1
|
Ghiasi Tabari P, Sattari A, Mashhadi Keshtiban M, Karkuki Osguei N, Hardy JG, Samadikuchaksaraei A. Injectable hydrogel scaffold incorporating microspheres containing cobalt-doped bioactive glass for bone healing. J Biomed Mater Res A 2024; 112:2225-2242. [PMID: 38984402 DOI: 10.1002/jbm.a.37773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/07/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024]
Abstract
Injectable in situ-forming scaffolds that induce both angiogenesis and osteogenesis have been proven to be promising for bone healing applications. Here, we report the synthesis of an injectable hydrogel containing cobalt-doped bioactive glass (BG)-loaded microspheres. Silk fibroin (SF)/gelatin microspheres containing BG particles were fabricated through microfluidics. The microspheres were mixed in an injectable alginate solution, which formed an in situ hydrogel by adding CaCl2. The hydrogel was evaluated for its physicochemical properties, in vitro interactions with osteoblast-like and endothelial cells, and bone healing potential in a rat model of calvarial defect. The microspheres were well-dispersed in the hydrogel and formed pores of >100 μm. The hydrogel displayed shear-thinning behavior and modulated the cobalt release so that the optimal cobalt concentration for angiogenic stimulation, cell proliferation, and deposition of mineralized matrix was only achieved by the scaffold that contained BG doped with 5% wt/wt cobalt (A-S-G5Co). In the scaffold containing higher cobalt content, a reduced biomimetic mineralization on the surface was observed. The gene expression study indicated an upregulation of the osteogenic genes of COL1A1, ALPL, OCN, and RUNX2 and angiogenic genes of HIF1A and VEGF at different time points in the cells cultured with the A-S-G5Co. Finally, the in vivo study demonstrated that A-S-G5Co significantly promoted both angiogenesis and osteogenesis and improved bone healing after 12 weeks of follow-up. These results show that incorporation of SF/gelatin microspheres containing cobalt-doped BG in an injectable in situ-forming scaffold can effectively enhance its bone healing potential through promotion of angiogenesis and osteogenesis.
Collapse
Affiliation(s)
- Parmida Ghiasi Tabari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirmohammad Sattari
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | | | | - John G Hardy
- Department of Chemistry, Lancaster University, Lancaster, UK
- Materials Science Lancaster, Lancaster University, Lancaster, UK
| | - Ali Samadikuchaksaraei
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Borciani G, Montalbano G, Perut F, Ciapetti G, Baldini N, Vitale-Brovarone C. Osteoblast and osteoclast activity on collagen-based 3D printed scaffolds enriched with strontium-doped bioactive glasses and hydroxyapatite nanorods for bone tissue engineering. Biomed Mater 2024; 19:065007. [PMID: 39173660 DOI: 10.1088/1748-605x/ad72c3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024]
Abstract
Bone tissue engineering (BTE) aims to promote bone regeneration by means of the synergistic effect of biomaterials, cells, and other factors, as potential alternative to conventional treatments for bone fractures. To this aim, a composite material was developed, based on collagen type I, strontium-enriched mesoporous bioactive glasses, and hydroxyapatite nanorods as bioactive and biomimetic components. Nanostructured scaffolds were 3D printed and subsequently chemically crosslinked with genipin to improve mechanical properties and stability. The developed nanostructured system was maintained in culture until 3 weeks with a co-culture of human bone cells to provide anex vivomodel of bone microenvironment and examine the cellular crosstalk and signaling pathways through paracrine cell activities. Human osteoblasts (OBs), derived from trabecular bone, and human osteoclast precursors (OCs), isolated from buffy coat samples were involved, with OBs seeded on the scaffold and OC precursors seeded in a transwell device. When compared to the material without inorganic components, the bioactive and biomimetic scaffold positively influenced cell proliferation and cell metabolic activity, boosting alkaline phosphatase activity of OBs, and reducing OC differentiation. Thus, the bioactive and biomimetic system promoted an enhanced cellular response, highlighting its potential application in BTE.
Collapse
Affiliation(s)
- Giorgia Borciani
- Biomedical Science and Technologies and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Giorgia Montalbano
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Francesca Perut
- Biomedical Science and Technologies and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Gabriela Ciapetti
- Biomedical Science and Technologies and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Nicola Baldini
- Biomedical Science and Technologies and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Chiara Vitale-Brovarone
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
3
|
Seifi S, Shamloo A, Barzoki AK, Bakhtiari MA, Zare S, Cheraghi F, Peyrovan A. Engineering biomimetic scaffolds for bone regeneration: Chitosan/alginate/polyvinyl alcohol-based double-network hydrogels with carbon nanomaterials. Carbohydr Polym 2024; 339:122232. [PMID: 38823905 DOI: 10.1016/j.carbpol.2024.122232] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/23/2024] [Accepted: 05/02/2024] [Indexed: 06/03/2024]
Abstract
In this study, new types of hybrid double-network (DN) hydrogels composed of polyvinyl alcohol (PVA), chitosan (CH), and sodium alginate (SA) are introduced, with the hypothesis that this combination and incorporating multi-walled carbon nanotubes (MWCNTs) and graphene nanoplatelets (GNPs) will enhance osteogenetic differentiation and the structural and mechanical properties of scaffolds for bone tissue engineering applications. Initially, the impact of varying mass ratios of the PVA/CH/SA mixture on mechanical properties, swelling ratio, and degradability was examined. Based on this investigation, a mass ratio of 4:6:6 was determined to be optimal. At this ratio, the hydrogel demonstrated a Young's modulus of 47.5 ± 5 kPa, a swelling ratio of 680 ± 6 % after 3 h, and a degradation rate of 46.5 ± 5 % after 40 days. In the next phase, following the determination of the optimal mass ratio, CNTs and GNPs were incorporated into the 4:6:6 composite resulting in a significant enhancement in the electrical conductivity and stiffness of the scaffolds. The introduction of CNTs led to a notable increase of 36 % in the viability of MG63 osteoblast cells. Additionally, the inhibition zone test revealed that GNPs and CNTs increased the diameter of the inhibition zone by 49.6 % and 52.6 %, respectively.
Collapse
Affiliation(s)
- Saeed Seifi
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran 11155-9161, Iran
| | - Amir Shamloo
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran 11155-9161, Iran.
| | - Ali Kheirkhah Barzoki
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran 11155-9161, Iran
| | - Mohammad Ali Bakhtiari
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran 11155-9161, Iran
| | - Sona Zare
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran 11155-9161, Iran; Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Cheraghi
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., P.O. Box 11155-9466, Tehran, Iran
| | - Aisan Peyrovan
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran 11155-9161, Iran; Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Forte YS, Nascimento-Silva V, Andrade-Santos C, Ramos-Andrade I, Atella GC, Kraemer-Aguiar LG, Leal PRF, Renovato-Martins M, Barja-Fidalgo C. Unlocking the Secrets of Adipose Tissue: How an Obesity-Associated Secretome Promotes Osteoblast Dedifferentiation via TGF-β1 Signaling, Paving the Path to an Adipogenic Phenotype. Cells 2024; 13:1418. [PMID: 39272990 PMCID: PMC11394205 DOI: 10.3390/cells13171418] [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: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Obesity poses a significant global health challenge, given its association with the excessive accumulation of adipose tissue (AT) and various systemic disruptions. Within the adipose microenvironment, expansion and enrichment with immune cells trigger the release of inflammatory mediators and growth factors, which can disrupt tissues, including bones. While obesity's contribution to bone loss is well established, the direct impact of obese AT on osteoblast maturation remains uncertain. This study aimed to explore the influence of the secretomes from obese and lean AT on osteoblast differentiation and activity. METHODS SAOS-2 cells were exposed to the secretomes obtained by culturing human subcutaneous AT from individuals with obesity (OATS) or lean patients, and their effects on osteoblasts were evaluated. RESULTS In the presence of the OATS, mature osteoblasts underwent dedifferentiation, showing an increased proliferation accompanied by a morphological shift towards a mesenchymal phenotype, with detrimental effects on osteogenic markers and the calcification capacity. Concurrently, the OATS promoted the expression of mesenchymal and adipogenic markers, inducing the formation of cytoplasmic lipid droplets in SAOS-2 cells exposed to an adipogenic differentiation medium. Additionally, TGF-β1 emerged as a key mediator of these effects, as the OATS was enriched with this growth factor. CONCLUSIONS Our findings demonstrate that obese subcutaneous AT promotes the dedifferentiation of osteoblasts and increases the adipogenic profile in these cells.
Collapse
Affiliation(s)
- Yasmin Silva Forte
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| | - Vany Nascimento-Silva
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| | - Caio Andrade-Santos
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| | - Isadora Ramos-Andrade
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| | - Georgia Correa Atella
- Institute of Medical Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Luiz Guilherme Kraemer-Aguiar
- Obesity Unit, Multiuser Clinical Research Center (CePEM), Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (L.G.K.-A.); (P.R.F.L.)
| | - Paulo Roberto Falcão Leal
- Obesity Unit, Multiuser Clinical Research Center (CePEM), Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (L.G.K.-A.); (P.R.F.L.)
| | - Mariana Renovato-Martins
- Department of Molecular & Cellular Biology, Universidade Federal Fluminense, Rio de Janeiro 24020-141, Brazil;
| | - Christina Barja-Fidalgo
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| |
Collapse
|
5
|
Chen A, Wang W, Mao Z, He Y, Chen S, Liu G, Su J, Feng P, Shi Y, Yan C, Lu J. Multimaterial 3D and 4D Bioprinting of Heterogenous Constructs for Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307686. [PMID: 37737521 DOI: 10.1002/adma.202307686] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/06/2023] [Indexed: 09/23/2023]
Abstract
Additive manufacturing (AM), which is based on the principle of layer-by-layer shaping and stacking of discrete materials, has shown significant benefits in the fabrication of complicated implants for tissue engineering (TE). However, many native tissues exhibit anisotropic heterogenous constructs with diverse components and functions. Consequently, the replication of complicated biomimetic constructs using conventional AM processes based on a single material is challenging. Multimaterial 3D and 4D bioprinting (with time as the fourth dimension) has emerged as a promising solution for constructing multifunctional implants with heterogenous constructs that can mimic the host microenvironment better than single-material alternatives. Notably, 4D-printed multimaterial implants with biomimetic heterogenous architectures can provide a time-dependent programmable dynamic microenvironment that can promote cell activity and tissue regeneration in response to external stimuli. This paper first presents the typical design strategies of biomimetic heterogenous constructs in TE applications. Subsequently, the latest processes in the multimaterial 3D and 4D bioprinting of heterogenous tissue constructs are discussed, along with their advantages and challenges. In particular, the potential of multimaterial 4D bioprinting of smart multifunctional tissue constructs is highlighted. Furthermore, this review provides insights into how multimaterial 3D and 4D bioprinting can facilitate the realization of next-generation TE applications.
