alginate-casein-WS2-based microspheres exponentially enhance the genes for ALP, BMP-2, OCN, and Collage type-1. The produced alginate-casein-WS2 microspheres could be a suitable synthetic graft for a bone transplant replacement.
Collapse
Affiliation(s)
- Pandurang Appana Dalavi
- Biomaterials Research Laboratory, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Ashwini Prabhu
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Sajida M
- Biomaterials Research Laboratory, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Sesha Subramanian Murugan
- Biomaterials Research Laboratory, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Venkatesan Jayachandran
- Biomaterials Research Laboratory, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| |
Collapse
|
156
|
Syahruddin MH, Anggraeni R, Ana ID. A microfluidic organ-on-a-chip: into the next decade of bone tissue engineering applied in dentistry. Future Sci OA 2023; 9:FSO902. [PMID: 37753360 PMCID: PMC10518836 DOI: 10.2144/fsoa-2023-0061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
A comprehensive understanding of the complex physiological and pathological processes associated with alveolar bones, their responses to different therapeutics strategies, and cell interactions with biomaterial becomes necessary in precisely treating patients with severe progressive periodontitis, as a bone-related issue in dentistry. However, existing monolayer cell culture or pre-clinical models have been unable to mimic the complex physiological, pathological and regeneration processes in the bone microenvironment in response to different therapeutic strategies. In this point, 'organ-on-a-chip' (OOAC) technology, specifically 'alveolar-bone-on-a-chip', is expected to resolve the problems by better imitating infection site microenvironment and microphysiology within the oral tissues. The OOAC technology is assessed in this study toward better approaches in disease modeling and better therapeutics strategy for bone tissue engineering applied in dentistry.
Collapse
Affiliation(s)
- Muhammad Hidayat Syahruddin
- Postgraduate Student, Dental Science Doctoral Study Program, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Rahmi Anggraeni
- Research Center for Preclinical & Clinical Medicine, National Research & Innovation Agency of the Republic of Indonesia, Cibinong Science Center, Bogor, 16915, Indonesia
- Research Collaboration Center for Biomedical Scaffolds, National Research & Innovation Agency (BRIN) – Universitas Gadjah Mada (UGM), Yogyakarta, 55281, Indonesia
| | - Ika Dewi Ana
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
- Research Collaboration Center for Biomedical Scaffolds, National Research & Innovation Agency (BRIN) – Universitas Gadjah Mada (UGM), Yogyakarta, 55281, Indonesia
| |
Collapse
|
157
|
Abdulaziz D, Anastasiou AD, Panagiotopoulou V, Raif EM, Giannoudis PV, Jha A. Physiologically engineered porous titanium/brushite scaffolds for critical-size bone defects: A design and manufacturing study. J Mech Behav Biomed Mater 2023; 148:106223. [PMID: 37976684 DOI: 10.1016/j.jmbbm.2023.106223] [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: 09/13/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Repairing critical-size bone defects still represents a critical clinical challenge in the field of trauma surgery. This study focuses on a physiological design and manufacturing of porous composite scaffold (titanium Ti with 10 % mole iron doped brushite DCPD-Fe3+) which can mimic the biomechanical properties of natural cortical bone, specifically for the purpose of repairing critical-size defects. To achieve this, the principle of design of experiments (DOE) was applied for investigating the impact of sintering temperature, mineral ratio, and volume fraction of porosity on the mechanical properties of the fabricated scaffolds. The fabricated scaffolds had open porosity up to 60 %, with pore size approximately between 100 μm and 850 μm. The stiffness of the porous composite scaffolds varied between 3.30 GPa and 20.50 GPa, while the compressive strength ranged from approximately 130 MPa-165 MPa at sintering temperatures equal to or exceeding 1000 °C. Scaffolds with higher porosity and mineral content demonstrated lower stiffness values, resembling natural bone. Numerical simulation was employed by Ansys Workbench to investigate the stress and strain distribution of a critical size defect in mid-shaft femur which was designed to be replaced with the fabricated scaffold. The fabricated scaffolds showed flexible biomechanical behaviour at the bone/scaffold interface, generating lower stress levels and indicating a better match with the femoral shaft stiffness. The experimental and numerical findings demonstrated promising applications for manufacturing a patient-specific bone scaffold for critical and potentially large defects for reducing stress shielding and minimizing non-union risk.
Collapse
Affiliation(s)
- Dina Abdulaziz
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, UK.
| | - Antonios D Anastasiou
- Department of Chemical Engineering, University of Manchester, Manchester, M1 3AL, UK
| | | | - El Mostafa Raif
- Faculty of Medicine and Health, School of Dentistry, University of Leeds, Leeds, LS2 9JT, UK
| | - Peter V Giannoudis
- Academic Department of Trauma and Orthopaedic Surgery, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Animesh Jha
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
158
|
Tikkala S, Tirkkonen K, Ekman E, Lehtimäki K. Experience with Tissue Bank Services in 2014 and 2020 in Turku, Finland. Transplant Proc 2023; 55:2345-2353. [PMID: 37891018 DOI: 10.1016/j.transproceed.2023.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND The objective of a musculoskeletal tissue bank is to collect, test, store, and provide musculoskeletal tissue allografts required in orthopedic procedures. Strict exclusion criteria are followed when selecting suitable cadaver musculoskeletal tissue donors, and the allografts are procured under sterile conditions to avoid bacterial contamination. Tissue banking in Turku, Finland, began in 1972, and tissue bank services were last reviewed in 2003. This study aimed to review the operation of the musculoskeletal tissue bank in Turku, Finland, between 2014 and 2020 and to analyze the number, types, and contamination rate of the allografts procured from the cadaver donors. Potential donor-related factors causing bacterial contamination of the allografts and whether potential musculoskeletal tissue donors were overlooked among multiorgan donors were also studied. METHODS A retrospective review of all cadaver musculoskeletal tissue donors used in the Hospital District of Southwest Finland Tyks Orto Musculoskeletal Tissue Bank during the study period was conducted, and data on the procured allograft was collected and presented. The donors were selected among patients treated in the intensive care unit (ICU) of Turku University Hospital (TYKS). RESULTS A total of 28 cadaver donors were used, and 636 allografts were procured between 2014 and 2020. The bacterial contamination rate was 2.5%, which was lower than that in the previous international literature. The median treatment time in the ICU was significantly longer, and the median value of the highest C-reactive protein level was significantly higher in the group of donors with positive allograft bacterial cultures. CONCLUSIONS The bacterial contamination rate in the tissue bank was low on an international scale. Some suitable musculoskeletal tissue donors were overlooked among multiorgan donors.
Collapse
Affiliation(s)
| | - Kari Tirkkonen
- Department of Orthopaedic Surgery, Turku University Hospital, Turku, Finland
| | - Elina Ekman
- Department of Orthopaedic Surgery, Turku University Hospital, Turku, Finland
| | - Kaisa Lehtimäki
- Department of Orthopaedic Surgery, Turku University Hospital, Turku, Finland
| |
Collapse
|
159
|
Chaaban M, Moya A, García-García A, Paillaud R, Schaller R, Klein T, Power L, Buczak K, Schmidt A, Kappos E, Ismail T, Schaefer DJ, Martin I, Scherberich A. Harnessing human adipose-derived stromal cell chondrogenesis in vitro for enhanced endochondral ossification. Biomaterials 2023; 303:122387. [PMID: 37977007 DOI: 10.1016/j.biomaterials.2023.122387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/19/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
Endochondral ossification (ECO), the major ossification process during embryogenesis and bone repair, involves the formation of a cartilaginous template remodelled into a functional bone organ. Adipose-derived stromal cells (ASC), non-skeletal multipotent progenitors from the stromal vascular fraction (SVF) of human adipose tissue, were shown to recapitulate ECO and generate bone organs in vivo when primed into a hypertrophic cartilage tissue (HCT) in vitro. However, the reproducibility of ECO was limited and the major triggers remain unknown. We studied the effect of the expansion of cells and maturation of HCT on the induction of the ECO process. SVF cells or expanded ASC were seeded onto collagen sponges, cultured in chondrogenic medium for 3-6 weeks and implanted ectopically in nude mice to evaluate their bone-forming capacities. SVF cells from all tested donors formed mature HCT in 3 weeks whereas ASC needed 4-5 weeks. A longer induction increased the degree of maturation of the HCT, with a gradually denser cartilaginous matrix and increased mineralization. This degree of maturation was highly predictive of their bone-forming capacity in vivo, with ECO achieved only for an intermediate maturation degree. In parallel, expanding ASC also resulted in an enrichment of the stromal fraction characterized by a rapid change of their proteomic profile from a quiescent to a proliferative state. Inducing quiescence rescued their chondrogenic potential. Our findings emphasize the role of monolayer expansion and chondrogenic maturation degree of ASC on ECO and provides a simple, yet reproducible and effective approach for bone formation to be tested in specific clinical models.
Collapse
Affiliation(s)
- Mansoor Chaaban
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Adrien Moya
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andres García-García
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Robert Paillaud
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Romain Schaller
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Thibaut Klein
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Laura Power
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Katarzyna Buczak
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Elisabeth Kappos
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Tarek Ismail
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
160
|
Wang X, Huang S, Peng Q. Metal Ion-Doped Hydroxyapatite-Based Materials for Bone Defect Restoration. Bioengineering (Basel) 2023; 10:1367. [PMID: 38135958 PMCID: PMC10741145 DOI: 10.3390/bioengineering10121367] [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: 09/20/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
Hydroxyapatite (HA)-based materials are widely used in the bone defect restoration field due to their stable physical properties, good biocompatibility, and bone induction potential. To further improve their performance with extra functions such as antibacterial activity, various kinds of metal ion-doped HA-based materials have been proposed and synthesized. This paper offered a comprehensive review of metal ion-doped HA-based materials for bone defect restoration based on the introduction of the physicochemical characteristics of HA followed by the synthesis methods, properties, and applications of different kinds of metal ion (Ag+, Zn2+, Mg2+, Sr2+, Sm3+, and Ce3+)-doped HA-based materials. In addition, the underlying challenges for bone defect restoration using these materials and potential solutions were discussed.
Collapse
Affiliation(s)
- Xuan Wang
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China;
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Shan Huang
- Changsha Health Vocational College, Changsha 410100, China;
| | - Qian Peng
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China;
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| |
Collapse
|
161
|
Choi CE, Chakraborty A, Adzija H, Shamiya Y, Hijazi K, Coyle A, Rizkalla A, Holdsworth DW, Paul A. Metal Organic Framework-Incorporated Three-Dimensional (3D) Bio-Printable Hydrogels to Facilitate Bone Repair: Preparation and In Vitro Bioactivity Analysis. Gels 2023; 9:923. [PMID: 38131909 PMCID: PMC10742699 DOI: 10.3390/gels9120923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/15/2023] [Accepted: 11/18/2023] [Indexed: 12/23/2023] Open
Abstract
Hydrogels are three-dimensional (3D) water-swellable polymeric matrices that are used extensively in tissue engineering and drug delivery. Hydrogels can be conformed into any desirable shape using 3D bio-printing, making them suitable for personalized treatment. Among the different 3D bio-printing techniques, digital light processing (DLP)-based printing offers the advantage of quickly fabricating high resolution structures, reducing the chances of cell damage during the printing process. Here, we have used DLP to 3D bio-print biocompatible gelatin methacrylate (GelMA) scaffolds intended for bone repair. GelMA is biocompatible, biodegradable, has integrin binding motifs that promote cell adhesion, and can be crosslinked easily to form hydrogels. However, GelMA on its own is incapable of promoting bone repair and must be supplemented with pharmaceutical molecules or growth factors, which can be toxic or expensive. To overcome this limitation, we introduced zinc-based metal-organic framework (MOF) nanoparticles into GelMA that can promote osteogenic differentiation, providing safer and more affordable alternatives to traditional methods. Incorporation of this nanoparticle into GelMA hydrogel has demonstrated significant improvement across multiple aspects, including bio-printability, and favorable mechanical properties (showing a significant increase in the compressive modulus from 52.14 ± 19.42 kPa to 128.13 ± 19.46 kPa with the addition of ZIF-8 nanoparticles). The designed nanocomposite hydrogels can also sustain drug (vancomycin) release (maximum 87.52 ± 1.6% cumulative amount) and exhibit a remarkable ability to differentiate human adipose-derived mesenchymal stem cells toward the osteogenic lineage. Furthermore, the formulated MOF-integrated nanocomposite hydrogel offers the unique capability to coat metallic implants intended for bone healing. Overall, the remarkable printability and coating ability displayed by the nanocomposite hydrogel presents itself as a promising candidate for drug delivery, cell delivery and bone tissue engineering applications.