Collapse
Affiliation(s)
- Annan Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Wanying Wang
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| | - Zhengyi Mao
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Yunhu He
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Shiting Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Guo Liu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Jin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Pei Feng
- State Key Laboratory of High-Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Yusheng Shi
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Chunze Yan
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Jian Lu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Hong Kong Branch of National Precious Metals Material Engineering Research, Center (NPMM), City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
6
|
Re F, Sartore L, Pasini C, Ferroni M, Borsani E, Pandini S, Bianchetti A, Almici C, Giugno L, Bresciani R, Mutti S, Trenta F, Bernardi S, Farina M, Russo D. In Vitro Biocompatibility Assessment of Bioengineered PLA-Hydrogel Core-Shell Scaffolds with Mesenchymal Stromal Cells for Bone Regeneration. J Funct Biomater 2024; 15:217. [PMID: 39194655 DOI: 10.3390/jfb15080217] [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: 06/28/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Human mesenchymal stromal cells (hMSCs), whether used alone or together with three-dimensional scaffolds, are the best-studied postnatal stem cells in regenerative medicine. In this study, innovative composite scaffolds consisting of a core-shell architecture were seeded with bone-marrow-derived hMSCs (BM-hMSCs) and tested for their biocompatibility and remarkable capacity to promote and support bone regeneration and mineralization. The scaffolds were prepared by grafting three different amounts of gelatin-chitosan (CH) hydrogel into a 3D-printed polylactic acid (PLA) core (PLA-CH), and the mechanical and degradation properties were analyzed. The BM-hMSCs were cultured in the scaffolds with the presence of growth medium (GM) or osteogenic medium (OM) with differentiation stimuli in combination with fetal bovine serum (FBS) or human platelet lysate (hPL). The primary objective was to determine the viability, proliferation, morphology, and spreading capacity of BM-hMSCs within the scaffolds, thereby confirming their biocompatibility. Secondly, the BM-hMSCs were shown to differentiate into osteoblasts and to facilitate scaffold mineralization. This was evinced by a positive Von Kossa result, the modulation of differentiation markers (osteocalcin and osteopontin), an expression of a marker of extracellular matrix remodeling (bone morphogenetic protein-2), and collagen I. The results of the energy-dispersive X-ray analysis (EDS) clearly demonstrate the presence of calcium and phosphorus in the samples that were incubated in OM, in the presence of FBS and hPL, but not in GM. The chemical distribution maps of calcium and phosphorus indicate that these elements are co-localized in the same areas of the sections, demonstrating the formation of hydroxyapatite. In conclusion, our findings show that the combination of BM-hMSCs and PLA-CH, regardless of the amount of hydrogel content, in the presence of differentiation stimuli, can provide a construct with enhanced osteogenicity for clinically relevant bone regeneration.
Collapse
Affiliation(s)
- Federica Re
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
| | - Luciana Sartore
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Materials Science and Technology Laboratory, Department of Mechanical and Industrial Engineering, University of Brescia, 25123 Brescia, Italy
| | - Chiara Pasini
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Materials Science and Technology Laboratory, Department of Mechanical and Industrial Engineering, University of Brescia, 25123 Brescia, Italy
| | - Matteo Ferroni
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Department of Civil, Environmental, Architectural Engineering and Mathematics (DICATAM), University of Brescia, Via Valotti 9, 25123 Brescia, Italy
- National Research Council (CNR)-Institute for Microelectronics and Microsystems, Via Gobetti 101, 40129 Bologna, Italy
| | - Elisa Borsani
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs (ARTO)", University of Brescia, 25123 Brescia, Italy
| | - Stefano Pandini
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Materials Science and Technology Laboratory, Department of Mechanical and Industrial Engineering, University of Brescia, 25123 Brescia, Italy
| | - Andrea Bianchetti
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Laboratory for Stem Cells Manipulation and Cryopreservation, Department of Transfusion Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Camillo Almici
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Laboratory for Stem Cells Manipulation and Cryopreservation, Department of Transfusion Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Lorena Giugno
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Roberto Bresciani
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Highly Specialized Laboratory, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Silvia Mutti
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
| | - Federica Trenta
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
| | - Simona Bernardi
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- National Center for Gene Therapy and Drugs based on RNA Technology-CN3, 35122 Padua, Italy
| | - Mirko Farina
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
7
|
Agostinacchio F, Biada E, Gambari L, Grassi F, Bucciarelli A, Motta A. Surfactant-assisted photo-crosslinked silk fibroin sponges: A versatile platform for the design of bone scaffolds. BIOMATERIALS ADVANCES 2024; 161:213887. [PMID: 38735199 DOI: 10.1016/j.bioadv.2024.213887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/16/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Critical size bone defects cannot heal without aid and current clinical approaches exhibit some limitations, underling the need for novel solutions. Silk fibroin, derived from silkworms, is widely utilized in tissue engineering and regenerative medicine due to its remarkable properties, making it a promising candidate for bone tissue regeneration in vitro and in vivo. However, the clinical translation of silk-based materials requires refinements in 3D architecture, stability, and biomechanical properties. In earlier research, improved mechanical resistance and stability of chemically crosslinked methacrylate silk fibroin (Sil-Ma) sponges over physically crosslinked counterparts were highlighted. Furthermore, the influence of photo-initiator and surfactant concentrations on silk properties was investigated. However, the characterization of sponges with Sil-Ma solution concentrations above 10 % (w/V) was hindered by production optimization challenges, with only cell viability assessed. This study focuses on the evaluation of methacrylate sponges' suitability as temporal bone tissue regeneration scaffolds. Sil-Ma sponge fabrication at a fixed concentration of 20 % (w/V) was optimized and the impact of photo-initiator (LAP) concentrations and surfactant (Tween 80) presence/absence was studied. Their effects on pore formation, silk secondary structure, mechanical properties, and osteogenic differentiation of hBM-MSCs were investigated. We demonstrated that, by tuning silk sponges' composition, the optimal combination boosted osteogenic gene expression, offering a strategy to tailor biomechanical properties for effective bone regeneration. Utilizing Design of Experiment (DoE), correlations between sponge composition, porosity, and mechanical properties are established, guiding tailored material outcomes. Additionally, correlation matrices elucidate the microstructure's influence on gene expressions, providing insights for personalized approaches in bone tissue regeneration.
Collapse
Affiliation(s)
- Francesca Agostinacchio
- National Interuniversity Consortium of Material Science and Technology, Florence, Italy; BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Department of Industrial Engineering, University of Trento, Trento, Italy
| | - Elisa Biada
- BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Department of Industrial Engineering, University of Trento, Trento, Italy
| | - Laura Gambari
- Laboratorio Ramses, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesco Grassi
- Laboratorio Ramses, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Antonella Motta
- BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Department of Industrial Engineering, University of Trento, Trento, Italy.
| |
Collapse
|
8
|
Pecorini G, Braccini S, Simoni S, Corti A, Parrini G, Puppi D. Additive Manufacturing of Wet-Spun Poly(3-hydroxybutyrate-co-3-hydroxyvalerate)-Based Scaffolds Loaded with Hydroxyapatite. Macromol Biosci 2024; 24:e2300538. [PMID: 38534197 DOI: 10.1002/mabi.202300538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/20/2024] [Indexed: 03/28/2024]
Abstract
Tissue engineering represents an advanced therapeutic approach for the treatment of bone tissue defects. Polyhydroxyalkanoates are a promising class of natural polymers in this context thanks to their biocompatibility, processing versatility, and mechanical properties. The aim of this study is the development by computer-aided wet-spinning of novel poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV)-based composite scaffolds for bone engineering. In particular, PHBV scaffolds are loaded with hydroxyapatite (HA), an osteoinductive ceramic, in order to tailor their biological activity and mechanical properties. PHBV blending with poly(lactide-co-glycolide) (PLGA) is also explored to increase the processing properties of the polymeric mixture used for composite scaffold fabrication. Different HA percentages, up to 15% wt., can be loaded into the PHBV or PHBV/PLGA scaffolds without compromising their interconnected porous architecture, as well as the polymer morphological and thermal properties, as demonstrated by scanning electron microscopy, thermogravimetric analysis, and differential scanning calorimetry. In addition, HA loading results in increased scaffold compressive stiffness to levels comparable to those of trabecular bone tissue, as well as in higher in vitro MC3T3-E1 cell viability and production of mineralized extracellular matrix, in comparison to what observed for unloaded scaffolds. The observed mechanical and biological properties suggest the suitability of the developed scaffolds for bone engineering.
Collapse
Affiliation(s)
- Gianni Pecorini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | - Simona Braccini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | - Stefano Simoni
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | - Andrea Corti
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | | | - Dario Puppi
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| |
Collapse
|
9
|
Moghaddam A, Bahrami M, Mirzadeh M, Khatami M, Simorgh S, Chimehrad M, Kruppke B, Bagher Z, Mehrabani D, Khonakdar HA. Recent trends in bone tissue engineering: a review of materials, methods, and structures. Biomed Mater 2024; 19:042007. [PMID: 38636500 DOI: 10.1088/1748-605x/ad407d] [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: 09/23/2023] [Accepted: 04/18/2024] [Indexed: 04/20/2024]
Abstract
Bone tissue engineering (BTE) provides the treatment possibility for segmental long bone defects that are currently an orthopedic dilemma. This review explains different strategies, from biological, material, and preparation points of view, such as using different stem cells, ceramics, and metals, and their corresponding properties for BTE applications. In addition, factors such as porosity, surface chemistry, hydrophilicity and degradation behavior that affect scaffold success are introduced. Besides, the most widely used production methods that result in porous materials are discussed. Gene delivery and secretome-based therapies are also introduced as a new generation of therapies. This review outlines the positive results and important limitations remaining in the clinical application of novel BTE materials and methods for segmental defects.
Collapse
Affiliation(s)
| | - Mehran Bahrami
- Department of Mechanical Engineering and Mechanics, Lehigh University, 27 Memorial Dr W, Bethlehem, PA 18015, United States of America
| | | | - Mehrdad Khatami
- Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Chimehrad
- Department of Mechanical & Aerospace Engineering, College of Engineering & Computer Science, University of Central Florida, Orlando, FL, United States of America
| | - Benjamin Kruppke
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, 01069 Dresden, Germany
| | - Zohreh Bagher
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Mehrabani
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71345-1744, Iran
| | - Hossein Ali Khonakdar
- Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, 01069 Dresden, Germany
| |
Collapse
|
10
|
Quek J, Vizetto-Duarte C, Teoh SH, Choo Y. Towards Stem Cell Therapy for Critical-Sized Segmental Bone Defects: Current Trends and Challenges on the Path to Clinical Translation. J Funct Biomater 2024; 15:145. [PMID: 38921519 PMCID: PMC11205181 DOI: 10.3390/jfb15060145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
The management and reconstruction of critical-sized segmental bone defects remain a major clinical challenge for orthopaedic clinicians and surgeons. In particular, regenerative medicine approaches that involve incorporating stem cells within tissue engineering scaffolds have great promise for fracture management. This narrative review focuses on the primary components of bone tissue engineering-stem cells, scaffolds, the microenvironment, and vascularisation-addressing current advances and translational and regulatory challenges in the current landscape of stem cell therapy for critical-sized bone defects. To comprehensively explore this research area and offer insights for future treatment options in orthopaedic surgery, we have examined the latest developments and advancements in bone tissue engineering, focusing on those of clinical relevance in recent years. Finally, we present a forward-looking perspective on using stem cells in bone tissue engineering for critical-sized segmental bone defects.
Collapse
Affiliation(s)
- Jolene Quek
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| | - Catarina Vizetto-Duarte
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| | - Swee Hin Teoh
- Centre for Advanced Medical Engineering, College of Materials Science and Engineering, Hunan University, Changsha 410012, China
| | - Yen Choo
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| |
Collapse
|
11
|
Seo YH, Lee JM, Park SY, Kim MH, Kim SB, Oh TH. PVA Hydrogels Supplemented with PLA Mesh for Tissue Regeneration Scaffold. Gels 2024; 10:364. [PMID: 38920911 PMCID: PMC11202865 DOI: 10.3390/gels10060364] [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: 04/28/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
This study examined the tensile strength and biocompatibility properties of polyvinyl alcohol (PVA) hydrogel tissue regeneration scaffolds with polylactic acid (PLA) mesh fabric added as reinforcement, with a focus on the impact of heat treatment temperature and the number of layers of the PLA mesh fabric. The hydrogel scaffolds were prepared using a freeze-thaw method to create PVA hydrogel, with the PLA mesh fabric placed inside the hydrogel. The swelling ratio of the PVA/PLA hydrogel scaffolds decreased with increasing layer number and heat treatment temperature of the PLA mesh. The gel strength was highest when five layers of PLA mesh fabric were added, heat-treated at 120 °C, and confirmed to be properly placed inside the hydrogel by SEM images. The MTT assay and DAPI staining using HaCaT cells demonstrated that the cell proliferation was uninterrupted throughout the experimental period, confirming the biocompatibility of the scaffold. Therefore, we confirmed the possibility of using PLA mesh fabric as a reinforcement for PVA hydrogel to improve the strength of scaffolds for tissue regeneration, and we confirmed the potential of PLA mesh fabric as a reinforcement for various biomaterials.
Collapse
Affiliation(s)
- Young-Ho Seo
- Department of Advanced Organic Materials Engineering, Graduate School, Yeungnam University, Gyeongsan 38541, Republic of Korea;
- Institute for Future Earth, Pusan National University, Busan 46241, Republic of Korea; (S.-Y.P.); (M.-H.K.); (S.-B.K.)
| | - Jae-Man Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Sun-Young Park
- Institute for Future Earth, Pusan National University, Busan 46241, Republic of Korea; (S.-Y.P.); (M.-H.K.); (S.-B.K.)
| | - Myung-Hoo Kim
- Institute for Future Earth, Pusan National University, Busan 46241, Republic of Korea; (S.-Y.P.); (M.-H.K.); (S.-B.K.)
- Department of Animal Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Seon-Beom Kim
- Institute for Future Earth, Pusan National University, Busan 46241, Republic of Korea; (S.-Y.P.); (M.-H.K.); (S.-B.K.)
- Department of Food Science & Technology, Pusan National University, Miryang 50463, Republic of Korea
| | - Tae-Hwan Oh
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
12
|
Thangavelu M, Kim PY, Cho H, Song JE, Park S, Bucciarelli A, Khang G. A Gellan Gum, Polyethylene Glycol, Hydroxyapatite Composite Scaffold with the Addition of Ginseng Derived Compound K with Possible Applications in Bone Regeneration. Gels 2024; 10:257. [PMID: 38667676 PMCID: PMC11049517 DOI: 10.3390/gels10040257] [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: 03/08/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Engineered bone scaffolds should mimic the natural material to promote cell adhesion and regeneration. For this reason, natural biopolymers are becoming a gold standard in scaffold production. In this study, we proposed a hybrid scaffold produced using gellan gum, hydroxyapatite, and Poly (ethylene glycol) within the addition of the ginseng compound K (CK) as a candidate for bone regeneration. The fabricated scaffold was physiochemically characterized. The morphology studied by scanning electron microscopy (SEM) and image analysis revealed a pore distribution suitable for cells growth. The addition of CK further improved the biological activity of the hybrid scaffold as demonstrated by the MTT assay. The addition of CK influenced the scaffold morphology, decreasing the mean pore diameter. These findings can potentially help the development of a new generation of hybrid scaffolds to best mimic the natural tissue.
Collapse
Affiliation(s)
| | - Pil-Yun Kim
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea; (P.-Y.K.); (H.C.); (J.-E.S.)
| | - Hunhwi Cho
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea; (P.-Y.K.); (H.C.); (J.-E.S.)
| | - Jeong-Eun Song
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea; (P.-Y.K.); (H.C.); (J.-E.S.)
| | - Sunjae Park
- Department of Polymer Nano Science & Technology and Polymer Materials Fusion Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea;
| | - Alessio Bucciarelli
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Gilson Khang
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea; (P.-Y.K.); (H.C.); (J.-E.S.)
| |
Collapse
|
13
|
Moghaddasi M, Özdemir MMM, Noshahr AT, Özadenç HM, Oktay B, Bingöl AB, Arayıcı PP, Eraslan A, Şenel İ, Chifiriuc MC, Üstündağ CB. Blend Electrospinning of Nigella sativa-Incorporating PCL/PLA/HA Fibers and Its Investigation for Bone Healing Applications. ACS OMEGA 2024; 9:10267-10275. [PMID: 38463250 PMCID: PMC10918662 DOI: 10.1021/acsomega.3c07523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/25/2023] [Accepted: 01/16/2024] [Indexed: 03/12/2024]
Abstract
One of the well-known postoperative complications that requires a number of prophylactic and curative treatments is infection. The implications of postsurgical infections are further exacerbated by the emergence of antibiotic-resistant strains. Reduced effectiveness of synthetic antibiotics has led to an interest in plant-based substances. Extracts obtained from Nigella sativa have been shown to possess effective anti-infectious agents against bacteria frequently seen in bone infections. In this study, a fiber-based bone scaffold containing polycaprolactone, poly(lactic acid), and hydroxyapatite with N. sativa oil at varying concentrations was developed. Solvent electrospinning was used to fabricate the fibers with the specified composition. According to FE-SEM analysis, fibers with average diameters of 751 ± 82, 1000 ± 100, 1020 ± 90, and 1223 ± 112 nm were formed and successful integration of N. sativa oil into the fiber's structure was confirmed via FTIR. Staphylococcus aureus showed moderate susceptibility against the fibers with a maximum inhibition zone diameter of 11.5 ± 1.6 mm. MTT assay analysis exhibited concentration-dependent cell toxicity against fibroblast cells. In short, the antibacterial fibers synthesized in this study possessed antibacterial properties while also allowing moderate accommodation of CDD fibroblast cells at low oil concentrations, which can be a potential application for bone healing and mitigating postsurgical infections.
Collapse
Affiliation(s)
- Mohammad Moghaddasi
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
| | - Muhammed Mustafa Mert Özdemir
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
| | - Ali Torabkhani Noshahr
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
| | - Hüseyin Murat Özadenç
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
| | - Busra Oktay
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
| | - Ayşe Betül Bingöl
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
| | - Pelin Pelit Arayıcı
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, 34220 Istanbul, Türkiye
| | - Azime Eraslan
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, 34220 Istanbul, Türkiye
| | - İlkay Şenel
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, 34220 Istanbul, Türkiye
- Central Research Laboratory, Yıldız Technical University, Esenler, 34220 Istanbul, Türkiye
| | - Mariana Carmen Chifiriuc
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
- Research Institute of the University of Bucharest (ICUB), 050568 Bucharest, Romania
- Romanian Academy, 050045 Bucharest, Romania
| | - Cem Bülent Üstündağ
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, 34220 Istanbul, Türkiye
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, 34220 Istanbul, Türkiye
| |
Collapse
|
14
|
Shanmugavadivu A, Lekhavadhani S, Miranda PJ, Selvamurugan N. Current approaches in tissue engineering-based nanotherapeutics for osteosarcoma treatment. Biomed Mater 2024; 19:022003. [PMID: 38324905 DOI: 10.1088/1748-605x/ad270b] [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: 09/16/2023] [Accepted: 02/07/2024] [Indexed: 02/09/2024]
Abstract
Osteosarcoma (OS) is a malignant bone neoplasm plagued by poor prognosis. Major treatment strategies include chemotherapy, radiotherapy, and surgery. Chemotherapy to treat OS has severe adverse effects due to systemic toxicity to healthy cells. A possible way to overcome the limitation is to utilize nanotechnology. Nanotherapeutics is an emerging approach in treating OS using nanoparticulate drug delivery systems. Surgical resection of OS leaves a critical bone defect requiring medical intervention. Recently, tissue engineered scaffolds have been reported to provide physical support to bone defects and aid multimodal treatment of OS. These scaffolds loaded with nanoparticulate delivery systems could also actively repress tumor growth and aid new bone formation. The rapid developments in nanotherapeutics and bone tissue engineering have paved the way for improved treatment efficacy for OS-related bone defects. This review focuses on current bifunctional nanomaterials-based tissue engineered (NTE) scaffolds that use novel approaches such as magnetic hyperthermia, photodynamic therapy, photothermal therapy, bioceramic and polymeric nanotherapeutics against OS. With further optimization and screening, NTE scaffolds could meet clinical applications for treating OS patients.
Collapse
Affiliation(s)
- Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Sundaravadhanan Lekhavadhani
- 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
|
15
|
Subramaniam D, Sekaran S. In Vitro Biocompatibility Assessment of a Novel Membrane Containing Magnesium-Chitosan/Carboxymethyl Cellulose and Alginate Intended for Bone Tissue Regeneration. Cureus 2024; 16:e54597. [PMID: 38523973 PMCID: PMC10959466 DOI: 10.7759/cureus.54597] [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: 08/15/2023] [Accepted: 02/18/2024] [Indexed: 03/26/2024] Open
Abstract
Bone tissue engineering (BTE) is an emerging interdisciplinary field that aims to develop new strategies and materials for repairing, regenerating, or replacing damaged bone tissues. This field combines engineering, biology, and medicine principles to create functional bone tissues in the laboratory and in vivo. The main goal of BTE is to create biological substitutes that mimic the structure, function, and properties of natural bone tissue, thereby promoting the regeneration of bone defects caused by trauma, disease, or aging. In this study, we developed a biocomposite membrane using magnesium-chitosan, carboxymethyl cellulose, and alginate through a simple cast drying method. The biocompatibility of the membrane was evaluated using human osteoblastic cells, and it was found to be nontoxic to these cells. Both metabolic activity measurements (24 and 48 hours) and the lactate dehydrogenase release assay (72 hours) indicated that the membrane was biocompatible and did not exert significant toxic effects. These results suggest that the developed biocomposite membrane has the potential to be used as a safe and effective biomaterial for various biomedical applications, such as BTE, wound healing, and drug delivery. Further studies are warranted to explore the full potential of this membrane and its performance in different biological environments.