Collapse
Affiliation(s)
- Cho-E Choi
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
| | - Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
- Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, ON N6A 5B9, Canada
| | - Hailey Adzija
- Department of Chemistry, The University of Western Ontario, London, ON N6A 5B9, Canada
| | - Yasmeen Shamiya
- Department of Chemistry, The University of Western Ontario, London, ON N6A 5B9, Canada
| | - Khaled Hijazi
- Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, ON N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
| | - Ali Coyle
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
| | - Amin Rizkalla
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
- Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, ON N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
- Department of Medical Biophysics, The University of Western Ontario, London, ON N6A 5B9, Canada
- Dentistry, The University of Western Ontario, London, ON N5A 5B9, Canada
| | - David W. Holdsworth
- Department of Medical Biophysics, The University of Western Ontario, London, ON N6A 5B9, Canada
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
- Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, ON N6A 5B9, Canada
- Department of Chemistry, The University of Western Ontario, London, ON N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
| |
Collapse
|
162
|
Luo X, Niu J, Su G, Zhou L, Zhang X, Liu Y, Wang Q, Sun N. Research progress of biomimetic materials in oral medicine. J Biol Eng 2023; 17:72. [PMID: 37996886 PMCID: PMC10668381 DOI: 10.1186/s13036-023-00382-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/02/2023] [Indexed: 11/25/2023] Open
Abstract
Biomimetic materials are able to mimic the structure and functional properties of native tissues especially natural oral tissues. They have attracted growing attention for their potential to achieve configurable and functional reconstruction in oral medicine. Though tremendous progress has been made regarding biomimetic materials, significant challenges still remain in terms of controversy on the mechanism of tooth tissue regeneration, lack of options for manufacturing such materials and insufficiency of in vivo experimental tests in related fields. In this review, the biomimetic materials used in oral medicine are summarized systematically, including tooth defect, tooth loss, periodontal diseases and maxillofacial bone defect. Various theoretical foundations of biomimetic materials research are reviewed, introducing the current and pertinent results. The benefits and limitations of these materials are summed up at the same time. Finally, challenges and potential of this field are discussed. This review provides the framework and support for further research in addition to giving a generally novel and fundamental basis for the utilization of biomimetic materials in the future.
Collapse
Affiliation(s)
- Xinyu Luo
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, 110001, China
| | - Jiayue Niu
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, 110001, China
| | - Guanyu Su
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, 110001, China
| | - Linxi Zhou
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China.
- National Center for Stomatology, Shanghai, 200011, China.
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
| | - Xue Zhang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, 110001, China
| | - Ying Liu
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, 110001, China
| | - Qiang Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, 110001, China
| | - Ningning Sun
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, 110001, China.
| |
Collapse
|
163
|
Yaralı Çevik ZB, Karaman O, Topaloğlu N. Investigation of the optimal light parameters for photobiomodulation to induce osteogenic differentiation of the human bone marrow stem cell and human umbilical vein endothelial cell co-culture. Lasers Med Sci 2023; 38:273. [PMID: 37991573 DOI: 10.1007/s10103-023-03941-9] [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: 03/10/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023]
Abstract
Bones have an important role in the human body with their complex nature. Mesenchymal stem cells and endothelial cells together support their unique and complex nature. Photobiomodulation (PBM) is a promising method that provides cell proliferation, osteogenic differentiation, and bone regeneration. However, there are still unknowns in the mechanism of osteogenic differentiation induced by PBM. The main aim of the study is to understand the molecular mechanism of PBM at 655 and 808 nm of wavelengths and identify the most effective energy densities of both wavelengths for osteogenic differentiation. The effect of PBM on osteogenic differentiation of Human Bone Marrow Stem Cell (hBMSC) and Human Umbilical Vein Endothelial Cell (HUVEC) co-culture was examined at 1, 3, and 5 J/cm2 energy densities of red and near-infrared light through different analysis such as cell viability, scratch assay, intracellular reactive oxygen species production, and ATP synthesis, nitric oxide release, temperature monitoring, and osteogenic differentiation analyses. Even though all PBM-treated groups exhibited better results compared to the control group, 5 J/cm2 energy density induced faster cell proliferation and migration at both wavelengths. The increases in ATP and NO levels as signaling molecules, and the increases in DNA, ALPase, and calcium contents as osteogenic markers were higher in the groups treated with 5 J/cm2 energy density at both wavelengths. Only a slight change was obtained in the level of intracellular ROS after any light applications. It can be concluded that NO release has a very important role together with ATP production in PBM therapy to trigger DNA synthesis, ALPase activity, and mineralization for osteogenic differentiation of the hBMSC and HUVEC co-culture at 655 and 808 nm of wavelengths.
Collapse
Affiliation(s)
- Ziyşan Buse Yaralı Çevik
- Biomedical Test Calibration Application and Research Center, Izmir Katip Celebi University, Çiğli, Izmir, 35620, Turkey
- Department of Biomedical Technologies, Graduate School of Natural and Applied Sciences, Izmir Katip Celebi University, Çiğli, Izmir, 35620, Turkey
| | - Ozan Karaman
- Biomedical Test Calibration Application and Research Center, Izmir Katip Celebi University, Çiğli, Izmir, 35620, Turkey
- Department of Biomedical Technologies, Graduate School of Natural and Applied Sciences, Izmir Katip Celebi University, Çiğli, Izmir, 35620, Turkey
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Çiğli, Izmir, 35620, Turkey
| | - Nermin Topaloğlu
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Çiğli, Izmir, 35620, Turkey.
| |
Collapse
|
164
|
Karbowniczek JE, Berniak K, Knapczyk-Korczak J, Williams G, Bryant JA, Nikoi ND, Banzhaf M, de Cogan F, Stachewicz U. Strategies of nanoparticles integration in polymer fibers to achieve antibacterial effect and enhance cell proliferation with collagen production in tissue engineering scaffolds. J Colloid Interface Sci 2023; 650:1371-1381. [PMID: 37480652 DOI: 10.1016/j.jcis.2023.07.066] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/24/2023]
Abstract
Current design strategies for biomedical tissue scaffolds are focused on multifunctionality to provide beneficial microenvironments to support tissue growth. We have developed a simple yet effective approach to create core-shell fibers of poly(3-hydroxybuty-rate-co-3-hydroxyvalerate) (PHBV), which are homogenously covered with titanium dioxide (TiO2) nanoparticles. Unlike the blend process, co-axial electrospinning enabled the uniform distribution of nanoparticles without the formation of large aggregates. We observed 5 orders of magnitude reduction in Escherichia coli survival after contact with electrospun scaffolds compared to the non-material control. In addition, our hybrid cores-shell structure supported significantly higher osteoblast proliferation after 7 days of cell culture and profound generation of 3D networked collagen fibers after 14 days. The organic-inorganic composite scaffold produced in this study demonstrates a unique combination of antibacterial properties and increased bone regeneration properties. In summary, the multifunctionality of the presented core-shell cPHBV+sTiO2 scaffolds shows great promise for biomedical applications.
Collapse
Affiliation(s)
- J E Karbowniczek
- AGH University of Krakow, Faculty of Metals Engineering and Industrial Computer Science, Cracow, Poland
| | - K Berniak
- AGH University of Krakow, Faculty of Metals Engineering and Industrial Computer Science, Cracow, Poland
| | - J Knapczyk-Korczak
- AGH University of Krakow, Faculty of Metals Engineering and Industrial Computer Science, Cracow, Poland
| | - G Williams
- University of Birmingham, Institute for Microbiology and Infection, B15 2TT Birmingham, UK
| | - J A Bryant
- University of Birmingham, Institute for Microbiology and Infection, B15 2TT Birmingham, UK
| | - N D Nikoi
- University of Nottingham, School of Pharmacy, NG7 2RD Nottingham, UK
| | - M Banzhaf
- University of Birmingham, Institute for Microbiology and Infection, B15 2TT Birmingham, UK
| | - F de Cogan
- University of Nottingham, School of Pharmacy, NG7 2RD Nottingham, UK
| | - U Stachewicz
- AGH University of Krakow, Faculty of Metals Engineering and Industrial Computer Science, Cracow, Poland.
| |
Collapse
|
165
|
Yıldız A, Birer M, Turgut Birer Y, Uyar R, Yurdakök-Dikmen B, Acartürk F. Silk fibroin nanoparticles and β-tricalcium phosphate loaded tissue engineered gelatin bone scaffolds: A Nature-based, low-cost solution. J Biomater Appl 2023; 38:646-661. [PMID: 37889125 DOI: 10.1177/08853282231207578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Tissue engineering has recently attracted attention as an alternative to traditional treatment methods for tissue and organ damage. Since bone is one of the most important vital parts of the body, the treatment of bone damage is important. Silk fibroin is a natural polymer with properties such as biocompatibility and biodegradability, which attracts attention with its controlled release, especially in drug delivery systems. In this study, gelatin-based scaffolds loaded with silk fibroin nanoparticles and β -tricalcium phosphate (β -TCP) were developed to be used as a potential drug delivery system in bone tissue engineering. The chosen nanoparticle formulation has a 294 nm average diameter with a 0.380 polidispersity index (PDI). In vitro characterization of scaffolds was performed by mechanical, morphological characterization, swelling capacity, Differential Scanning Calorimetry (DSC), Fourier-Transform Infrared Spectroscopy (FT-IR) measurements, and biocompatibility was evaluated by cell culture studies. Swelling index, tensile strength, elongation at break, and Young modulus of the β -TCP and silk nanoparticles loaded scaffold were found as 456%, 1.476 MPa, 6.75%, and 24 MPa, respectively. In vitro cell culture studies have shown that scaffolds prepared in the present study can accelerate osteoblast differentiation and increase the healing rate of bone tissues. In addition, they have the potential to be used as a drug delivery system in bone tissue engineering that needs to be evaluated with further studies.
Collapse
Affiliation(s)
- Ayşegül Yıldız
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Mehmet Birer
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Yağmur Turgut Birer
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Recep Uyar
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Begüm Yurdakök-Dikmen
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Füsun Acartürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| |
Collapse
|
166
|
Li M, Li D, Jiang Y, He P, Li Y, Wu Y, Lei W, de Bruijn JD, Cannon RD, Mei L, Zhang H, Ji P, Zhang H, Yuan H. The genetic background determines material-induced bone formation through the macrophage-osteoclast axis. Biomaterials 2023; 302:122356. [PMID: 37898023 DOI: 10.1016/j.biomaterials.2023.122356] [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: 11/15/2022] [Revised: 06/28/2023] [Accepted: 10/15/2023] [Indexed: 10/30/2023]
Abstract
Osteoinductive materials are characterized by their ability to induce bone formation in ectopic sites. Thus, osteoinductive materials hold promising potential for repairing bone defects. However, the mechanism of material-induced bone formation remains unknown, which limits the design of highly potent osteoinductive materials. Here, we demonstrated a genetic background link among macrophage polarization, osteoclastogenesis and material-induced bone formation. The intramuscular implantation of an osteoinductive material in FVB/NCrl (FVB) mice resulted in more M2 macrophages at week 1, more osteoclasts at week 2 and increased bone formation after week 4 compared with the results obtained in C57BL/6JOlaHsd (C57) mice. Similarly, in vitro, with a greater potential to form M2 macrophages, monocytes derived from FVB mice formed more osteoclasts than those derived from C57 mice. A transcriptomic analysis identified Csf1, Cxcr4 and Tgfbr2 as the main genes controlling macrophage-osteoclast coupling, which were further confirmed by related inhibitors. With such coupling, macrophage polarization and osteoclast formation of monocytes in vitro successfully predicted in vivo bone formation in four other mouse strains. Considering material-induced bone formation as an example of acquired heterotopic bone formation, the current findings shed a light on precision medicine for both bone regeneration and the treatment of pathological heterotopic bone formation.
Collapse
Affiliation(s)
- Mingzheng Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Dan Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yucan Jiang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Ping He
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yeming Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yan Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Wei Lei
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Joost D de Bruijn
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, 3723 MB Bilthoven, the Netherlands; Queen Mary University of London, London, UK
| | - Richard D Cannon
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Li Mei
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China; Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Hua Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Ji
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| | - Hongmei Zhang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| | - Huipin Yuan
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, 3723 MB Bilthoven, the Netherlands; Huipin Yuan's Lab, Chengdu, China.
| |
Collapse
|
167
|
Jurczak P, Lach S. Hydrogels as Scaffolds in Bone-Related Tissue Engineering and Regeneration. Macromol Biosci 2023; 23:e2300152. [PMID: 37276333 DOI: 10.1002/mabi.202300152] [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: 04/11/2023] [Revised: 05/22/2023] [Indexed: 06/07/2023]
Abstract
Several years have passed since the medical and scientific communities leaned toward tissue engineering as the most promising field to aid bone diseases and defects resulting from degenerative conditions or trauma. Owing to their histocompatibility and non-immunogenicity, bone grafts, precisely autografts, have long been the gold standard in bone tissue therapies. However, due to issues associated with grafting, especially the surgical risks and soaring prices of the procedures, alternatives are being extensively sought and researched. Fibrous and non-fibrous materials, synthetic substitutes, or cell-based products are just a few examples of research directions explored as potential solutions. A very promising subgroup of these replacements involves hydrogels. Biomaterials resembling the bone extracellular matrix and therefore acting as 3D scaffolds, providing the appropriate mechanical support and basis for cell growth and tissue regeneration. Additional possibility of using various stimuli in the form of growth factors, cells, etc., within the hydrogel structure, extends their use as bioactive agent delivery platforms and acts in favor of their further directed development. The aim of this review is to bring the reader closer to the fascinating subject of hydrogel scaffolds and present the potential of these materials, applied in bone and cartilage tissue engineering and regeneration.
Collapse
Affiliation(s)
- Przemyslaw Jurczak
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Centre Polish Academy of Sciences, Gdansk, 80-308, Poland
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk, 80-308, Poland
| | - Slawomir Lach
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk, 80-308, Poland
| |
Collapse
|
168
|
Dos Anjos LM, Rocha ADO, Magrin GL, Kammer PV, Benfatti CAM, Matias de Souza JC, Sanz M, Henriques BAPDC. Bibliometric analysis of the 100 most cited articles on bone grafting in dentistry. Clin Oral Implants Res 2023; 34:1198-1216. [PMID: 37577958 DOI: 10.1111/clr.14152] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023]
Abstract
AIM This bibliometric study analyzed the characteristics of the 100 most cited articles on bone grafts in dentistry. MATERIALS AND METHODS A database search was performed on the Web of Science Core Collection using a specific search strategy. Scopus and Google Scholar were also consulted for citation comparisons. Data extracted included: title, citation metrics, publication year, journal, study design, graft material, surgical technique, authors, institution, and country. Bibliometric networks were generated using VOSviewer. RESULTS The identified articles were published between 1991 and 2019. Citation counts ranged from 120 to 1161 (mean: 240, 30). Clinical Oral Implants Research was the most cited journal (5175 citations; 25/100). Xenogeneic bone graft material was the most frequently used (5130 citations; 22/100). Europe had 62 articles (14,604 citations), and the United States was the most prominent country (5209 citations; 22/100). The University of Bern had the highest number of citations (2565 citations; 13/100), with Buser D as the author with the largest number of articles (2648 citations; 12/100). CONCLUSION This study shows the scientific progress on bone grafts in dentistry. The use of xenogeneic grafts for horizontal and/or vertical ridge augmentation was the most prominent trend.
Collapse
Affiliation(s)
- Lucas Menezes Dos Anjos
- Department of Dentistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Aurélio de Oliveira Rocha
- Department of Dentistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Gabriel Leonardo Magrin
- Department of Dentistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
- Department of Periodontology, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
| | - Pedro Vitali Kammer
- Department of Dentistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | | | | | - Mariano Sanz
- Department of Periodontology, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
| | | |
Collapse
|
169
|
Radha G, Manjubaashini N, Balakumar S. Nano-hydroxyapatite/natural polymer composite scaffolds for bone tissue engineering: a brief review of recent trend. IN VITRO MODELS 2023; 2:125-151. [PMID: 39872168 PMCID: PMC11756495 DOI: 10.1007/s44164-023-00049-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 01/29/2025]
Abstract
Nanostructured inorganic biomaterial emerged as the most essential platform to address traumatic and non-traumatic conditions of hard tissues in the current scenario. Synthetic inorganic biomaterials serve as an efficient and pathogen-free choice that overcomes the obstructions associated with autografts and allografts to promote new tissue regeneration, since nano-hydroxyapatite (nHAp) is a biomaterial that mimics the natural mineral composition of bones and teeth of human hard tissues, which is widely employed in orthopedics and dentistry. The nHAp-based materials exhibit bioactive, biocompatible, and osteoconductive features under in vitro and in vivo conditions. The brittle nature of synthetic nHAp leads to weak mechanical properties, which eventually confines the utility of nHAp in load-bearing applications. Hence, this review focuses on the recent trends in the fabrication and investigation of nHAp-based polymer nanocomposite scaffolds for bone regeneration. Employing different polymers and fabrication strategies would efficiently tailor the physicochemical properties, and tailor-made mechanical properties in competence with biodegradation, thereby enhancing their potential in biomedical utility, and exploring their efficacy under in vitro and in vivo conditions to make "HAp-based smart-biomaterials" for bone tissue engineering.
Collapse
Affiliation(s)
- G. Radha
- National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai, 600 025 Tamilnadu India
| | - N. Manjubaashini
- National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai, 600 025 Tamilnadu India
| | - S. Balakumar
- National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai, 600 025 Tamilnadu India
| |
Collapse
|
170
|
Aadinath W, Muthuvijayan V. Antibacterial and angiogenic potential of iron oxide nanoparticles-stabilized acrylate-based scaffolds for bone tissue engineering applications. Colloids Surf B Biointerfaces 2023; 231:113572. [PMID: 37797467 DOI: 10.1016/j.colsurfb.2023.113572] [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: 08/23/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023]
Abstract
Pickering emulsion polymerization, stabilized by inorganic nanoparticles such as iron oxide nanoparticles (IONPs), can be used to fabricate scaffolds with the desired porosity and pore size. These nanoparticles create stable emulsions that can be processed under harsh polymerization conditions. IONPs, apart from serving as an emulsifier, impart beneficial bioactivities such as antibacterial and pro-angiogenic activity. Here, we coated IONPs with three different weights of oleic acid (5.0 g, 7.5 g, and 10.0 g) to synthesize oleic acid-IONPs (OA-IONPs) that possess the desired hydrophobicity (contact angle > 100°). Next, glycidyl methacrylate and trimethylolpropane triacrylate were polymerized using the Pickering emulsion polymerization technique stabilized by the OA-IONPs. The physicochemical properties of the resulting porous scaffolds were thoroughly characterized using scanning electron microscopy (SEM), thermogravimetric analysis (TGA), Fourier transform infrared spectroscopy (FT-IR), vibrating sample magnetometry (VSM), and a universal testing machine (UTM). The SEM images confirmed the formation of a porous scaffold. The IONPs content, measured using inductively coupled plasma mass spectrometry (ICP-MS), was in the range of 22-26 µg/mg of the scaffold. The mechanical strengths of the scaffolds were in the range of cancellous bone. The degradation profile of the scaffolds varied between 29% and 41% degradation over 30 days. In vitro cytotoxicity studies conducted using the fibroblast (L929) and osteosarcoma (MG-63) cell lines proved that these scaffolds were non-toxic. SEM images showed that the MG-63 cells adhered firmly to the scaffolds and exhibited a well-spread morphology. The antibacterial activity was confirmed by percentage inhibition studies, SEM analysis of bacterial membrane distortion, and reactive oxygen species (ROS) generation in the bacteria. Chick chorioallantoic membrane assay showed that the total vessel length and branch points were significantly increased in the presence of the scaffolds. These results confirm the pro-angiogenic potential of the fabricated scaffolds. The physicochemical, mechanical, and biological properties of the material suggest that the developed scaffolds would be suitable for bone tissue engineering applications.
Collapse
Affiliation(s)
- W Aadinath
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India
| | - Vignesh Muthuvijayan
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India.
| |
Collapse
|
171
|
Stocco TD, Zhang T, Dimitrov E, Ghosh A, da Silva AMH, Melo WCMA, Tsumura WG, Silva ADR, Sousa GF, Viana BC, Terrones M, Lobo AO. Carbon Nanomaterial-Based Hydrogels as Scaffolds in Tissue Engineering: A Comprehensive Review. Int J Nanomedicine 2023; 18:6153-6183. [PMID: 37915750 PMCID: PMC10616695 DOI: 10.2147/ijn.s436867] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/12/2023] [Indexed: 11/03/2023] Open
Abstract
Carbon-based nanomaterials (CBNs) are a category of nanomaterials with various systems based on combinations of sp2 and sp3 hybridized carbon bonds, morphologies, and functional groups. CBNs can exhibit distinguished properties such as high mechanical strength, chemical stability, high electrical conductivity, and biocompatibility. These desirable physicochemical properties have triggered their uses in many fields, including biomedical applications. In this review, we specifically focus on applying CBNs as scaffolds in tissue engineering, a therapeutic approach whereby CBNs can act for the regeneration or replacement of damaged tissue. Here, an overview of the structures and properties of different CBNs will first be provided. We will then discuss state-of-the-art advancements of CBNs and hydrogels as scaffolds for regenerating various types of human tissues. Finally, a perspective of future potentials and challenges in this field will be presented. Since this is a very rapidly growing field, we expect that this review will promote interdisciplinary efforts in developing effective tissue regeneration scaffolds for clinical applications.
Collapse
Affiliation(s)
- Thiago Domingues Stocco
- Bioengineering Program, Scientific and Technological Institute, Brazil University, São Paulo, SP, Brazil
| | - Tianyi Zhang
- Pennsylvania State University, University Park, PA, USA
| | | | - Anupama Ghosh
- Department of Chemical and Materials Engineering (DEQM), Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Wanessa C M A Melo
- FTMC, State Research institute Center for Physical Sciences and Technology, Department of Functional Materials and Electronics, Vilnius, Lithuanian
| | - Willian Gonçalves Tsumura
- Bioengineering Program, Scientific and Technological Institute, Brazil University, São Paulo, SP, Brazil
| | - André Diniz Rosa Silva
- FATEC, Ribeirão Preto, SP, Brazil
- Interdisciplinary Laboratory for Advanced Materials (LIMAV), BioMatLab Group, Materials Science and Engineering Graduate Program, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Gustavo F Sousa
- Interdisciplinary Laboratory for Advanced Materials (LIMAV), BioMatLab Group, Materials Science and Engineering Graduate Program, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Bartolomeu C Viana
- Interdisciplinary Laboratory for Advanced Materials (LIMAV), BioMatLab Group, Materials Science and Engineering Graduate Program, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | | | - Anderson Oliveira Lobo
- Interdisciplinary Laboratory for Advanced Materials (LIMAV), BioMatLab Group, Materials Science and Engineering Graduate Program, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| |
Collapse
|
172
|
Wang Y, Chen S, Liang H, Bai J, Wang M. Design and fabrication of biomimicking radially graded scaffolds via digital light processing 3D printing for bone regeneration. J Mater Chem B 2023; 11:9961-9974. [PMID: 37818766 DOI: 10.1039/d3tb01573d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Scaffolds are an essential component in bone tissue engineering (BTE). However, most of the current BTE scaffolds are homogeneous structures and do not resemble the graded architectures of native bone. In the current study, four types of biomimicking scaffold designs based on gyroid (G) and primitive (P) units with radially graded pore sizes were devised, and scaffolds of these designs with two porosity groups (65 vol% and 75 vol%) were fabricated via digital light processing (DLP) 3D printing using biphasic calcium phosphate (BCP). Scaffolds of the gyroid-gyroid (G-G) design displayed better dimensional accuracy, compressive property, and cell proliferation rate than gyroid-primitive (G-P), primitive-gyroid (P-G), and primitive-primitive (P-P) scaffolds. Subsequently, graded G-G scaffolds with different porosities were fabricated and the relationship between compressive strength and porosity was determined. Furthermore, the sintered BCP bioceramics fabricated via current manufacturing process exhibited excellent biocompatibility and bioactivity, indicating their high potential for BTE.