Collapse
Affiliation(s)
- Deepika Subramaniam
- General Pathology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Saravanan Sekaran
- Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
16
|
Salehi Abar E, Vandghanooni S, Torab A, Jaymand M, Eskandani M. A comprehensive review on nanocomposite biomaterials based on gelatin for bone tissue engineering. Int J Biol Macromol 2024; 254:127556. [PMID: 37884249 DOI: 10.1016/j.ijbiomac.2023.127556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023]
Abstract
The creation of a suitable scaffold is a crucial step in the process of bone tissue engineering (BTE). The scaffold, acting as an artificial extracellular matrix, plays a significant role in determining the fate of cells by affecting their proliferation and differentiation in BTE. Therefore, careful consideration should be given to the fabrication approach and materials used for scaffold preparation. Natural polypeptides such as gelatin and collagen have been widely used for this purpose. The unique properties of nanoparticles, which vary depending on their size, charge, and physicochemical properties, have demonstrated potential in solving various challenges encountered in BTE. Therefore, nanocomposite biomaterials consisting of polymers and nanoparticles have been extensively used for BTE. Gelatin has also been utilized in combination with other nanomaterials to apply for this purpose. Composites of gelatin with various types of nanoparticles are particularly promising for creating scaffolds with superior biological and physicochemical properties. This review explores the use of nanocomposite biomaterials based on gelatin and various types of nanoparticles together for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Elaheh Salehi Abar
- Department of Prosthodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Torab
- Department of Prosthodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
17
|
Wang YM, Shen JT. Chitosan-based promising scaffolds for the construction of tailored nanosystems against osteoporosis: Current status and future prospects. J Appl Biomater Funct Mater 2024; 22:22808000241266487. [PMID: 39129376 DOI: 10.1177/22808000241266487] [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: 08/13/2024] Open
Abstract
Despite advancements in therapeutic techniques, restoring bone tissue after damage remains a challenging task. Tissue engineering or targeted drug delivery solutions aim to meet the pressing clinical demand for treatment alternatives by creating substitute materials that imitate the structural and biological characteristics of healthy tissue. Polymers derived from natural sources typically exhibit enhanced biological compatibility and bioactivity when compared to manufactured polymers. Chitosan is a unique polysaccharide derived from chitin through deacetylation, offering biodegradability, biocompatibility, and antibacterial activity. Its cationic charge sets it apart from other polymers, making it a valuable resource for various applications. Modifications such as thiolation, alkylation, acetylation, or hydrophilic group incorporation can enhance chitosan's swelling behavior, cross-linking, adhesion, permeation, controllable drug release, enzyme inhibition, and antioxidative properties. Chitosan scaffolds possess considerable potential for utilization in several biological applications. An intriguing application is its use in the areas of drug distribution and bone tissue engineering. Due to their excellent biocompatibility and lack of toxicity, they are an optimal material for this particular usage. This article provides a comprehensive analysis of osteoporosis, including its pathophysiology, current treatment options, the utilization of natural polymers in disease management, and the potential use of chitosan scaffolds for drug delivery systems aimed at treating the condition.
Collapse
Affiliation(s)
- Ya-Ming Wang
- Department of Endocrine, Shengzhou People's Hospital (Shengzhou Branch of the First Affiliated Hospital of Zhejiang University School of Medicine, the Shengzhou Hospital of Shaoxing University), Shengzhou, Zhejiang, China
| | - Jiang-Tao Shen
- Department of Orthopedics, Shengzhou People's Hospital (Shengzhou Branch of the First Affiliated Hospital of Zhejiang University School of Medicine, the Shengzhou Hospital of Shaoxing University), Shengzhou, Zhejiang, China
| |
Collapse
|
18
|
Ahangar P, Li J, Nkindi LS, Mohammadrezaee Z, Cooke ME, Martineau PA, Weber MH, Saade E, Nateghi N, Rosenzweig DH. A Nanoporous 3D-Printed Scaffold for Local Antibiotic Delivery. MICROMACHINES 2023; 15:83. [PMID: 38258202 PMCID: PMC10819679 DOI: 10.3390/mi15010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/14/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024]
Abstract
Limitations of bone defect reconstruction include poor bone healing and osteointegration with acrylic cements, lack of strength with bone putty/paste, and poor osteointegration. Tissue engineering aims to bridge these gaps through the use of bioactive implants. However, there is often a risk of infection and biofilm formation associated with orthopedic implants, which may develop anti-microbial resistance. To promote bone repair while also locally delivering therapeutics, 3D-printed implants serve as a suitable alternative. Soft, nanoporous 3D-printed filaments made from a thermoplastic polyurethane and polyvinyl alcohol blend, LAY-FOMM and LAY-FELT, have shown promise for drug delivery and orthopedic applications. Here, we compare 3D printability and sustained antibiotic release kinetics from two types of commercial 3D-printed porous filaments suitable for bone tissue engineering applications. We found that both LAY-FOMM and LAY-FELT could be consistently printed into scaffolds for drug delivery. Further, the materials could sustainably release Tetracycline over 3 days, independent of material type and infill geometry. The drug-loaded materials did not show any cytotoxicity when cultured with primary human fibroblasts. We conclude that both LAY-FOMM and LAY-FELT 3D-printed scaffolds are suitable devices for local antibiotic delivery applications, and they may have potential applications to prophylactically reduce infections in orthopedic reconstruction surgery.
Collapse
Affiliation(s)
- Pouyan Ahangar
- Department of Surgery, McGill University, Montreal, QC H3G 1A4, Canada; (P.A.); (M.E.C.); (P.A.M.); (M.H.W.)
| | - Jialiang Li
- Department of Science, TAV College, Montreal, QC H3W 3E1, Canada; (J.L.); (L.S.N.); (Z.M.); (E.S.); (N.N.)
| | - Leslie S. Nkindi
- Department of Science, TAV College, Montreal, QC H3W 3E1, Canada; (J.L.); (L.S.N.); (Z.M.); (E.S.); (N.N.)
| | - Zohreh Mohammadrezaee
- Department of Science, TAV College, Montreal, QC H3W 3E1, Canada; (J.L.); (L.S.N.); (Z.M.); (E.S.); (N.N.)
| | - Megan E. Cooke
- Department of Surgery, McGill University, Montreal, QC H3G 1A4, Canada; (P.A.); (M.E.C.); (P.A.M.); (M.H.W.)
| | - Paul A. Martineau
- Department of Surgery, McGill University, Montreal, QC H3G 1A4, Canada; (P.A.); (M.E.C.); (P.A.M.); (M.H.W.)
| | - Michael H. Weber
- Department of Surgery, McGill University, Montreal, QC H3G 1A4, Canada; (P.A.); (M.E.C.); (P.A.M.); (M.H.W.)
| | - Elie Saade
- Department of Science, TAV College, Montreal, QC H3W 3E1, Canada; (J.L.); (L.S.N.); (Z.M.); (E.S.); (N.N.)
| | - Nima Nateghi
- Department of Science, TAV College, Montreal, QC H3W 3E1, Canada; (J.L.); (L.S.N.); (Z.M.); (E.S.); (N.N.)
| | - Derek H. Rosenzweig
- Department of Surgery, McGill University, Montreal, QC H3G 1A4, Canada; (P.A.); (M.E.C.); (P.A.M.); (M.H.W.)
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| |
Collapse
|
19
|
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
|
20
|
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: 1.0] [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
|
21
|
Lewns FK, Tsigkou O, Cox LR, Wildman RD, Grover LM, Poologasundarampillai G. Hydrogels and Bioprinting in Bone Tissue Engineering: Creating Artificial Stem-Cell Niches for In Vitro Models. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301670. [PMID: 37087739 PMCID: PMC11478930 DOI: 10.1002/adma.202301670] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Advances in bioprinting have enabled the fabrication of complex tissue constructs with high speed and resolution. However, there remains significant structural and biological complexity within tissues that bioprinting is unable to recapitulate. Bone, for example, has a hierarchical organization ranging from the molecular to whole organ level. Current bioprinting techniques and the materials employed have imposed limits on the scale, speed, and resolution that can be achieved, rendering the technique unable to reproduce the structural hierarchies and cell-matrix interactions that are observed in bone. The shift toward biomimetic approaches in bone tissue engineering, where hydrogels provide biophysical and biochemical cues to encapsulated cells, is a promising approach to enhancing the biological function and development of tissues for in vitro modeling. A major focus in bioprinting of bone tissue for in vitro modeling is creating dynamic microenvironmental niches to support, stimulate, and direct the cellular processes for bone formation and remodeling. Hydrogels are ideal materials for imitating the extracellular matrix since they can be engineered to present various cues whilst allowing bioprinting. Here, recent advances in hydrogels and 3D bioprinting toward creating a microenvironmental niche that is conducive to tissue engineering of in vitro models of bone are reviewed.
Collapse
Affiliation(s)
| | - Olga Tsigkou
- Department of MaterialsUniversity of ManchesterManchesterM1 5GFUK
| | - Liam R. Cox
- School of ChemistryUniversity of BirminghamBirminghamB15 2TTUK
| | - Ricky D. Wildman
- Faculty of EngineeringUniversity of NottinghamNottinghamNG7 2RDUK
| | - Liam M. Grover
- Healthcare Technologies InstituteSchool of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | | |
Collapse
|
22
|
Bar JK, Lis-Nawara A, Kowalczyk T, Grelewski PG, Stamnitz S, Gerber H, Klimczak A. Osteogenic Potential of Human Dental Pulp Stem Cells (hDPSCs) Growing on Poly L-Lactide-Co-Caprolactone and Hyaluronic Acid (HYAFF-11 TM) Scaffolds. Int J Mol Sci 2023; 24:16747. [PMID: 38069071 PMCID: PMC10705868 DOI: 10.3390/ijms242316747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/12/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Bone tissue engineering using different scaffolds is a new therapeutic approach in regenerative medicine. This study explored the osteogenic potential of human dental pulp stem cells (hDPSCs) grown on a hydrolytically modified poly(L-lactide-co-caprolactone) (PLCL) electrospun scaffold and a non-woven hyaluronic acid (HYAFF-11™) mesh. The adhesion, immunophenotype, and osteogenic differentiation of hDPSCs seeded on PLCL and HYAFF-11™ scaffolds were analyzed. The results showed that PLCL and HYAFF-11™ scaffolds significantly supported hDPSCs adhesion; however, hDPSCs' adhesion rate was significantly higher on PLCL than on HYAFF-11™. SEM analysis confirmed good adhesion of hDPSCs on both scaffolds before and after osteogenesis. Alizarin red S staining showed mineral deposits on both scaffolds after hDPSCs osteogenesis. The mRNA levels of runt-related transcription factor 2 (Runx2), collagen type I (Coll-I), osterix (Osx), osteocalcin (Ocn), osteopontin (Opn), bone sialoprotein (Bsp), and dentin sialophosphoprotein (Dspp) gene expression and their proteins were higher in hDPSCs after osteogenic differentiation on both scaffolds compared to undifferentiated hDPSCs on PLCL and HYAFF-11™. These results showed that PLCL scaffolds provide a better environment that supports hDPSCs attachment and osteogenic differentiation than HYAFF-11™. The high mRNA of early osteogenic gene expression and mineral deposits observed after hDPSCs osteogenesis on a PLCL mat indicated its better impact on hDPSCs' osteogenic potential than that of HYAFF-11™, and hDPSC/PLCL constructs might be considered in the future as an innovative approach to bone defect repair.
Collapse
Affiliation(s)
- Julia K. Bar
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.L.-N.); (P.G.G.)
| | - Anna Lis-Nawara
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.L.-N.); (P.G.G.)
| | - Tomasz Kowalczyk
- Laboratory of Polymers and Biomaterials, Institute of Fundamental Technological Research (IPPT PAN), Polish Academy of Sciences, Adolfa Pawińskiego 5B St., 02-106 Warsaw, Poland;
| | - Piotr G. Grelewski
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.L.-N.); (P.G.G.)
| | - Sandra Stamnitz
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland;
| | - Hanna Gerber
- Department of Maxillofacial Surgery, Wroclaw Medical University, Borowska 213, 50-556Wroclaw, Poland;
| | - Aleksandra Klimczak
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland;
| |
Collapse
|
23
|
Chen H, Lin YM, Bupphathong S, Lim J, Huang JE, Huang W, Hsieh TAS, Lin CH. Synthesis of Silanized Bioactive Glass/Gelatin Methacrylate (GelMA/Si-BG) composite hydrogel for Bone Tissue Engineering Application. J Mech Behav Biomed Mater 2023; 147:106159. [PMID: 37797555 DOI: 10.1016/j.jmbbm.2023.106159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/07/2023]
Abstract
Bioactive glass (BG) has been widely employed in the field of bone tissue engineering owing to its osteoconductive properties. These properties increase the stiffness and bioactivity of polymeric hydrogels, making them ideal for the repair, replacement, and regeneration of damaged bones. In this study, we investigated the effects of incorporating silanized 45S5 bioactive glass (Si-BG) into gelatin methacrylate (GelMA) hydrogel (GelMA/Si-BG) for potential bone tissue engineering. Our findings revealed that crosslinking GelMA with Si-BG had a striking increase in bioactivity with and without osteogenic induction of human mesenchymal stem cells (hMSCs) when compared to GelMA/BG hydrogels. Meanwhile, both GelMA/Si-BG and GelMA/BG hydrogels were able to maintain the cell viability of hMSC for up to 14 days. Additionally, GelMA/Si-BG hydrogels were shown to have a significantly higher compressive modulus than GelMA/BG hydrogels. This study has demonstrated the introduction of silanized 45S5 BG into GelMA hydrogel bioactivity and mechanical properties of GelMA hydrogels, exemplifying the potential application of silanization of BG in bone tissue engineering.