Collapse
Affiliation(s)
- Yue Wang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong.
| | - Shangsi Chen
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong.
| | - Haowen Liang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Jiaming Bai
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Min Wang
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong.
| |
Collapse
|
173
|
Dürig J, Calcagni M, Buschmann J. Transition metals in angiogenesis - A narrative review. Mater Today Bio 2023; 22:100757. [PMID: 37593220 PMCID: PMC10430620 DOI: 10.1016/j.mtbio.2023.100757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
The aim of this paper is to offer a narrative review of the literature regarding the influence of transition metals on angiogenesis, excluding lanthanides and actinides. To our knowledge there are not any reviews up to date offering such a summary, which inclined us to write this paper. Angiogenesis describes the process of blood vessel formation, which is an essential requirement for human growth and development. When the complex interplay between pro- and antiangiogenic mediators falls out of balance, angiogenesis can quickly become harmful. As it is so fundamental, both its inhibition and enhancement take part in various diseases, making it a target for therapeutic treatments. Current methods come with limitations, therefore, novel agents are constantly being researched, with metal agents offering promising results. Various transition metals have already been investigated in-depth, with studies indicating both pro- and antiangiogenic properties, respectively. The transition metals are being applied in various formulations, such as nanoparticles, complexes, or scaffold materials. Albeit the increasing attention this field is receiving, there remain many unanswered questions, mostly regarding the molecular mechanisms behind the observed effects. Notably, approximately half of all the transition metals have not yet been investigated regarding potential angiogenic effects. Considering the promising results which have already been established, it should be of great interest to begin investigating the remaining elements whilst also further analyzing the established effects.
Collapse
Affiliation(s)
- Johannes Dürig
- University of Zürich, Faculty of Medicine, Pestalozzistrasse 3, 8032, Zurich, Switzerland
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Maurizio Calcagni
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Johanna Buschmann
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| |
Collapse
|
174
|
Chen YT, Chuang YH, Chen CM, Wang JY, Wang J. Development of hybrid scaffolds with biodegradable polymer composites and bioactive hydrogels for bone tissue engineering. BIOMATERIALS ADVANCES 2023; 153:213562. [PMID: 37549480 DOI: 10.1016/j.bioadv.2023.213562] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/19/2023] [Accepted: 07/17/2023] [Indexed: 08/09/2023]
Abstract
The development of treatments for critical-sized bone defects has been considered an important topic in the biomedical field because of the high demand for transplantable bone grafts. Following the concept of tissue engineering, implantation of biocompatible porous scaffolds carrying cells and regulating factors is the most efficient strategy to stimulate clinical bone regeneration. With the advancement in the development of 3D-printing techniques, scaffolds with highly controllable architectures can be fabricated to further improve healing efficacies. However, challenges such as the limited biocompatibility of resin materials and poor cell-carrying capacities still exist in the application of current scaffolds. In this study, a novel biodegradable polymer, poly (ethylene glycol)-co-poly (glycerol sebacate) acrylate (PEGSA), was synthesized and blended with hydroxyapatite (HAP) nanoparticles to produce osteoinductive and photocurable resins for 3D printing. The composites were optimized and applied in the fabrication of gyroid scaffolds with biomimetic characteristics and high permeability, followed by the combination of bioactive hydrogels containing Wharton's jelly-derived mesenchymal stem cells (WJMSC) to increase the efficiency of cell delivery. The promotion of osteogenesis from 3D-printed scaffolds was confirmed in-vivo while the hybrid scaffolds were proven to be great platforms for WJMSC culture and differentiation in-vitro. These results indicate that the proposed hybrid systems, combining osteoinductive 3D-printed scaffolds and cell-laden hydrogels, have great potential for bone tissue engineering and are expected to be applied in the treatment of bone defects based on active tissue regeneration.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Ya-Han Chuang
- Interdisciplinary Program of Life Science and Medicine, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Chao-Ming Chen
- Department of Orthopedics Surgery and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Jir-You Wang
- Department of Orthopedics Surgery and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Jane Wang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan.
| |
Collapse
|
175
|
Tuygunov N, Zakaria MN, Yahya NA, Abdul Aziz A, Cahyanto A. Efficacy and bone-contact biocompatibility of glass ionomer cement as a biomaterial for bone regeneration: A systematic review. J Mech Behav Biomed Mater 2023; 146:106099. [PMID: 37660446 DOI: 10.1016/j.jmbbm.2023.106099] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Bone regeneration is a rapidly growing field that seeks to develop new biomaterials to regenerate bone defects. Conventional bone graft materials have limitations, such as limited availability, complication, and rejection. Glass ionomer cement (GIC) is a biomaterial with the potential for bone regeneration due to its bone-contact biocompatibility, ease of use, and cost-effectiveness. GIC is a two-component material that adheres to the bone and releases ions that promote bone growth and mineralization. A systematic literature search was conducted using PubMed-MEDLINE, Scopus, and Web of Science databases and registered in the PROSPERO database to determine the evidence regarding the efficacy and bone-contact biocompatibility of GIC as bone cement. Out of 3715 initial results, thirteen studies were included in the qualitative synthesis. Two tools were employed in evaluating the Risk of Bias (RoB): the QUIN tool for assessing in vitro studies and SYRCLE for in vivo. The results indicate that GIC has demonstrated the ability to adhere to bone and promote bone growth. Establishing a chemical bond occurs at the interface between the GIC and the mineral phase of bone. This interaction allows the GIC to exhibit osteoconductive properties and promote the growth of bone tissue. GIC's bone-contact biocompatibility, ease of preparation, and cost-effectiveness make it a promising alternative to conventional bone grafts. However, further research is required to fully evaluate the potential application of GIC in bone regeneration. The findings hold implications for advancing material development in identifying the optimal composition and fabrication of GIC as a bone repair material.
Collapse
Affiliation(s)
- Nozimjon Tuygunov
- Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Myrna Nurlatifah Zakaria
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia; Biomaterials Technology Research Groups, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Noor Azlin Yahya
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia; Biomaterials Technology Research Groups, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Azwatee Abdul Aziz
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia; Biomaterials Technology Research Groups, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Arief Cahyanto
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia; Biomaterials Technology Research Groups, Faculty of Dentistry, University of Malaya, Kuala Lumpur, 50603, Malaysia; Department of Dental Materials Science and Technology, Faculty of Dentistry, Padjadjaran University, Jatinangor, 45363, Indonesia.
| |
Collapse
|
176
|
Teimoori M, Nokhbatolfoghahaei H, Khojasteh A. Bilayer scaffolds/membranes for bone tissue engineering applications: A systematic review. BIOMATERIALS ADVANCES 2023; 153:213528. [PMID: 37352742 DOI: 10.1016/j.bioadv.2023.213528] [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: 04/09/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
OBJECTIVE This systematic review evaluates the purpose, materials, physio-mechanical, and biological effects of bilayer scaffolds/membranes used for bone tissue engineering applications. METHODS A comprehensive electronic search of English-language literature from 2012 to October 2022 was conducted in PubMed, Scopus, ScienceDirect, and Google Scholar online databases according to the PRISMA 2020 guidelines. The quality of animal studies was evaluated through the SYRCLE's risk of bias tool. RESULTS A total of 77 studies were sought for retrieval, and 39 studies met the inclusion criteria. According to the synthesis results, most bilayers had a dense barrier layer that prevented connective tissue penetration and a loose osteogenic layer that supported cell migration and osteogenesis. PLGA, PCL, and chitosan were the most common polymers in the barrier layers, while the most utilized polymers in osteogenic layers were PLGA and gelatin. Electrospinning and solvent casting were the most common fabrication methods to design the bilayer structures. Many studies reported higher biological results for bilayers compared to their single layers. Also, fabricated bilayers' in vitro osteogenesis and in vivo new bone formation were significantly superior or at least comparable to the frequently used commercial membranes. CONCLUSION 1) Bilayers with two distinct layers and different materials, porosities, mechanical properties, and biological behavior can significantly improve heterogeneous bone regeneration; 2) the addition of ceramics and/or drugs to the osteogenic layer enhances the osteogenic properties of the bilayers; 3) fabrication method and pore size of the layers play an important role in determining the mechanical and biological behavior of them.
Collapse
Affiliation(s)
- Mahdis Teimoori
- Student Research Committee, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Khojasteh
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Cranio-Maxillofacial Surgery, University Hospital, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
177
|
Moradi L, Witek L, Vivekanand Nayak V, Cabrera Pereira A, Kim E, Good J, Liu CJ. Injectable hydrogel for sustained delivery of progranulin derivative Atsttrin in treating diabetic fracture healing. Biomaterials 2023; 301:122289. [PMID: 37639975 PMCID: PMC11232488 DOI: 10.1016/j.biomaterials.2023.122289] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/22/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
Hydrogels with long-term storage stability, controllable sustained-release properties, and biocompatibility have been garnering attention as carriers for drug/growth factor delivery in tissue engineering applications. Chitosan (CS)/Graphene Oxide (GO)/Hydroxyethyl cellulose (HEC)/β-glycerol phosphate (β-GP) hydrogel is capable of forming a 3D gel network at physiological temperature (37 °C), rendering it an excellent candidate for use as an injectable biomaterial. This work focused on an injectable thermo-responsive CS/GO/HEC/β-GP hydrogel, which was designed to deliver Atsttrin, an engineered derivative of a known chondrogenic and anti-inflammatory growth factor-like molecule progranulin. The combination of the CS/GO/HEC/β-GP hydrogel and Atsttrin provides a unique biochemical and biomechanical environment to enhance fracture healing. CS/GO/HEC/β-GP hydrogels with increased amounts of GO exhibited rapid sol-gel transition, higher viscosity, and sustained release of Atsttrin. In addition, these hydrogels exhibited a porous interconnected structure. The combination of Atsttrin and hydrogel successfully promoted chondrogenesis and osteogenesis of bone marrow mesenchymal stem cells (bmMSCs) in vitro. Furthermore, the work also presented in vivo evidence that injection of Atsttrin-loaded CS/GO/HEC/β-GP hydrogel stimulated diabetic fracture healing by simultaneously inhibiting inflammatory and stimulating cartilage regeneration and endochondral bone formation signaling pathways. Collectively, the developed injectable thermo-responsive CS/GO/HEC/βG-P hydrogel yielded to be minimally invasive, as well as capable of prolonged and sustained delivery of Atsttrin, for therapeutic application in impaired fracture healing, particularly diabetic fracture healing.
Collapse
Affiliation(s)
- Lida Moradi
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA; Department of Orthopaedics & Rehabilitation, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Lukasz Witek
- Biomaterials Division - Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA; Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY, 11201, USA
| | - Vasudev Vivekanand Nayak
- Biomaterials Division - Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Angel Cabrera Pereira
- Biomaterials Division - Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Ellen Kim
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA
| | - Julia Good
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA; Department of Orthopaedics & Rehabilitation, Yale University School of Medicine, New Haven, CT, 06510, USA; Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
178
|
Woodbury SM, Swanson WB, Douglas L, Niemann D, Mishina Y. Temperature-responsive PCL-PLLA nanofibrous tissue engineering scaffolds with memorized porous microstructure recovery. FRONTIERS IN DENTAL MEDICINE 2023; 4:1240397. [PMID: 38606037 PMCID: PMC11008614 DOI: 10.3389/fdmed.2023.1240397] [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] [Indexed: 04/13/2024] Open
Abstract
Biomaterial scaffolds in tissue engineering facilitate tissue regeneration and integration with the host. Poor healing outcomes arise from lack of cell and tissue infiltration, and ill-fitting interfaces between matrices or grafts, resulting in fibrous tissue formation, inflammation, and resorption. Existing tissue engineering scaffolds struggle to recover from deformation to fit irregularly shaped defects encountered in clinical settings without compromising their mechanical properties and favorable internal architecture. This study introduces a synthetic biomaterial scaffold composed of high molecular weight poly (L-lactic acid) (PLLA) and an interpenetrating network of poly (ε-caprolactone) (PCL), in a composition aiming to address the need for conformal fitting synthetic matrices which retain and recover their advantageous morphologies. The scaffold, known as thermosensitive memorized microstructure (TS-MMS), forms nanofibrous materials with memorized microstructures capable of recovery after deformation, including macropores and nanofibers. TS-MMS nanofibers, with 50-500 nm diameters, are formed via thermally induced phase separation (TIPS) of PLLA after in situ polymerization of PCL-diacrylate. A critical partial-melting temperature of TS-MMS at 52°C enables bulk deformation above this temperature, while retaining the nanofibrous and macroporous structures upon cooling to 37°C. Incorporation of drug-loaded poly (lactide-co-glycolide) (PLGA) nanoparticles directly into TS-MMS nanofibers during fabrication allows sustained release of a model drug for up to 40 days. Subcutaneous implantation in vivo using LysM-Cre;td-Tomato; Col1eGFP mice demonstrates successful cellularization and integration of deformed/recovered TS-MMS materials, surpassing the limitations of deformed PLLA scaffolds, to facilitate cell and vasculature infiltration requisite for successful bone regeneration. Additionally we demonstrated a method for embedding controlled release vehicles directly into the scaffold nanofibers; controlled release of simvastatin enhances vascularization and tissue maturation. TS-MMS scaffolds offer promising improvements in clinical handling and performance compared to existing biomaterial scaffolds.
Collapse
Affiliation(s)
- Seth M. Woodbury
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, Ann Arbor, MI, United States
- Department of Physics, College of Literature, Science and the Arts, University of Michigan, Ann Arbor, MI, United States
| | - W. Benton Swanson
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Lindsey Douglas
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, Ann Arbor, MI, United States
| | - David Niemann
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, Ann Arbor, MI, United States
| | - Yuji Mishina
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
179
|
Alonso-Fernández I, Haugen HJ, López-Peña M, González-Cantalapiedra A, Muñoz F. Use of 3D-printed polylactic acid/bioceramic composite scaffolds for bone tissue engineering in preclinical in vivo studies: A systematic review. Acta Biomater 2023; 168:1-21. [PMID: 37454707 DOI: 10.1016/j.actbio.2023.07.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
3D-printed composite scaffolds have emerged as an alternative to deal with existing limitations when facing bone reconstruction. The aim of the study was to systematically review the feasibility of using PLA/bioceramic composite scaffolds manufactured by 3D-printing technologies as bone grafting materials in preclinical in vivo studies. Electronic databases were searched using specific search terms, and thirteen manuscripts were selected after screening. The synthesis of the scaffolds was carried out using mainly extrusion-based techniques. Likewise, hydroxyapatite was the most used bioceramic for synthesizing composites with a PLA matrix. Among the selected studies, seven were conducted in rats and six in rabbits, but the high variability that exists regarding the experimental process made it difficult to compare them. Regarding the results, PLA/Bioceramic composite scaffolds have shown to be biocompatible and mechanically resistant. Preclinical studies elucidated the ability of the scaffolds to be used as bone grafts, allowing bone growing without adverse reactions. In conclusion, PLA/Bioceramics scaffolds have been demonstrated to be a promising alternative for treating bone defects. Nevertheless, more care should be taken when designing and performing in vivo trials, since the lack of standardization of the processes, which prevents the comparison of the results and reduces the quality of the information. STATEMENT OF SIGNIFICANCE: 3D-printed polylactic acid/bioceramic composite scaffolds have emerged as an alternative to deal with existing limitations when facing bone reconstruction. Since preclinical in vivo studies with animal models represent a mandatory step for clinical translation, the present manuscript analyzed and discussed not only those aspects related to the selection of the bioceramic material, the synthesis of the implants and their characterization. But provides a new approach to understand how the design and perform of clinical trials, as well as the selection of the analysis methods, may affect the obtained results, by covering authors' knowledgebase from veterinary medicine to biomaterial science. Thus, this study aims to systematically review the feasibility of using polylactic acid/bioceramic scaffolds as grafting materials in preclinical trials.
Collapse
Affiliation(s)
- Iván Alonso-Fernández
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain.
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Mónica López-Peña
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain
| | - Antonio González-Cantalapiedra
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain
| | - Fernando Muñoz
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain
| |
Collapse
|
180
|
Pei B, Hu M, Wu X, Lu D, Zhang S, Zhang L, Wu S. Investigations into the effects of scaffold microstructure on slow-release system with bioactive factors for bone repair. Front Bioeng Biotechnol 2023; 11:1230682. [PMID: 37781533 PMCID: PMC10537235 DOI: 10.3389/fbioe.2023.1230682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
In recent years, bone tissue engineering (BTE) has played an essential role in the repair of bone tissue defects. Although bioactive factors as one component of BTE have great potential to effectively promote cell differentiation and bone regeneration, they are usually not used alone due to their short effective half-lives, high concentrations, etc. The release rate of bioactive factors could be controlled by loading them into scaffolds, and the scaffold microstructure has been shown to significantly influence release rates of bioactive factors. Therefore, this review attempted to investigate how the scaffold microstructure affected the release rate of bioactive factors, in which the variables included pore size, pore shape and porosity. The loading nature and the releasing mechanism of bioactive factors were also summarized. The main conclusions were achieved as follows: i) The pore shapes in the scaffold may have had no apparent effect on the release of bioactive factors but significantly affected mechanical properties of the scaffolds; ii) The pore size of about 400 μm in the scaffold may be more conducive to controlling the release of bioactive factors to promote bone formation; iii) The porosity of scaffolds may be positively correlated with the release rate, and the porosity of 70%-80% may be better to control the release rate. This review indicates that a slow-release system with proper scaffold microstructure control could be a tremendous inspiration for developing new treatment strategies for bone disease. It is anticipated to eventually be developed into clinical applications to tackle treatment-related issues effectively.
Collapse
Affiliation(s)
- Baoqing Pei
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Mengyuan Hu
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xueqing Wu
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Da Lu
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shijia Zhang
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Le Zhang
- Beijing Key Laboratory for Design and Evaluation Technology of Advanced Implantable and Interventional Medical Devices, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shuqin Wu
- School of Big Data and Information, Shanxi College of Technology, Taiyuan, Shanxi, China
| |
Collapse
|
181
|
Lázár I, Čelko L, Menelaou M. Aerogel-Based Materials in Bone and Cartilage Tissue Engineering-A Review with Future Implications. Gels 2023; 9:746. [PMID: 37754427 PMCID: PMC10530393 DOI: 10.3390/gels9090746] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
Aerogels are fascinating solid materials known for their highly porous nanostructure and exceptional physical, chemical, and mechanical properties. They show great promise in various technological and biomedical applications, including tissue engineering, and bone and cartilage substitution. To evaluate the bioactivity of bone substitutes, researchers typically conduct in vitro tests using simulated body fluids and specific cell lines, while in vivo testing involves the study of materials in different animal species. In this context, our primary focus is to investigate the applications of different types of aerogels, considering their specific materials, microstructure, and porosity in the field of bone and cartilage tissue engineering. From clinically approved materials to experimental aerogels, we present a comprehensive list and summary of various aerogel building blocks and their biological activities. Additionally, we explore how the complexity of aerogel scaffolds influences their in vivo performance, ranging from simple single-component or hybrid aerogels to more intricate and organized structures. We also discuss commonly used formulation and drying methods in aerogel chemistry, including molding, freeze casting, supercritical foaming, freeze drying, subcritical, and supercritical drying techniques. These techniques play a crucial role in shaping aerogels for specific applications. Alongside the progress made, we acknowledge the challenges ahead and assess the near and far future of aerogel-based hard tissue engineering materials, as well as their potential connection with emerging healing techniques.
Collapse
Affiliation(s)
- István Lázár
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Ladislav Čelko
- Central European Institute of Technology, Brno University of Technology, Purkynova 656/123, 612 00 Brno, Czech Republic;
| | - Melita Menelaou
- Department of Chemical Engineering, Cyprus University of Technology, 30 Arch. Kyprianos Str., Limassol 3036, Cyprus
| |
Collapse
|
182
|
Alavi MS, Memarpour S, Pazhohan-Nezhad H, Salimi Asl A, Moghbeli M, Shadmanfar S, Saburi E. Applications of poly(lactic acid) in bone tissue engineering: A review article. Artif Organs 2023; 47:1423-1430. [PMID: 37475653 DOI: 10.1111/aor.14612] [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: 04/27/2023] [Revised: 06/04/2023] [Accepted: 06/12/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Bone tissue engineering is a promising approach to large-scale bone regeneration. This involves the use of an artificial extracellular matrix or scaffold and osteoblasts to promote osteogenesis and ossification at defect sites. Scaffolds are constructed using biomaterials that typically have properties similar to those of natural bone. METHOD In this study, which is a review of the literature, various evidences have been discussed in the field of Poly Lactic acid (PLA) polymer application and modifications made on it in order to induce osteogenesis and repair bone lesions. RESULTS PLA is a synthetic aliphatic polymer that has been extensively used for scaffold construction in bone tissue engineering owing to its good processability, biocompatibility, and flexibility in design. However, PLA has some drawbacks, including low osteoconductivity, low cellular adhesion, and the possibility of inflammatory reactions owing to acidic discharge in a living environment. To overcome these issues, a combination of PLA and other biomaterials has been introduced. CONCLUSIONS This short review discusses PLA's characteristics of PLA, its applications in bone regeneration, and its combination with other biomaterials.
Collapse
Affiliation(s)
- Mahya Sadat Alavi
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Memarpour
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Salimi Asl
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soraya Shadmanfar
- Health Research Center, Life Style Institute, Department of Rheumatology, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
183
|
Mai TP, Park JB, Nguyen HD, Min KA, Moon C. Current application of dexamethasone-incorporated drug delivery systems for enhancing bone formation. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2023; 53:643-665. [DOI: 10.1007/s40005-023-00629-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/31/2023] [Indexed: 03/10/2025]
|
184
|
Gholami Farashah MS, Mohammadi A, Javadi M, Soleimani Rad J, Shakouri SK, Meshgi S, Roshangar L. Bone marrow mesenchymal stem cells' osteogenic potential: superiority or non-superiority to other sources of mesenchymal stem cells? Cell Tissue Bank 2023; 24:663-681. [PMID: 36622494 DOI: 10.1007/s10561-022-10066-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/14/2022] [Indexed: 01/10/2023]
Abstract
Skeletal problems are an increasing issue due to the increase in the global aging population. Different statistics reports show that today, the global population is aging that results in skeletal problems, increased health system costs, and even higher mortality associated with skeletal problems. Common treatments such as surgery and bone grafts are not always effective and in some cases, they can even cause secondary problems such as infections or improper repair. Cell therapy is a method that can be utilized along with common treatments independently. Mesenchymal stem cells (MSCs) are a very important and efficient source in terms of different diseases, especially bone problems. These cells are present in different tissues such as bone marrow, adipose tissue, umbilical cord, placenta, dental pulp, peripheral blood, amniotic fluid and others. Among the types of MSCs, bone marrow mesenchymal stem cells (BMMSCs) are the most widely used source of these cells, which have appeared to be very effective and promising in terms of skeletal diseases, especially compared to the other sources of MSCs. This study focuses on the specific potential and content of BMMSCs from which the specific capacity of these cells originates, and compares their osteogenic potential with other types of MSCs, and also the future directions in the application of BMMSCs as a source for cell therapy.