Collapse
Affiliation(s)
- Hsuan Chen
- Department of Dentistry, School of Dentistry, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Yuan-Min Lin
- Department of Dentistry, School of Dentistry, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan; Department of Stomatology, Taipei Veterans Hospital, Taipei, 11221, Taiwan; Institute of Oral Tissue Engineering and Biomaterials, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Sasinan Bupphathong
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan; High-value Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei, 10608, Taiwan
| | - Joshua Lim
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Jing-En Huang
- Department of Dentistry, School of Dentistry, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Wei Huang
- Department of Orthodontics, Rutgers School of Dental Medicine, Newark, NJ, 07103, USA
| | - Tiffany Angela S Hsieh
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chih-Hsin Lin
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
24
|
Lekhavadhani S, Shanmugavadivu A, Selvamurugan N. Role and architectural significance of porous chitosan-based scaffolds in bone tissue engineering. Int J Biol Macromol 2023; 251:126238. [PMID: 37567529 DOI: 10.1016/j.ijbiomac.2023.126238] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/26/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023]
Abstract
In designing and fabricating scaffolds to fill the bone defects and stimulate new bone formation, the biomimetics of the construct is a crucial factor in invoking the bone microenvironment to promote osteogenic differentiation. Regarding structural traits, changes in porous characteristics of the scaffolds, such as pore size, pore morphology, and percentage porosity, may patronize or jeopardize their other physicochemical and biological properties. Chitosan (CS), a biodegradable naturally occurring polymer, has recently drawn considerable attention as a scaffolding material in tissue engineering and regenerative medicine. CS-based microporous scaffolds have been reported to aid osteogenesis under both in vitro and in vivo conditions by supporting cellular attachment and proliferation of osteoblast cells and the formation of mineralized bone matrix. This related notion may be found in numerous earlier research, even though the precise mechanism of action that encourages the development of new bone still needs to be understood completely. This article presents the potential correlations and the significance of the porous properties of the CS-based scaffolds to influence osteogenesis and angiogenesis during bone regeneration. This review also goes over resolving the mechanical limitations of CS by blending it with other polymers and ceramics.
Collapse
Affiliation(s)
- Sundaravadhanan Lekhavadhani
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
25
|
Vijayan V, Sreekumar S, Ahina KM, Lakra R, Kiran MS. Lanthanum Oxide Nanoparticles Reinforced Collagen ƙ-Carrageenan Hydroxyapatite Biocomposite as Angio-Osteogenic Biomaterial for In Vivo Osseointegration and Bone Repair. Adv Biol (Weinh) 2023; 7:e2300039. [PMID: 37080950 DOI: 10.1002/adbi.202300039] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/22/2023] [Indexed: 04/22/2023]
Abstract
A composite biomatrix fabricated with collagen, ƙ-carrageenan, hydroxyapatite reinforced with lanthanum oxide nanoparticles is explored as proangiogenic and osteogenic bone tissue repair biomaterial. The biomatrix shows increased physical and biological stability as observed from proteolytic degradation and thermal stability studies. The addition of lanthanum oxide nanoparticles facilitates good osseointegration coupled with simultaneous activation of proangiogenic properties to act as a bone mimicking material. The minimal level of reactive oxygen species and superior cytocompatibility help the as-synthesized biomatrix in achieving capillary migration into the bone micro environment. The composite biomatrix upregulates the expression of VEGF, VEGF-R2 genes in endothelial cells and osteopontin, osteocalcin in osteoblasts cells, respectively. The in vivo hard tissue repair experiment conducted in a rat model shows complete healing of the bone defect by eight weeks with the application of collagen-ƙ-carrageenan-hydroxyapatite-lanthanum oxide nanoparticle biomaterial when compared to the biomaterial made out of individual constituents alone. The biomaterial matrix gets biointegrated into the bone tissue and exerts its therapeutic value in bringing a faster osseo repair process. The study shows the feasibility of using rare-earth metal nanoparticles in combination with protein-polysaccharide biopolymers for bone regeneration.
Collapse
Affiliation(s)
- Vinu Vijayan
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
- University of Madras, Chennai, Tamil Nadu, 600005, India
| | - Sreelekshmi Sreekumar
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kannoth Madappurakkal Ahina
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
| | - Rachita Lakra
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
| | - Manikantan Syamala Kiran
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, 600020, India
- University of Madras, Chennai, Tamil Nadu, 600005, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
26
|
Urzì O, Gasparro R, Costanzo E, De Luca A, Giavaresi G, Fontana S, Alessandro R. Three-Dimensional Cell Cultures: The Bridge between In Vitro and In Vivo Models. Int J Mol Sci 2023; 24:12046. [PMID: 37569426 PMCID: PMC10419178 DOI: 10.3390/ijms241512046] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Although historically, the traditional bidimensional in vitro cell system has been widely used in research, providing much fundamental information regarding cellular functions and signaling pathways as well as nuclear activities, the simplicity of this system does not fully reflect the heterogeneity and complexity of the in vivo systems. From this arises the need to use animals for experimental research and in vivo testing. Nevertheless, animal use in experimentation presents various aspects of complexity, such as ethical issues, which led Russell and Burch in 1959 to formulate the 3R (Replacement, Reduction, and Refinement) principle, underlying the urgent need to introduce non-animal-based methods in research. Considering this, three-dimensional (3D) models emerged in the scientific community as a bridge between in vitro and in vivo models, allowing for the achievement of cell differentiation and complexity while avoiding the use of animals in experimental research. The purpose of this review is to provide a general overview of the most common methods to establish 3D cell culture and to discuss their promising applications. Three-dimensional cell cultures have been employed as models to study both organ physiology and diseases; moreover, they represent a valuable tool for studying many aspects of cancer. Finally, the possibility of using 3D models for drug screening and regenerative medicine paves the way for the development of new therapeutic opportunities for many diseases.
Collapse
Affiliation(s)
- Ornella Urzì
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Roberta Gasparro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Elisa Costanzo
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Angela De Luca
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche, 40136 Bologna, Italy; (A.D.L.); (G.G.)
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche, 40136 Bologna, Italy; (A.D.L.); (G.G.)
| | - Simona Fontana
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| | - Riccardo Alessandro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy; (O.U.); (R.G.); (E.C.); (R.A.)
| |
Collapse
|
27
|
Toropitsyn E, Ščigalková I, Pravda M, Toropitsyna J, Velebný V. Enzymatically cross-linked hyaluronic acid hydrogels as in situ forming carriers of platelet-rich plasma: Mechanical properties and bioactivity levels evaluation. J Mech Behav Biomed Mater 2023; 143:105916. [PMID: 37224645 DOI: 10.1016/j.jmbbm.2023.105916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
New studies have shown the great potential of the combination of in situ enzymatically cross-linked hydrogels based on tyramine derivative of hyaluronic acid (HA-TA) with platelet-rich plasma (PRP) and platelet lysate in regenerative medicine. This study describes how the presence of PRP and platelet lysate affects the kinetics of gelation, viscoelastic properties, swelling ratio, and the network structure of HA-TA hydrogels and how the encapsulation of PRP in hydrogels affects the bioactivity of released PRP determined as the ability to induce cell proliferation. The properties of hydrogels were tuned by a degree of substitution and concentration of HA-TA derivatives. The addition of platelet derivatives to the reaction mixture slowed down the cross-linking reaction and reduced elastic modulus (G') and thus cross-linking efficiency. However, low-swellable hydrogels (7-190%) suitable for soft tissue engineering with G' 200-1800 Pa were prepared with a gelation time within 1 min. It was confirmed that tested cross-linking reaction conditions are suitable for PRP incorporation because the total bioactivity level of PRP released from HA-TA hydrogels was ≥87% and HA-TA content in the hydrogels and thus mesh size (285-482 nm) has no significant effect on the bioactivity level of released PRP.
Collapse
Affiliation(s)
- Evgeniy Toropitsyn
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic; Biocev, First Faculty of Medicine Charles University, Průmyslová 595, 25250, Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, Prague, 120 00, Czech Republic.
| | - Ivana Ščigalková
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic
| | - Martin Pravda
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic
| | - Jelena Toropitsyna
- Department of Power Engineering, University of Chemistry and Technology, Prague, Technická 5, Prague, 166 28, Czech Republic
| | - Vladimír Velebný
- Contipro a.s., Dolní Dobrouč 401, 56102, Dolní Dobrouč, Czech Republic
| |
Collapse
|
28
|
Agnes CJ, Karoichan A, Tabrizian M. The Diamond Concept Enigma: Recent Trends of Its Implementation in Cross-linked Chitosan-Based Scaffolds for Bone Tissue Engineering. ACS APPLIED BIO MATERIALS 2023. [PMID: 37310896 PMCID: PMC10354806 DOI: 10.1021/acsabm.3c00108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
An increasing number of publications over the past ten years have focused on the development of chitosan-based cross-linked scaffolds to regenerate bone tissue. The design of biomaterials for bone tissue engineering applications relies heavily on the ideals set forth by a polytherapy approach called the "Diamond Concept". This methodology takes into consideration the mechanical environment, scaffold properties, osteogenic and angiogenic potential of cells, and benefits of osteoinductive mediator encapsulation. The following review presents a comprehensive summarization of recent trends in chitosan-based cross-linked scaffold development within the scope of the Diamond Concept, particularly for nonload-bearing bone repair. A standardized methodology for material characterization, along with assessment of in vitro and in vivo potential for bone regeneration, is presented based on approaches in the literature, and future directions of the field are discussed.
Collapse
Affiliation(s)
- Celine J Agnes
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
- Shriner's Hospital for Children, Montreal, Quebec H4A 0A9 Canada
| | - Antoine Karoichan
- Shriner's Hospital for Children, Montreal, Quebec H4A 0A9 Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec H3A 1G1 Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec H3A 1G1 Canada
| |
Collapse
|
29
|
Re F, Borsani E, Rezzani R, Sartore L, Russo D. Bone Regeneration Using Mesenchymal Stromal Cells and Biocompatible Scaffolds: A Concise Review of the Current Clinical Trials. Gels 2023; 9:gels9050389. [PMID: 37232981 DOI: 10.3390/gels9050389] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Bone regenerative medicine is a clinical approach combining live osteoblast progenitors, such as mesenchymal stromal cells (MSCs), with a biocompatible scaffold that can integrate into host bone tissue and restore its structural integrity. Over the last few years, many tissue engineering strategies have been developed and thoroughly investigated; however, limited approaches have been translated to clinical application. Consequently, the development and clinical validation of regenerative approaches remain a centerpiece of investigational efforts towards the clinical translation of advanced bioengineered scaffolds. The aim of this review was to identify the latest clinical trials related to the use of scaffolds with or without MSCs to regenerate bone defects. A revision of the literature was performed in PubMed, Embase, and Clinicaltrials.gov from 2018 up to 2023. Nine clinical trials were analyzed according to the inclusion criteria: six presented in the literature and three reported in Clinicaltrials.gov. Data were extracted covering background trial information. Six of the clinical trials added cells to scaffolds, while three used scaffolds alone. The majority of scaffolds were composed of calcium phosphate ceramic alone, such as β-tricalcium phosphate (TCP) (two clinical trials), biphasic calcium phosphate bioceramic granules (three clinical trials), and anorganic bovine bone (two clinical trials), while bone marrow was the primary source of the MSCs (five clinical trials). The MSC expansion was performed in GMP facilities, using human platelet lysate (PL) as a supplement without osteogenic factors. Only one trial reported minor adverse events. Overall, these findings highlight the importance and efficacy of cell-scaffold constructs in regenerative medicine under different conditions. Despite the encouraging clinical results obtained, further studies are needed to assess their clinical efficacy in treating bone diseases to optimize their application.