Collapse
Affiliation(s)
- Mohammad Sadegh Gholami Farashah
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mohammadi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Javadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleimani Rad
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Kazem Shakouri
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahla Meshgi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
185
|
Bianconi S, Oliveira KMC, Klein KL, Wolf J, Schaible A, Schröder K, Barker J, Marzi I, Leppik L, Henrich D. Pretreatment of Mesenchymal Stem Cells with Electrical Stimulation as a Strategy to Improve Bone Tissue Engineering Outcomes. Cells 2023; 12:2151. [PMID: 37681884 PMCID: PMC10487010 DOI: 10.3390/cells12172151] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
Electrical stimulation (EStim), whether used alone or in combination with bone tissue engineering (BTE) approaches, has been shown to promote bone healing. In our previous in vitro studies, mesenchymal stem cells (MSCs) were exposed to EStim and a sustained, long-lasting increase in osteogenic activity was observed. Based on these findings, we hypothesized that pretreating MSC with EStim, in 2D or 3D cultures, before using them to treat large bone defects would improve BTE treatments. Critical size femur defects were created in 120 Sprague-Dawley rats and treated with scaffold granules seeded with MSCs that were pre-exposed or not (control group) to EStim 1 h/day for 7 days in 2D (MSCs alone) or 3D culture (MSCs + scaffolds). Bone healing was assessed at 1, 4, and 8 weeks post-surgery. In all groups, the percentage of new bone increased, while fibrous tissue and CD68+ cell count decreased over time. However, these and other healing features, like mineral density, bending stiffness, the amount of new bone and cartilage, and the gene expression of osteogenic markers, did not significantly differ between groups. Based on these findings, it appears that the bone healing environment could counteract the long-term, pro-osteogenic effects of EStim seen in our in vitro studies. Thus, EStim seems to be more effective when administered directly and continuously at the defect site during bone healing, as indicated by our previous studies.
Collapse
Affiliation(s)
- Santiago Bianconi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Karla M. C. Oliveira
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Kari-Leticia Klein
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Jakob Wolf
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Alexander Schaible
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Katrin Schröder
- Vascular Research Centre, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - John Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, Goethe University Frankfurt, 60528 Frankfurt am Main, Germany;
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Liudmila Leppik
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Dirk Henrich
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| |
Collapse
|
186
|
Kazimierczak P, Kalisz G, Sroka-Bartnicka A, Przekora A. Effectiveness of the production of tissue-engineered living bone graft: a comparative study using perfusion and rotating bioreactor systems. Sci Rep 2023; 13:13737. [PMID: 37612367 PMCID: PMC10447456 DOI: 10.1038/s41598-023-41003-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/20/2023] [Indexed: 08/25/2023] Open
Abstract
Bioreactor systems are very precious tools to generate living bone grafts in vitro. The aim of this study was to compare the effectiveness of rotating and perfusion bioreactor in the production of a living bone construct. Human bone marrow-derived mesenchymal stem cells (BMDSCs) were seeded on the surfaces of hydroxyapatite-based scaffolds and cultured for 21 days in three different conditions: (1) static 3D culture, (2) 3D culture in a perfusion bioreactor, and (3) dynamic 3D culture in a rotating bioreactor. Quantitative evaluation of cell number showed that cultivation in the perfusion bioreactor significantly reduced cell proliferation compared to the rotating bioreactor and static culture. Osteogenic differentiation test demonstrated that BMDSCs cultured in the rotating bioreactor produced significantly greater amount of osteopontin compared to the cells cultured in the perfusion bioreactor. Moreover, Raman spectroscopy showed that cultivation of BMDSCs in the rotating bioreactor enhanced extracellular matrix (ECM) mineralization that was characterized by B-type carbonated substitution of hydroxyapatite (associated with PO43- groups) and higher mineral-to-matrix ratio compared to the ECM of cells cultured in the perfusion system. Thus, it was concluded that the rotating bioreactor was much more effective than the perfusion one in the generation of bone tissue construct in vitro.
Collapse
Affiliation(s)
- Paulina Kazimierczak
- Independent Unit of Tissue Engineering and Regenerative Medicine, Medical University of Lublin, Chodzki 1 Street, 20-093, Lublin, Poland.
| | - Grzegorz Kalisz
- Independent Unit of Spectroscopy and Chemical Imaging, Medical University of Lublin, Chodzki 4a Street, 20-093, Lublin, Poland
| | - Anna Sroka-Bartnicka
- Independent Unit of Spectroscopy and Chemical Imaging, Medical University of Lublin, Chodzki 4a Street, 20-093, Lublin, Poland
| | - Agata Przekora
- Independent Unit of Tissue Engineering and Regenerative Medicine, Medical University of Lublin, Chodzki 1 Street, 20-093, Lublin, Poland
| |
Collapse
|
187
|
Ray S, Nandi SK, Dasgupta S. Enhanced bone regeneration using Antheraea mylittasilk fibroin and chitosan based scaffold: in-vivoand in-vitrostudy. Biomed Mater 2023; 18:055019. [PMID: 37552994 DOI: 10.1088/1748-605x/acee3c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/08/2023] [Indexed: 08/10/2023]
Abstract
In this study, highly interconnected porous scaffolds fromAntheraea mylittasilk fibroin (SF) and chitosan (CH) were fabricated using the freeze-drying method. The weight ratios of SF to CH were varied from 90:10 (SF90/CH10) to 50:50 (SF50/CH50) to prepare the scaffolds from the aqueous suspension of the protein-polysaccharide mix. From the initial optimization of scaffold composition with respect to their microstructure, porosity, and mechanical properties, the SF80/CH20scaffold exhibited the most suitable properties for bone tissue engineering application as compared to others compositions. Hencein-vitrohemocompatibility, protein adsorption, and MG-63 cell culture studies were carried out for SF80/CH20scaffold. The fabricated SF80/CH20scaffold showed a more controlled swelling percentage of 42.8%, with high BSA protein adsorption of 0.39 mg of BSA per gm of the scaffold at 24 h incubation period. Furthermore,in-vitroMG-63 cell culture study onto the fabricated SF80/CH20scaffold elicited excellent MG-63 cell attachment with better biocompatibility and cell viability with increased F-action production from day 3 to day 7 of the cell culture period.In vivobone defect healing in a rabbit tibia model revealed excellent bone healing capacity in SF80/CH20scaffold implanted specimens compared to control ones, as evident from histology and fluorochrome labeling analysis.
Collapse
Affiliation(s)
- Sambit Ray
- Department of Ceramic Engineering, NIT Rourkela, Rourkela, Odisha 769008, India
| | - Samit Kumar Nandi
- West Bengal University of Animal and Fishery Science, WBAFS, Kolkata 700037, India
| | - Sudip Dasgupta
- Department of Ceramic Engineering, NIT Rourkela, Rourkela, Odisha 769008, India
| |
Collapse
|
188
|
Xing Y, Qiu L, Liu D, Dai S, Sheu CL. The role of smart polymeric biomaterials in bone regeneration: a review. Front Bioeng Biotechnol 2023; 11:1240861. [PMID: 37662432 PMCID: PMC10469876 DOI: 10.3389/fbioe.2023.1240861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
Addressing critical bone defects necessitates innovative solutions beyond traditional methods, which are constrained by issues such as immune rejection and donor scarcity. Smart polymeric biomaterials that respond to external stimuli have emerged as a promising alternative, fostering endogenous bone regeneration. Light-responsive polymers, employed in 3D-printed scaffolds and photothermal therapies, enhance antibacterial efficiency and bone repair. Thermo-responsive biomaterials show promise in controlled bioactive agent release, stimulating osteocyte differentiation and bone regeneration. Further, the integration of conductive elements into polymers improves electrical signal transmission, influencing cellular behavior positively. Innovations include advanced 3D-printed poly (l-lactic acid) scaffolds, polyurethane foam scaffolds promoting cell differentiation, and responsive polymeric biomaterials for osteogenic and antibacterial drug delivery. Other developments focus on enzyme-responsive and redox-responsive polymers, which offer potential for bone regeneration and combat infection. Biomaterials responsive to mechanical, magnetic, and acoustic stimuli also show potential in bone regeneration, including mechanically-responsive polymers, magnetic-responsive biomaterials with superparamagnetic iron oxide nanoparticles, and acoustic-responsive biomaterials. In conclusion, smart biopolymers are reshaping scaffold design and bone regeneration strategies. However, understanding their advantages and limitations is vital, indicating the need for continued exploratory research.
Collapse
Affiliation(s)
| | | | | | | | - Chia-Lin Sheu
- Department of Biomedical Engineering, Shantou University, Shantou, China
| |
Collapse
|
189
|
Yamada S, Ockermann PN, Schwarz T, Mustafa K, Hansmann J. Translation of biophysical environment in bone into dynamic cell culture under flow for bone tissue engineering. Comput Struct Biotechnol J 2023; 21:4395-4407. [PMID: 37711188 PMCID: PMC10498129 DOI: 10.1016/j.csbj.2023.08.008] [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: 05/13/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Bone is a dynamic environment where osteocytes, osteoblasts, and mesenchymal stem/progenitor cells perceive mechanical cues and regulate bone metabolism accordingly. In particular, interstitial fluid flow in bone and bone marrow serves as a primary biophysical stimulus, which regulates the growth and fate of the cellular components of bone. The processes of mechano-sensory and -transduction towards bone formation have been well studied mainly in vivo as well as in two-dimensional (2D) dynamic cell culture platforms, which elucidated mechanically induced osteogenesis starting with anabolic responses, such as production of nitrogen oxide and prostaglandins followed by the activation of canonical Wnt signaling, upon mechanosensation. The knowledge has been now translated into regenerative medicine, particularly into the field of bone tissue engineering, where multipotent stem cells are combined with three-dimensional (3D) scaffolding biomaterials to produce transplantable constructs for bone regeneration. In the presence of 3D scaffolds, the importance of suitable dynamic cell culture platforms increases further not only to improve mass transfer inside the scaffolds but to provide appropriate biophysical cues to guide cell fate. In principle, the concept of dynamic cell culture platforms is rooted to bone mechanobiology. Therefore, this review primarily focuses on biophysical environment in bone and its translation into dynamic cell culture platforms commonly used for 2D and 3D cell expansion, including their advancement, challenges, and future perspectives. Additionally, it provides the literature review of recent empirical studies using 2D and 3D flow-based dynamic cell culture systems for bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Center of Translational Oral Research-Tissue Engineering, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Philipp Niklas Ockermann
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
| | - Thomas Schwarz
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
| | - Kamal Mustafa
- Center of Translational Oral Research-Tissue Engineering, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Jan Hansmann
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Germany
- Department of Electrical Engineering, University of Applied Sciences Würzburg-Schweinfurt, Germany
| |
Collapse
|
190
|
Kaou MH, Furkó M, Balázsi K, Balázsi C. Advanced Bioactive Glasses: The Newest Achievements and Breakthroughs in the Area. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2287. [PMID: 37630871 PMCID: PMC10459405 DOI: 10.3390/nano13162287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023]
Abstract
Bioactive glasses (BGs) are especially useful materials in soft and bone tissue engineering and even in dentistry. They can be the solution to many medical problems, and they have a huge role in the healing processes of bone fractures. Interestingly, they can also promote skin regeneration and wound healing. Bioactive glasses are able to attach to the bone tissues and form an apatite layer which further initiates the biomineralization process. The formed intermediate apatite layer makes a connection between the hard tissue and the bioactive glass material which results in faster healing without any complications or side effects. This review paper summarizes the most recent advancement in the preparation of diverse types of BGs, such as silicate-, borate- and phosphate-based bioactive glasses. We discuss their physical, chemical, and mechanical properties detailing how they affect their biological performances. In order to get a deeper insight into the state-of-the-art in this area, we also consider their medical applications, such as bone regeneration, wound care, and dental/bone implant coatings.
Collapse
Affiliation(s)
- Maroua H. Kaou
- Centre for Energy Research, Institute of Technical Physics and Materials Science, Konkoly-Thege M. Str. 29-33, 1121 Budapest, Hungary; (M.H.K.); (M.F.); (K.B.)
- Doctoral School of Materials Science and Technologies, Óbuda University, Bécsi Str. 96/B, 1030 Budapest, Hungary
| | - Mónika Furkó
- Centre for Energy Research, Institute of Technical Physics and Materials Science, Konkoly-Thege M. Str. 29-33, 1121 Budapest, Hungary; (M.H.K.); (M.F.); (K.B.)
| | - Katalin Balázsi
- Centre for Energy Research, Institute of Technical Physics and Materials Science, Konkoly-Thege M. Str. 29-33, 1121 Budapest, Hungary; (M.H.K.); (M.F.); (K.B.)
| | - Csaba Balázsi
- Centre for Energy Research, Institute of Technical Physics and Materials Science, Konkoly-Thege M. Str. 29-33, 1121 Budapest, Hungary; (M.H.K.); (M.F.); (K.B.)
| |
Collapse
|
191
|
Liu XL, Zhang CJ, Shi JJ, Ke QF, Ge YW, Zhu ZA, Guo YP. Nacre-mimetic cerium-doped nano-hydroxyapatite/chitosan layered composite scaffolds regulate bone regeneration via OPG/RANKL signaling pathway. J Nanobiotechnology 2023; 21:259. [PMID: 37550715 PMCID: PMC10408205 DOI: 10.1186/s12951-023-01988-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/07/2023] [Indexed: 08/09/2023] Open
Abstract
Autogenous bone grafting has long been considered the gold standard for treating critical bone defects. However, its use is plagued by numerous drawbacks, such as limited supply, donor site morbidity, and restricted use for giant-sized defects. For this reason, there is an increasing need for effective bone substitutes to treat these defects. Mollusk nacre is a natural structure with outstanding mechanical property due to its notable "brick-and-mortar" architecture. Inspired by the nacre architecture, our team designed and fabricated a nacre-mimetic cerium-doped layered nano-hydroxyapatite/chitosan layered composite scaffold (CeHA/CS). Hydroxyapatite can provide a certain strength to the material like a brick. And as a polymer material, chitosan can slow down the force when the material is impacted, like an adhesive. As seen in natural nacre, the combination of these inorganic and organic components results in remarkable tensile strength and fracture toughness. Cerium ions have been demonstrated exceptional anti-osteoclastogenesis capabilities. Our scaffold featured a distinct layered HA/CS composite structure with intervals ranging from 50 to 200 μm, which provided a conducive environment for human bone marrow mesenchymal stem cell (hBMSC) adhesion and proliferation, allowing for in situ growth of newly formed bone tissue. In vitro, Western-blot and qPCR analyses showed that the CeHA/CS layered composite scaffolds significantly promoted the osteogenic process by upregulating the expressions of osteogenic-related genes such as RUNX2, OCN, and COL1, while inhibiting osteoclast differentiation, as indicated by reduced TRAP-positive osteoclasts and decreased bone resorption. In vivo, calvarial defects in rats demonstrated that the layered CeHA/CS scaffolds significantly accelerated bone regeneration at the defect site, and immunofluorescence indicated a lowered RANKL/OPG ratio. Overall, our results demonstrate that CeHA/CS scaffolds offer a promising platform for bone regeneration in critical defect management, as they promote osteogenesis and inhibit osteoclast activation.
Collapse
Affiliation(s)
- Xiao-Liang Liu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chuan-Jian Zhang
- The Education Ministry Key Lab of Resource Chemistry, Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, 200234, China
| | - Jing-Jing Shi
- The Education Ministry Key Lab of Resource Chemistry, Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, 200234, China
| | - Qin-Fei Ke
- The Education Ministry Key Lab of Resource Chemistry, Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, 200234, China
| | - Yu-Wei Ge
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Zhen-An Zhu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Ya-Ping Guo
- The Education Ministry Key Lab of Resource Chemistry, Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, 200234, China.
| |
Collapse
|
192
|
Assanah F, Grassie K, Anderson H, Xin X, Rowe D, Khan Y. Ultrasound-derived mechanical stimulation of cell-laden collagen hydrogels for bone repair. J Biomed Mater Res A 2023; 111:1200-1215. [PMID: 36728346 DOI: 10.1002/jbm.a.37508] [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: 07/12/2022] [Revised: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023]
Abstract
Cell therapy is emerging as an effective treatment strategy for many diseases. Here we describe a novel approach to bone tissue repair that combines hydrogel-based cell therapy with low intensity pulsed ultrasound (LIPUS), an FDA approved treatment for fracture repair. Bone marrow-derived stromal cells (BMSCs) have been encapsulated in type I collagen hydrogels and mechanically stimulated using LIPUS-derived acoustic radiation force (ARF). We observed the expression and upward trend of load-sensitive, osteoblast-specific markers and determined that the extent of cell response is dependent on an optimal combination of both hydrogel stiffness and ARF intensity. Specifically, cells encapsulated in hydrogels of optimal stiffness respond at the onset of ultrasound by upregulating early bone-sensitive markers such as calcium, cyclooxygenase-2, and prostaglandin E2 , and later by supporting mineralized tissue formation after 21 days of culture. In vivo evaluation of a critical size calvarial defect in NOD scid gamma (NSG) mice indicated that the implantation of BMSC-laden hydrogels of optimal stiffness improved healing of calvarial defects after daily administration of ARF over 4 weeks. Collectively, these findings validate the efficacy of our system of localized cell delivery for treating bone defects where undifferentiated BMSCs are induced to the osteoblastic lineage. Further, in vivo healing may be enhanced via non-invasive transdermal mechanical stimulation of implanted cells using ARF.
Collapse
Affiliation(s)
- Fayekah Assanah
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UCONN Health, Farmington, Connecticut, USA
| | - Kevin Grassie
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UCONN Health, Farmington, Connecticut, USA
| | - Hanna Anderson
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UCONN Health, Farmington, Connecticut, USA
| | - Xiaonan Xin
- Center for Regenerative Medicine and Skeletal Development, UCONN School of Dental Medicine, Farmington, Connecticut, USA
| | - David Rowe
- Center for Regenerative Medicine and Skeletal Development, UCONN School of Dental Medicine, Farmington, Connecticut, USA
| | - Yusuf Khan
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UCONN Health, Farmington, Connecticut, USA
- Department of Orthopedic Surgery, UCONN Health, Farmington, Connecticut, USA
| |
Collapse
|
193
|
DeMitchell-Rodriguez EM, Shen C, Nayak VV, Tovar N, Witek L, Torroni A, Yarholar LM, Cronstein BN, Flores RL, Coelho PG. Engineering 3D Printed Bioceramic Scaffolds to Reconstruct Critical-Sized Calvaria Defects in a Skeletally Immature Pig Model. Plast Reconstr Surg 2023; 152:270e-280e. [PMID: 36723712 PMCID: PMC11310574 DOI: 10.1097/prs.0000000000010258] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Three-dimensional printed bioceramic scaffolds composed of 100% β-tricalcium phosphate augmented with dipyridamole (3DPBC-DIPY) can regenerate bone across critically sized defects in skeletally mature and immature animal models. Before human application, safe and effective bone formation should be demonstrated in a large translational animal model. This study evaluated the ability of 3DPBC-DIPY scaffolds to restore critically sized calvarial defects in a skeletally immature, growing minipig. METHODS Unilateral calvarial defects (~1.4 cm) were created in 6-week-old Göttingen minipigs ( n = 12). Four defects were filled with a 1000 μm 3DPBC-DIPY scaffold with a cap (a solid barrier on the ectocortical side of the scaffold to prevent soft-tissue infiltration), four defects were filled with a 1000 μm 3DPBC-DIPY scaffold without a cap, and four defects served as negative controls (no scaffold). Animals were euthanized 12 weeks postoperatively. Calvariae were subjected to micro-computed tomography, 3D reconstruction with volumetric analysis, qualitative histologic analysis, and nanoindentation. RESULTS Scaffold-induced bone growth was statistically greater than in negative controls ( P ≤ 0.001), and the scaffolds with caps produced significantly more bone generation compared with the scaffolds without caps ( P ≤ 0.001). Histologic analysis revealed woven and lamellar bone with haversian canals throughout the regenerated bone. Cranial sutures were observed to be patent, and there was no evidence of ectopic bone formation or excess inflammatory response. Reduced elastic modulus and hardness of scaffold-regenerated bone were found to be statistically equivalent to native bone ( P = 0.148 for reduced elastic modulus of scaffolds with and without caps and P = 0.228 and P = 0.902 for hardness of scaffolds with and without caps, respectively). CONCLUSION 3DPBC-DIPY scaffolds have the capacity to regenerate bone across critically sized calvarial defects in a skeletally immature translational pig model. CLINICAL RELEVANCE STATEMENT This study assessed the bone generative capacity of 3D-printed bioceramic scaffolds composed of 100% β-tricalcium phosphate and augmented with dipyridamole placed within critical-sized calvarial defects in a growing porcine model.
Collapse
Affiliation(s)
| | - Chen Shen
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine
| | - Vasudev Vivekanand Nayak
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering
- Department of Biomaterials, New York University College of Dentistry
| | - Nick Tovar
- Department of Biomaterials, New York University College of Dentistry
| | - Lukasz Witek
- Department of Biomaterials, New York University College of Dentistry
- Department of Biomedical Engineering, New York University Tandon School of Engineering
| | - Andrea Torroni
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine
| | - Lauren M. Yarholar
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine
| | | | - Roberto L. Flores
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine
| | - Paulo G. Coelho
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine
| |
Collapse
|
194
|
Zhou S, Liu S, Wang Y, Li W, Wang J, Wang X, Wang S, Chen W, Lv H. Advances in the Study of Bionic Mineralized Collagen, PLGA, Magnesium Ionomer Materials, and Their Composite Scaffolds for Bone Defect Treatment. J Funct Biomater 2023; 14:406. [PMID: 37623651 PMCID: PMC10455784 DOI: 10.3390/jfb14080406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
The healing of bone defects after a fracture remains a key issue to be addressed. Globally, more than 20 million patients experience bone defects annually. Among all artificial bone repair materials that can aid healing, implantable scaffolds made from a mineralized collagen (MC) base have the strongest bionic properties. The MC/PLGA scaffold, created by adding Poly (lactic-co-glycolic acid) copolymer (PLGA) and magnesium metal to the MC substrate, plays a powerful role in promoting fracture healing because, on the one hand, it has good biocompatibility similar to that of MC; on the other hand, the addition of PLGA provides the scaffold with an interconnected porous structure, and the addition of magnesium allows the scaffold to perform anti-inflammatory, osteogenic, and angiogenic activities. Using the latest 3D printing technology for scaffold fabrication, it is possible to model the scaffold in advance according to the requirement and produce a therapeutic scaffold suitable for various bone-defect shapes with less time and effort, which can promote bone tissue healing and regeneration to the maximum extent. This study reviews the material selection and technical preparation of MC/PLGA scaffolds, and the progress of their research on bone defect treatment.
Collapse
Affiliation(s)
- Shuai Zhou
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Shihang Liu
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Yan Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Wenjing Li
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Juan Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, No. 30 Shuangqing Road, Beijing 100084, China
| | - Shuo Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, No. 30 Shuangqing Road, Beijing 100084, China
| | - Wei Chen
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Hongzhi Lv
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| |
Collapse
|
195
|
Lv S, Yuan X, Xiao J, Jiang X. Hemostasis-osteogenesis integrated Janus carboxymethyl chitin/hydroxyapatite porous membrane for bone defect repair. Carbohydr Polym 2023; 313:120888. [PMID: 37182974 DOI: 10.1016/j.carbpol.2023.120888] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/21/2023] [Accepted: 04/03/2023] [Indexed: 05/16/2023]
Abstract
Barrier membranes with osteogenesis are desirable for promoting bone repair. Janus membrane, which has a bilayered structure with different properties on each side, could meet the osteogenesis/barrier dual functions of guided bone regeneration. In this work, new biodegradable Janus carboxymethyl chitin membrane with asymmetric pore structure was prepared based on thermosensitive carboxymethyl chitin without using any crosslinkers. Nano-hydroxyapatites were cast on single-sided membrane. The obtained carboxymethyl chitin/nano-hydroxyapatite Janus membrane showed dual biofunctions: the dense layer of the Janus membrane could act as a barrier to prevent connective tissue cells from invading the bone defects, while the porous layer (with pore size 100-200 μm) containing nano-hydroxyapatite could guide bone regeneration. After implanted on the rat critical-sized calvarial defect 8 weeks, carboxymethyl chitin/nano-hydroxyapatite membrane showed the most newly formed bone tissue with the highest bone volume/total volume ratio (10.03 ± 1.81 %, analyzed by micro CT), which was significantly better than the commercial collagen membrane GTR® (5.05 ± 0.76 %). Meanwhile, this Janus membrane possessed good hemostatic ability. These results suggest a facile strategy to construct hemostasis-osteogenesis integrated Janus carboxymethyl chitin/hydroxyapatite membrane for guided bone regeneration.