Collapse
Affiliation(s)
- Federica Re
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
| | - Elisa Borsani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs (ARTO)", University of Brescia, 25123 Brescia, Italy
| | - Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs (ARTO)", University of Brescia, 25123 Brescia, Italy
| | - Luciana Sartore
- Department of Mechanical and Industrial Engineering, Materials Science and Technology Laboratory, University of Brescia, 25123 Brescia, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
| |
Collapse
|
30
|
Li D, Jiang Y, He P, Li Y, Wu Y, Lei W, Liu N, de Bruijn JD, Zhang H, Zhang H, Ji P, Yuan H, Li M. Hypoxia Drives Material-Induced Heterotopic Bone Formation by Enhancing Osteoclastogenesis via M2/Lipid-Loaded Macrophage Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207224. [PMID: 36970815 PMCID: PMC10214238 DOI: 10.1002/advs.202207224] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/19/2023] [Indexed: 05/27/2023]
Abstract
Heterotopic ossification (HO) is a double-edged sword. Pathological HO presents as an undesired clinical complication, whereas controlled heterotopic bone formation by synthetic osteoinductive materials shows promising therapeutic potentials for bone regeneration. However, the mechanism of material-induced heterotopic bone formation remains largely unknown. Early acquired HO being usually accompanied by severe tissue hypoxia prompts the hypothesis that hypoxia caused by the implantation coordinates serial cellular events and ultimately induces heterotopic bone formation in osteoinductive materials. The data presented herein shows a link between hypoxia, macrophage polarization to M2, osteoclastogenesis, and material-induced bone formation. Hypoxia inducible factor-1α (HIF-1α), a crucial mediator of cellular responses to hypoxia, is highly expressed in an osteoinductive calcium phosphate ceramic (CaP) during the early phase of implantation, while pharmacological inhibition of HIF-1α significantly inhibits M2 macrophage, subsequent osteoclast, and material-induced bone formation. Similarly, in vitro, hypoxia enhances M2 macrophage and osteoclast formation. Osteoclast-conditioned medium enhances osteogenic differentiation of mesenchymal stem cells, such enhancement disappears with the presence of HIF-1α inhibitor. Furthermore, metabolomics analysis reveals that hypoxia enhances osteoclastogenesis via the axis of M2/lipid-loaded macrophages. The current findings shed new light on the mechanism of HO and favor the design of more potent osteoinductive materials for bone regeneration.
Collapse
Affiliation(s)
- Dan Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Yucan Jiang
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Ping He
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Yeming Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Yan Wu
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Wei Lei
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Nanxin Liu
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Joost D. de Bruijn
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
- Kuros Biosciences BVProf. Bronkhorstlaan 10Bilthoven3723 MBThe Netherlands
| | - Hua Zhang
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400015P. R. China
| | - Hongmei Zhang
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Ping Ji
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| | - Huipin Yuan
- Kuros Biosciences BVProf. Bronkhorstlaan 10Bilthoven3723 MBThe Netherlands
- Huipin Yuan's LabChengdu610000P. R. China
| | - Mingzheng Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationStomatological Hospital of Chongqing Medical UniversityChongqing401120P. R. China
| |
Collapse
|
31
|
Zhang L, Wang M, Qiu H, Wei Y, Zhou L, Nian N, Shi Z, Hu D, Ma B. Epicatechin gallate promotes vascularization in co-culture of human osteoblasts and outgrowth endothelial cells. Exp Biol Med (Maywood) 2023; 248:732-745. [PMID: 37354086 PMCID: PMC10408553 DOI: 10.1177/15353702231171894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/27/2023] [Indexed: 06/26/2023] Open
Abstract
Prevascularization is crucial for the survival of tissue-engineered bone and further bone repair/regeneration. Since epicatechin gallate (ECG), the most abundant flavanol in green tea, shows potential beneficial effects on endothelial cells and bone cells, we decided to investigate whether it promotes vascularization/angiogenesis and osteogenesis using a co-culture system containing human primary osteoblasts (POBs) and outgrowth endothelial cells (OECs). We found that treatment with ECG (1) significantly enhanced microvessel formation in co-culture of POB and OECs, (2) improved cell viability/proliferation and the angiogenic/osteogenic capacities of OEC/POBs, (3) significantly increased the levels of E-selectin, IL-6, TNF-α, IFN-γ, VEGF, and PDGF-BB in co-cultures of POB and OEC, and (4) upregulated HIF-1α, HIF-2α, NF-κB, iNOS, GLUT1, VEGF, and Ang1/2 but downregulated PHD1 in monocultures of OEC or POB. Our findings demonstrate that ECG promotes angiogenesis and osteogenesis (probably via HIF signaling) in co-cultures of OECs and POBs. ECG thus has potential applications in the promotion of angiogenesis/vascularization in many tissue constructs including those of bone.
Collapse
Affiliation(s)
- Liyan Zhang
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Miaoran Wang
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Huiqing Qiu
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
- Department of Geriatrics, The First Hospital of Hebei Medical University, Shijiazhuang 050030, China
| | - Yusen Wei
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Lu Zhou
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Nannan Nian
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Zhongli Shi
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, The First Hospital of Hebei Medical University, Shijiazhuang 050030, China
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang 050030, China
| | - Dailun Hu
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, China
| | - Bin Ma
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA 6150, Australia
| |
Collapse
|
32
|
Huang X, Li Y, Liao H, Luo X, Zhao Y, Huang Y, Zhou Z, Xiang Q. Research Advances on Stem Cell-Derived Extracellular Vesicles Promoting the Reconstruction of Alveolar Bone through RANKL/RANK/OPG Pathway. J Funct Biomater 2023; 14:jfb14040193. [PMID: 37103283 PMCID: PMC10145790 DOI: 10.3390/jfb14040193] [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: 02/02/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023] Open
Abstract
Periodontal bone tissue defects and bone shortages are the most familiar and troublesome clinical problems in the oral cavity. Stem cell-derived extracellular vesicles (SC-EVs) have biological properties similar to their sources, and they could be a promising acellular therapy to assist with periodontal osteogenesis. In the course of alveolar bone remodeling, the RANKL/RANK/OPG signaling pathway is an important pathway involved in bone metabolism. This article summarizes the experimental studies of SC-EVs applied for the therapy of periodontal osteogenesis recently and explores the role of the RANKL/RANK/OPG pathway in their mechanism of action. Their unique patterns will open a new field of vision for people, and they will help to advance a possible future clinical treatment.
Collapse
Affiliation(s)
- Xia Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
- Department of Orthodontics, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Yuxiao Li
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Hui Liao
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Xin Luo
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Yueping Zhao
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Yadong Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Zhiying Zhou
- School of Stomatology, Jinan University, Guangzhou 510632, China
- Department of Orthodontics, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Qi Xiang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
33
|
Maciel GBM, Maciel RM, Danesi CC. Bone cells and their role in physiological remodeling. Mol Biol Rep 2023; 50:2857-2863. [PMID: 36609750 DOI: 10.1007/s11033-022-08190-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 12/07/2022] [Indexed: 01/09/2023]
Abstract
PURPOSE This work compiles the characteristics of bone cells involved in the physiological bone remodeling. METHODS A narrative review of the literature was performed. RESULTS Remodeling is a different process from modeling. Remodeling allows old or damaged bone tissue to be renewed, ensuring the maintenance of bone fracture resistance, as well as maintaining calcium and phosphorus homeostasis. We present the role of osteoclasts, a multinucleated cell with hematopoietic origin responsible for resorbing bone. The formation of osteoclasts depends on the cytokines macrophage colony stimulating factor (M-CSF) and receptor activator of NF-kB ligand (RANKL) and can be blocked by osteoprotegerin. Furthermore, this review highlights the features of osteoblasts, polarized cubic cells of mesenchymal origin that deposit bone and also covers osteocytes and bone lining cells. This review presents the five fundamental phases of bone remodeling and addresses aspects of its regulation through hormones and growth factors. CONCLUSIONS Knowledge of the current concepts of physiological bone remodeling is necessary for the study of the different pathologies that affect the bone tissue and thus helps in the search for new therapies.
Collapse
Affiliation(s)
- Gabriel Bassan Marinho Maciel
- Postgraduate Program in Dental Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil. .,Department of Pathology, Federal University of Santa Maria, Av. Roraima, 1000, Santa Maria, 97015-900, RS, Brazil.
| | | | | |
Collapse
|
34
|
Habibzadeh F, Sadraei SM, Mansoori R, Singh Chauhan NP, Sargazi G. Nanomaterials supported by polymers for tissue engineering applications: A review. Heliyon 2022; 8:e12193. [PMID: 36578390 PMCID: PMC9791886 DOI: 10.1016/j.heliyon.2022.e12193] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/21/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
In the biomedical sciences, particularly in wound healing, tissue engineering, and regenerative medicine, the development of natural-based biomaterials as a carrier has revealed a wide range of advantages. Tissue engineering is one of the therapeutic approaches used to replace damaged tissue. Polymers have received a lot of attention for their beneficial interactions with cells, but they have some drawbacks, such as poor mechanical properties. Due to their relatively large surface area, nanoparticles can cause significant changes in polymers and improve their mechanical properties. The nanoparticles incorporated into biomaterial scaffolds have been associated with positive effects on cell adhesion, viability, proliferation, and migration in the majority of studies. This review paper discusses recent applications of polymer-nanoparticle composites in the development of tissue engineering scaffolds, as well as the effects of these nanomaterials in the fields of cardiovascular, neural, bone, and skin tissue engineering.