Collapse
Affiliation(s)
- Siyao Lv
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China; Shandong Laboratory of Yantai Advanced Materials and Green Manufacturing, Yantai 264005, PR China.
| | - Xi Yuan
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| | - Xulin Jiang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| |
Collapse
|
196
|
Olevsky LM, Anup A, Jacques M, Keokominh N, Holmgren EP, Hixon KR. Direct Integration of 3D Printing and Cryogel Scaffolds for Bone Tissue Engineering. Bioengineering (Basel) 2023; 10:889. [PMID: 37627774 PMCID: PMC10451777 DOI: 10.3390/bioengineering10080889] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Cryogels, known for their biocompatibility and porous structure, lack mechanical strength, while 3D-printed scaffolds have excellent mechanical properties but limited porosity resolution. By combining a 3D-printed plastic gyroid lattice scaffold with a chitosan-gelatin cryogel scaffold, a scaffold can be created that balances the advantages of both fabrication methods. This study compared the pore diameter, swelling potential, mechanical characteristics, and cellular infiltration capability of combined scaffolds and control cryogels. The incorporation of the 3D-printed lattice demonstrated patient-specific geometry capabilities and significantly improved mechanical strength compared to the control cryogel. The combined scaffolds exhibited similar porosity and relative swelling ratio to the control cryogels. However, they had reduced elasticity, reduced absolute swelling capacity, and are potentially cytotoxic, which may affect their performance. This paper presents a novel approach to combine two scaffold types to retain the advantages of each scaffold type while mitigating their shortcomings.
Collapse
Affiliation(s)
- Levi M. Olevsky
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA; (L.M.O.); (A.A.)
| | - Amritha Anup
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA; (L.M.O.); (A.A.)
| | - Mason Jacques
- College of Engineering and Physical Sciences, University of New Hampshire, Durham, NH 03824, USA; (M.J.); (N.K.)
| | - Nadia Keokominh
- College of Engineering and Physical Sciences, University of New Hampshire, Durham, NH 03824, USA; (M.J.); (N.K.)
| | - Eric P. Holmgren
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA;
| | - Katherine R. Hixon
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA; (L.M.O.); (A.A.)
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA;
| |
Collapse
|
197
|
Najafabadi FM, Karbasi S, Benisi SZ, Shojaei S. Physical, mechanical, and biological performance of chitosan-based nanocomposite coating deposited on the polycaprolactone-based 3D printed scaffold: Potential application in bone tissue engineering. Int J Biol Macromol 2023; 243:125218. [PMID: 37285889 DOI: 10.1016/j.ijbiomac.2023.125218] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/09/2023]
Abstract
Recently, coating on composite scaffolds has attracted many researchers' attention to improve scaffolds' properties. In this research, a 3D printed scaffold was fabricated from polycaprolactone (PCL)/magnetic mesoporous bioactive glass (MMBG)/alumina nanowire (Al2O3, Optimal percentage 5 %) (PMA) and then coated with chitosan (Cs)/multi-walled carbon nanotubes (MWCNTs) by an immersion coating method. Structural analyses such as XRD and ATR-FTIR confirmed the presence of Cs and MWCNTs in the coated scaffolds. The SEM results of the coated scaffolds showed homogeneous three-dimensional structures with interconnected pores compared to the uncoated scaffolds. The coated scaffolds exhibited an increase in compression strength (up to 16.1 MPa) and compressive modulus (up to 40.83 MPa), improved surface hydrophilicity (up to 32.69°), and decrease in degradation rate (68 % remaining weight) compared to the uncoated scaffolds. The increase in apatite formation in the scaffold coated with Cs/MWCNTs was confirmed by SEM, EDAX, and XRD tests. Coating the PMA scaffold with Cs/MWCNTs leads to the viability and proliferation of MG-63 cells and more secretion of alkaline phosphatase and Ca activity, which can be introduced as a suitable candidate for use in bone tissue engineering.
Collapse
Affiliation(s)
| | - Saeed Karbasi
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Dental Implants Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Soheila Zamanlui Benisi
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Shahrokh Shojaei
- Department of Biomedical Engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran; Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
198
|
Seo JY, Park SB, Kim SY, Seo GJ, Jang HK, Lee TJ. Acoustic and Magnetic Stimuli-Based Three-Dimensional Cell Culture Platform for Tissue Engineering. Tissue Eng Regen Med 2023; 20:563-580. [PMID: 37052782 PMCID: PMC10313605 DOI: 10.1007/s13770-023-00539-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/16/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023] Open
Abstract
In a conventional two-dimensional (2D) culture method, cells are attached to the bottom of the culture dish and grow into a monolayer. These 2D culture methods are easy to handle, cost-effective, reproducible, and adaptable to growing many different types of cells. However, monolayer 2D cell culture conditions are far from those of natural tissue, indicating the need for a three-dimensional (3D) culture system. Various methods, such as hanging drop, scaffolds, hydrogels, microfluid systems, and bioreactor systems, have been utilized for 3D cell culture. Recently, external physical stimulation-based 3D cell culture platforms, such as acoustic and magnetic forces, were introduced. Acoustic waves can establish acoustic radiation force, which can induce suspended objects to gather in the pressure node region and aggregate to form clusters. Magnetic targeting consists of two components, a magnetically responsive carrier and a magnetic field gradient source. In a magnetic-based 3D cell culture platform, cells are aggregated by changing the magnetic force. Magnetic fields can manipulate cells through two different methods: positive magnetophoresis and negative magnetophoresis. Positive magnetophoresis is a way of imparting magnetic properties to cells by labeling them with magnetic nanoparticles. Negative magnetophoresis is a label-free principle-based method. 3D cell structures, such as spheroids, 3D network structures, and cell sheets, have been successfully fabricated using this acoustic and magnetic stimuli-based 3D cell culture platform. Additionally, fabricated 3D cell structures showed enhanced cell behavior, such as differentiation potential and tissue regeneration. Therefore, physical stimuli-based 3D cell culture platforms could be promising tools for tissue engineering.
Collapse
Affiliation(s)
- Ju Yeon Seo
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
- Department of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Song Bin Park
- Department of Bio-Health Technology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Seo Yeon Kim
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Gyeong Jin Seo
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Hyeon-Ki Jang
- Division of Chemical Engineering and Bioengineering, College of Art Culture and Engineering, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Tae-Jin Lee
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea.
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea.
| |
Collapse
|
199
|
Laubach M, Hildebrand F, Suresh S, Wagels M, Kobbe P, Gilbert F, Kneser U, Holzapfel BM, Hutmacher DW. The Concept of Scaffold-Guided Bone Regeneration for the Treatment of Long Bone Defects: Current Clinical Application and Future Perspective. J Funct Biomater 2023; 14:341. [PMID: 37504836 PMCID: PMC10381286 DOI: 10.3390/jfb14070341] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/31/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
The treatment of bone defects remains a challenging clinical problem with high reintervention rates, morbidity, and resulting significant healthcare costs. Surgical techniques are constantly evolving, but outcomes can be influenced by several parameters, including the patient's age, comorbidities, systemic disorders, the anatomical location of the defect, and the surgeon's preference and experience. The most used therapeutic modalities for the regeneration of long bone defects include distraction osteogenesis (bone transport), free vascularized fibular grafts, the Masquelet technique, allograft, and (arthroplasty with) mega-prostheses. Over the past 25 years, three-dimensional (3D) printing, a breakthrough layer-by-layer manufacturing technology that produces final parts directly from 3D model data, has taken off and transformed the treatment of bone defects by enabling personalized therapies with highly porous 3D-printed implants tailored to the patient. Therefore, to reduce the morbidities and complications associated with current treatment regimens, efforts have been made in translational research toward 3D-printed scaffolds to facilitate bone regeneration. Three-dimensional printed scaffolds should not only provide osteoconductive surfaces for cell attachment and subsequent bone formation but also provide physical support and containment of bone graft material during the regeneration process, enhancing bone ingrowth, while simultaneously, orthopaedic implants supply mechanical strength with rigid, stable external and/or internal fixation. In this perspective review, we focus on elaborating on the history of bone defect treatment methods and assessing current treatment approaches as well as recent developments, including existing evidence on the advantages and disadvantages of 3D-printed scaffolds for bone defect regeneration. Furthermore, it is evident that the regulatory framework and organization and financing of evidence-based clinical trials remains very complex, and new challenges for non-biodegradable and biodegradable 3D-printed scaffolds for bone regeneration are emerging that have not yet been sufficiently addressed, such as guideline development for specific surgical indications, clinically feasible design concepts for needed multicentre international preclinical and clinical trials, the current medico-legal status, and reimbursement. These challenges underscore the need for intensive exchange and open and honest debate among leaders in the field. This goal can be addressed in a well-planned and focused stakeholder workshop on the topic of patient-specific 3D-printed scaffolds for long bone defect regeneration, as proposed in this perspective review.
Collapse
Affiliation(s)
- Markus Laubach
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Frank Hildebrand
- Department of Orthopaedics, Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Sinduja Suresh
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Michael Wagels
- Department of Plastic Surgery, Princess Alexandra Hospital, Woolloongabba, QLD 4102, Australia;
- The Herston Biofabrication Institute, The University of Queensland, Herston, QLD 4006, Australia
- Southside Clinical Division, School of Medicine, University of Queensland, Woolloongabba, QLD 4102, Australia
- Department of Plastic and Reconstructive Surgery, Queensland Children’s Hospital, South Brisbane, QLD 4101, Australia
- The Australian Centre for Complex Integrated Surgical Solutions, Woolloongabba, QLD 4102, Australia
| | - Philipp Kobbe
- Department of Orthopaedics, Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Fabian Gilbert
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Ulrich Kneser
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, 67071 Ludwigshafen, Germany
| | - Boris M. Holzapfel
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Dietmar W. Hutmacher
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, QLD 4000, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies (CTET), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| |
Collapse
|
200
|
Sankaranarayanan A, Ramprasad A, Shree Ganesh S, Ganesh H, Ramanathan B, Shanmugavadivu A, Selvamurugan N. Nanogels for bone tissue engineering - from synthesis to application. NANOSCALE 2023. [PMID: 37305943 DOI: 10.1039/d3nr01246h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nanogels are cross-linked hydrogel nanoparticles with a three-dimensional, tunable porous structure that merges the best features of hydrogels and nanoparticles, including the ability to retain their hydrated nature and to swell and shrink in response to environmental changes. Nanogels have attracted increasing attention for use in bone tissue engineering as scaffolds for growth factor transport and cell adhesion. Their three-dimensional structures allow the encapsulation of a wide range of hydrophobic and hydrophilic drugs, enhance their half-life, and impede their enzymatic breakdown in vivo. Nanogel-based scaffolds are a viable treatment modality for enhanced bone regeneration. They act as carriers for cells and active ingredients capable of controlled release, enhanced mechanical support, and osteogenesis for enhanced bone tissue regeneration. However, the development of such nanogel constructs might involve combinations of several biomaterials to fabricate active ingredients that can control release, enhance mechanical support, and facilitate osteogenesis for more effective bone tissue regeneration. Hence, this review aims to highlight the potential of nanogel-based scaffolds to address the needs of bone tissue engineering.
Collapse
Affiliation(s)
- Aravind Sankaranarayanan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Anushikaa Ramprasad
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - S Shree Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Harini Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Bharathi Ramanathan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| |
Collapse
|