Collapse
Affiliation(s)
- Faezeh Habibzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahdi Sadraei
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Roghayeh Mansoori
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Narendra Pal Singh Chauhan
- Department of Chemistry, Faculty of Science, Bhupal Nobles' University, Udaipur, Rajasthan, India,Corresponding author.
| | - Ghasem Sargazi
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran,Corresponding author.
| |
Collapse
|
35
|
Klimek K, Palka K, Truszkiewicz W, Douglas TEL, Nurzynska A, Ginalska G. Could Curdlan/Whey Protein Isolate/Hydroxyapatite Biomaterials Be Considered as Promising Bone Scaffolds?-Fabrication, Characterization, and Evaluation of Cytocompatibility towards Osteoblast Cells In Vitro. Cells 2022; 11:cells11203251. [PMID: 36291119 PMCID: PMC9600130 DOI: 10.3390/cells11203251] [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: 09/12/2022] [Revised: 10/08/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
The number of bone fractures and cracks requiring surgical interventions increases every year; hence, there is a huge need to develop new potential bone scaffolds for bone regeneration. The goal of this study was to gain knowledge about the basic properties of novel curdlan/whey protein isolate/hydroxyapatite biomaterials in the context of their use in bone tissue engineering. The purpose of this research was also to determine whether the concentration of whey protein isolate in scaffolds has an influence on their properties. Thus, two biomaterials differing in the concentration of whey protein isolate (i.e., 25 wt.% and 35 wt.%; hereafter called Cur_WPI25_HAp and Cur_WPI35_HAp, respectively) were fabricated and subjected to evaluation of porosity, mechanical properties, swelling ability, protein release capacity, enzymatic biodegradability, bioactivity, and cytocompatibility towards osteoblasts in vitro. It was found that both biomaterials fulfilled a number of requirements for bone scaffolds, as they demonstrated limited swelling and the ability to undergo controllable enzymatic biodegradation, to form apatite layers on their surfaces and to support the viability, growth, proliferation, and differentiation of osteoblasts. On the other hand, the biomaterials were characterized by low open porosity, which may hinder the penetration of cells though their structure. Moreover, they had low mechanical properties compared to natural bone, which limits their use to filling of bone defects in non-load bearing implantation areas, e.g., in the craniofacial area, but then they will be additionally supported by application of mechanically strong materials such as titanium plates. Thus, this preliminary in vitro research indicates that biomaterials composed of curdlan, whey protein isolate, and hydroxyapatite seem promising for bone tissue engineering applications, but their porosity and mechanical properties should be improved. This will be the subject of our further work.
Collapse
Affiliation(s)
- Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
- Correspondence: ; Tel.: +48-448-70-28
| | - Krzysztof Palka
- Faculty of Mechanical Engineering, Lublin University of Technology, Nadbystrzycka 26 Street, 20-618 Lublin, Poland
| | - Wieslaw Truszkiewicz
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
| | - Timothy E. L. Douglas
- School of Engineering, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK
- Materials Science Institute (MSI), Lancaster University, Lancaster LA1 4YW, UK
| | - Aleksandra Nurzynska
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
| | - Grazyna Ginalska
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
| |
Collapse
|
36
|
Calcium Phosphate-Based Biomaterials for Bone Repair. J Funct Biomater 2022; 13:jfb13040187. [PMID: 36278657 PMCID: PMC9589993 DOI: 10.3390/jfb13040187] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
Traumatic, tumoral, and infectious bone defects are common in clinics, and create a big burden on patient's families and society. Calcium phosphate (CaP)-based biomaterials have superior properties and have been widely used for bone defect repair, due to their similarities to the inorganic components of human bones. The biological performance of CaPs, as a determining factor for their applications, are dependent on their physicochemical properties. Hydroxyapatite (HAP) as the most thermally stable crystalline phase of CaP is mostly used in the form of ceramics or composites scaffolds with polymers. Nanostructured CaPs with large surface areas are suitable for drug/gene delivery systems. Additionally, CaP scaffolds with hierarchical nano-/microstructures have demonstrated excellent ability in promoting bone regeneration. This review focuses on the relationships and interactions between the physicochemical/biological properties of CaP biomaterials and their species, sizes, and morphologies in bone regeneration, including synthesis strategies, structure control, biological behavior, and the mechanisms of CaP in promoting osteogenesis. This review will be helpful for scientists and engineers to further understand CaP-based biomaterials (CaPs), and be useful in developing new high-performance biomaterials for bone repair.
Collapse
|
37
|
Al Maruf DSA, Ghosh YA, Xin H, Cheng K, Mukherjee P, Crook JM, Wallace GG, Klein TJ, Clark JR. Hydrogel: A Potential Material for Bone Tissue Engineering Repairing the Segmental Mandibular Defect. Polymers (Basel) 2022; 14:polym14194186. [PMID: 36236133 PMCID: PMC9571534 DOI: 10.3390/polym14194186] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Free flap surgery is currently the only successful method used by surgeons to reconstruct critical-sized defects of the jaw, and is commonly used in patients who have had bony lesions excised due to oral cancer, trauma, infection or necrosis. However, donor site morbidity remains a significant flaw of this strategy. Various biomaterials have been under investigation in search of a suitable alternative for segmental mandibular defect reconstruction. Hydrogels are group of biomaterials that have shown their potential in various tissue engineering applications, including bone regeneration, both through in vitro and in vivo pre-clinical animal trials. This review discusses different types of hydrogels, their fabrication techniques, 3D printing, their potential for bone regeneration, outcomes, and the limitations of various hydrogels in preclinical models for bone tissue engineering. This review also proposes a modified technique utilizing the potential of hydrogels combined with scaffolds and cells for efficient reconstruction of mandibular segmental defects.
Collapse
Affiliation(s)
- D S Abdullah Al Maruf
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown 2050, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
- Correspondence:
| | - Yohaann Ali Ghosh
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown 2050, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
| | - Hai Xin
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown 2050, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
| | - Kai Cheng
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local, Camperdown 2050, Australia
| | - Payal Mukherjee
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown 2050, Australia
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local, Camperdown 2050, Australia
| | - Jeremy Micah Crook
- Biomedical Innovation, Chris O’Brien Lifehouse, Camperdown 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
- Sarcoma and Surgical Research Centre, Chris O’Brien Lifehouse, Camperdown 2050, Australia
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong 2522, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong 2522, Australia
- Illawarra Health and Medical Research Institute, The University of Wollongong, Wollongong 2522, Australia
| | - Gordon George Wallace
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong 2522, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong 2522, Australia
| | - Travis Jacob Klein
- Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove 4059, Australia
| | - Jonathan Robert Clark
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown 2050, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local, Camperdown 2050, Australia
| |
Collapse
|
38
|
Treatment Effect of Platelet Gel on Reconstructing Bone Defects and Nonunions: A Review of In Vivo Human Studies. Int J Mol Sci 2022; 23:ijms231911377. [PMID: 36232679 PMCID: PMC9570043 DOI: 10.3390/ijms231911377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022] Open
Abstract
In ideal circumstances, a fractured bone can heal properly by itself or with the aid of clinical interventions. However, around 5% to 10% of bone fractures fail to heal properly within the expected time even with the aid of clinical interventions, resulting in nonunions. Platelet gel is a blood-derived biomaterial used in regenerative medicine aiming to promote wound healing and regeneration of damaged tissues. The purpose of this paper is to review relevant articles in an attempt to explore the current consensus on the treatment effect of platelet gel on reconstructing bone defects and nonunions, hoping to provide a valuable reference for clinicians to make treatment decisions in clinical practice. Based on the present review, most of the studies applied the combination of platelet gel and bone graft to reconstruct bone defects and nonunions, and most of the results were positive, suggesting that this treatment strategy could promote successful reconstruction of bone defects and nonunions. Only two studies tried to apply platelet gel alone to reconstruct bone defects and nonunions, therefore a convincing conclusion could not be made yet regarding the treatment effect of platelet gel alone on reconstructing bone defects and nonunions. Only one study applied platelet gel combined with extracorporeal shock wave therapy to reconstruct nonunions, and the results were positive; the hypothetical mechanism of this treatment strategy is reasonable and sound, and more future clinical studies are encouraged to further justify the effectiveness of this promising treatment strategy. In conclusion, the application of platelet gel could be a promising and useful treatment method for reconstructing bone defects and nonunions, and more future clinical studies are encouraged to further investigate the effectiveness of this promising treatment method.
Collapse
|
39
|
Khan MUA, Razak SIA, Rehman S, Hasan A, Qureshi S, Stojanović GM. Bioactive scaffold (sodium alginate)-g-(nHAp@SiO 2@GO) for bone tissue engineering. Int J Biol Macromol 2022; 222:462-472. [PMID: 36155784 DOI: 10.1016/j.ijbiomac.2022.09.153] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/02/2022] [Accepted: 09/16/2022] [Indexed: 12/24/2022]
Abstract
Globally, people suffering from bone disorders are steadily increasing and bone tissue engineering is an advanced approach to treating fractured and defected bone tissues. In this study, we have prepared polymeric nanocomposite by free-radical polymerization from sodium alginate, hydroxyapatite, and silica with different GO amounts. The porous scaffolds were fabricated using the freeze drying technique. The structural, morphological, mechanical, and wetting investigation was conducted by Fourier-transform infrared spectroscopy, X-ray diffraction, scanning electron microscope, universal tensile machine, and water contact angle characterization techniques. The swelling, biodegradation, and water retention were also studied. The biological studies were performed (cell viability, cell adherence, proliferation, and mineralization) against osteoblast cell lines. Scaffolds have exhibited different pore morphology SAG-1 (pore size = 414.61 ± 56 μm and porosity = 81.45 ± 2.17 %) and SAG-4 (pore size = 195.97 ± 82 μm and porosity = 53.82 ± 2.45 %). They have different mechanical behavior as SAG-1 has the least compression strength and compression modulus 2.14 ± 2.35 and 16.51 ± 1.27 MPa. However, SAG-4 has maximum compression strength and compression modulus 13.67 ± 2.63 and 96.16 ± 1.97 MPa with wetting behavior 80.70° and 58.70°, respectively. Similarly, SAG-1 exhibited the least and SAG-4 presented maximum apatite mineral formation, cell adherence, cell viability, and cell proliferation against mouse pre-osteoblast cell lines. The increased GO amount provides different multifunctional materials with different characteristics. Hence, the fabricated scaffolds could be potential scaffold materials to treat and regenerate fracture bone tissues in bone tissue engineering.
Collapse
Affiliation(s)
- Muhammad Umar Aslam Khan
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar.
| | - Saiful Izwan Abd Razak
- BioInspired Device and Tissue Engineering Research Group, School of Biomedical Engineering and Health Sciences, Faculty of Engineering, Universiti Teknologi Malaysia, 81300 Skudai, Johor, Malaysia; Sports Innovation & Technology Centre, Institute of Human Centred Engineering, Universiti Teknologi Malaysia, 81300 Skudai, Johor, Malaysia
| | - Sarish Rehman
- Department of Chemistry, McGill University, Montreal H3A 0B8, Canada
| | - Anwarul Hasan
- Biomedical Research Center, Qatar University, Doha 2713, Qatar; Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar
| | - Saima Qureshi
- Faculty of Technical Sciences, University of Novi Sad, T. Dositeja Obradovića 6, 21000 Novi Sad, Serbia
| | - Goran M Stojanović
- Faculty of Technical Sciences, University of Novi Sad, T. Dositeja Obradovića 6, 21000 Novi Sad, Serbia
| |
Collapse
|
40
|
Saunders WB, Dejardin LM, Soltys-Niemann EV, Kaulfus CN, Eichelberger BM, Dobson LK, Weeks BR, Kerwin SC, Gregory CA. Angle-stable interlocking nailing in a canine critical-sized femoral defect model for bone regeneration studies: In pursuit of the principle of the 3R’s. Front Bioeng Biotechnol 2022; 10:921486. [PMID: 36118571 PMCID: PMC9479202 DOI: 10.3389/fbioe.2022.921486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction: Critical-sized long bone defects represent a major therapeutic challenge and current treatment strategies are not without complication. Tissue engineering holds much promise for these debilitating injuries; however, these strategies often fail to successfully translate from rodent studies to the clinical setting. The dog represents a strong model for translational orthopedic studies, however such studies should be optimized in pursuit of the Principle of the 3R’s of animal research (replace, reduce, refine). The objective of this study was to refine a canine critical-sized femoral defect model using an angle-stable interlocking nail (AS-ILN) and reduce total animal numbers by performing imaging, biomechanics, and histology on the same cohort of dogs. Methods: Six skeletally mature hounds underwent a 4 cm mid-diaphyseal femoral ostectomy followed by stabilization with an AS-ILN. Dogs were assigned to autograft (n = 3) or negative control (n = 3) treatment groups. At 6, 12, and 18 weeks, healing was quantified by ordinal radiographic scoring and quantified CT. After euthanasia, femurs from the autograft group were mechanically evaluated using an established torsional loading protocol. Femurs were subsequently assessed histologically. Results: Surgery was performed without complication and the AS-ILN provided appropriate fixation for the duration of the study. Dogs assigned to the autograft group achieved radiographic union by 12 weeks, whereas the negative control group experienced non-union. At 18 weeks, median bone and soft tissue callus volume were 9,001 mm3 (range: 4,939–10,061) for the autograft group and 3,469 mm3 (range: 3,085–3,854) for the negative control group. Median torsional stiffness for the operated, autograft treatment group was 0.19 Nm/° (range: 0.19–1.67) and torque at failure was 12.0 Nm (range: 1.7–14.0). Histologically, callus formation and associated endochondral ossification were identified in the autograft treatment group, whereas fibrovascular tissue occupied the critical-sized defect in negative controls. Conclusion: In a canine critical-sized defect model, the AS-ILN and described outcome measures allowed refinement and reduction consistent with the Principle of the 3R’s of ethical animal research. This model is well-suited for future canine translational bone tissue engineering studies.
Collapse
Affiliation(s)
- W. B. Saunders
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A & M University, College Station, TX, United States
- *Correspondence: W. B. Saunders,
| | - L. M. Dejardin
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - E. V. Soltys-Niemann
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A & M University, College Station, TX, United States
| | - C. N. Kaulfus
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A & M University, College Station, TX, United States
| | - B. M. Eichelberger
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A & M University, College Station, TX, United States
| | - L. K. Dobson
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A & M University, College Station, TX, United States
| | - B. R. Weeks
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A & M University, College Station, TX, United States
| | - S. C. Kerwin
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A & M University, College Station, TX, United States
| | - C. A. Gregory
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, School of Medicine, Texas A & M Health Science Center, College Station, TX, United States
| |
Collapse
|
41
|
Hayashi T, Asakura M, Kawase M, Matsubara M, Uematsu Y, Mieki A, Kawai T. Bone Tissue Engineering in Rat Calvarial Defects Using Induced Bone-like Tissue by rhBMPs from Immature Muscular Tissues In Vitro. Int J Mol Sci 2022; 23:ijms23136927. [PMID: 35805943 PMCID: PMC9266849 DOI: 10.3390/ijms23136927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
This study aimed to induce bone-like tissue from immature muscular tissue (IMT) in vitro using commercially available recombinant human bone morphogenetic protein (rhBMP)-2, rhBMP-4, and rhBMP-7, and then implanting this tissue into a calvarial defect in rats to assess healing. IMTs were extracted from 20-day-old Sprague-Dawley (SD) fetal rats, placed on expanded polytetrafluoroethylene (ePTFE) with 10 ng/μL each of rhBMP-2, BMP-4, and BMP-7, and cultured for two weeks. The specimens were implanted into calvarial defects in 3-week-old SD rats for up to three weeks. Relatively strong radiopacity was observed on micro-CT two weeks after culture, and bone-like tissue, comprising osteoblastic cells and osteoids, was partially observed by H&E staining. Calcium, phosphorus, and oxygen were detected in the extracellular matrix using an electron probe micro analyzer, and X-ray diffraction patterns and Fourier transform infrared spectroscopy spectra of the specimen were found to have typical apatite crystal peaks and spectra, respectively. Furthermore, partial strong radiopacity and ossification were confirmed one week after implantation, and a dominant novel bone was observed after two weeks in the defect site. Thus, rhBMP-2, BMP-4, and BMP-7 differentiated IMT into bone-like tissue in vitro, and this induced bone-like tissue has ossification potential and promotes the healing of calvarial defects. Our results suggest that IMT is an effective tissue source for bone tissue engineering.
Collapse
|
42
|
Noroozi R, Shamekhi MA, Mahmoudi R, Zolfagharian A, Asgari F, Mousavizadeh A, Bodaghi M, Hadi A, Haghighipour N. In vitro static and dynamic cell culture study of novel bone scaffolds based on 3D-printed PLA and cell-laden alginate hydrogel. Biomed Mater 2022; 17. [PMID: 35609602 DOI: 10.1088/1748-605x/ac7308] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/24/2022] [Indexed: 11/11/2022]
Abstract
The aim of this paper was to design and fabricate a novel composite scaffold based on the combination of 3D-printed PLA-based triply minimal surface structures (TPMS) and cell-laden alginate hydrogel. This novel scaffold improves the low mechanical properties of alginate hydrogel and can also provide a scaffold with a suitable pore size, which can be used in bone regeneration applications. In this regard, an implicit function was used to generate some Gyroid TPMS scaffolds. Then the fused deposition modeling (FDM) process was employed to print the scaffolds. Moreover, the micro-CT technique was employed to assess the microstructure of 3D-printed TPMS scaffolds and obtain the real geometries of printed scaffolds. The mechanical properties of composite scaffolds were investigated under compression tests experimentally. It was shown that different mechanical behaviors could be obtained for different implicit function parameters. In this research, to assess the mechanical behavior of printed scaffolds in terms of the strain-stress curves on, two approaches were presented: equivalent volume and finite element-based volume. Results of strain-stress curves showed that the finite-element based approach predicts a higher level of stress. Moreover, the biological response of composite scaffolds in terms of cell viability, cell proliferation, and cell attachment was investigated. In this vein, a dynamic cell culture system was designed and fabricated, which improves mass transport through the composite scaffolds and applies mechanical loading to the cells, which helps cell proliferation. Moreover, the results of the novel composite scaffolds were compared to those without Alginate, and it was shown that the composite scaffold could create more viability and cell proliferation in both dynamic and static cultures. Also, it was shown that scaffolds in dynamic cell culture have a better biological response than in static culture. In addition, Scanning electron microscopy was employed to study the cell adhesion on the composite scaffolds, which showed excellent attachment between the scaffolds and cells.
Collapse
Affiliation(s)
- Reza Noroozi
- Pasteur Institute of Iran, tehran, Tehran, 1316943551, Iran (the Islamic Republic of)
| | - Mohammad Amin Shamekhi
- Department of Polymer Engineering, Sarvestan Branch, Islamic Azad University, Sarvestan, Shiraz, Shiraz, 19585-466, Iran (the Islamic Republic of)
| | - Reza Mahmoudi
- Yasuj University of Medical Sciences, yasuj, Yasuj, 000, Iran (the Islamic Republic of)
| | - Ali Zolfagharian
- Engineering, Deakin University Faculty of Science Engineering and Built Environment, Waurn Ponds, Geelong, Victoria, 3217, AUSTRALIA
| | - Fatemeh Asgari
- Pasteur Institute of Iran, tehran, Tehran, 1316943551, Iran (the Islamic Republic of)
| | - Ali Mousavizadeh
- Yasuj University of Medical Sciences, yasuj, Yasuj, 00000, Iran (the Islamic Republic of)
| | - Mahdi Bodaghi
- Engineering , Nottingham Trent University - Clifton Campus, Nottingham, Nottingham, NG11 8NS, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Amin Hadi
- Cellular and Molecular Research Center , Yasuj University of Medical Sciences, Yasuj, Yasuj, 00000, Iran (the Islamic Republic of)
| | - Nooshin Haghighipour
- Pasteur Institute of Iran, Tehran, Tehran, Tehran, 1316943551, Iran (the Islamic Republic of)
| |
Collapse
|
43
|
Shen HX, Liu JZ, Yan XQ, Yang HN, Hu SQ, Yan XL, Xu T, El Haj AJ, Yang Y, Lü LX. Hydrostatic pressure stimulates the osteogenesis and angiogenesis of MSCs/HUVECs co-culture on porous PLGA scaffolds. Colloids Surf B Biointerfaces 2022; 213:112419. [PMID: 35227994 DOI: 10.1016/j.colsurfb.2022.112419] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 01/04/2023]
Abstract
In native bone tissue regeneration, blood vessels, providing oxygen and nutrition for tissues, can promote the regeneration of bone and accelerate the repair of a defected area. In this study, Poly(D, L-lactic-co-glycolic acid) (PLGA) inverse opal scaffolds with high pore interconnectivity were fabricated and further modified with vascular endothelial growth factor (VEGF). The rat bone marrow derived mesenchymal stem cells (rMSCs) and human umbilical vein endothelial cells (HUVECs) were co-cultured onto the scaffolds to enhance vascularization for bone tissue regeneration. Cell attachment, viability, proliferation, and morphology were detected by cell counting kit-8 (CCK-8) assay, live and dead staining and scanning electron microscopy (SEM). Hydrostatic pressure with 0-279 KPa and 1 Hz one hour per day for 7 days was applied to tissue engineered bone constructs to investigate whether the loading stimulation can promote osteogenesis and angiogenesis mutually evaluated in parallel by multiple in vitro assays and in an in vivo chicken chorioallantoic membrane (CAM) model. The results indicated that the immobilization of VEGF can improve biocompatibility of PLGA scaffolds and promote cell attachment and proliferation. The cell-scaffold constructs showed higher CD31 expression because of the angiogenic differentiation of rMSCs in hydrostatic loading culture condition in vitro. The in vivo CAM model experiment demonstrated that hydrostatic loading stimulated angiogenic differentiation of rMSCs can accelerate tubulogenesis. Furthermore, the new capillaries formed in cell-scaffold constructs were conducive to calcium deposition in vivo.
Collapse
Affiliation(s)
- Hong-Xian Shen
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Jing-Zhi Liu
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Xiao-Qing Yan
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Hong-Ning Yang
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Shu-Qun Hu
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Xian-Liang Yan
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China; Emergency Medicine Department of the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Tie Xu
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China; Emergency Medicine Department of the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Alicia J El Haj
- Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TH, UK
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK.
| | - Lan-Xin Lü
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China; Emergency Medicine Department of the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China.
| |
Collapse
|
44
|
Rajendran AK, Amirthalingam S, Hwang NS. A brief review of mRNA therapeutics and delivery for bone tissue engineering. RSC Adv 2022; 12:8889-8900. [PMID: 35424872 PMCID: PMC8985089 DOI: 10.1039/d2ra00713d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/15/2022] [Indexed: 11/21/2022] Open
Abstract
The therapeutics for bone tissue regeneration requires constant advancements owing to the steady increase in the number of patients suffering from bone-related disorders, and also to find efficient and cost-effective treatment modalities. One of the major advancements in the field of therapeutics is the development of mRNAs. mRNAs, which have been extensively tested for the vaccines, could be very well utilized as a potential inducer for bone regeneration. The ability of mRNAs to enter the cells and instruct the cellular machinery to produce the required native proteins such as BMP or VEGF is a great way to avoid the issues faced with growth factor deliveries such as the production cost, loss of biological function etc. However, there have been a few hurdles for using mRNAs as an effective therapeutic agent, such as proper dosing, tolerating the degradation by RNases, improving the half-life, controlling the spatio-temporal release and reducing the off-target effects. This brief review discusses the various developments in the field of mRNA therapeutics especially for bone tissue engineering, how nano-formulations are being developed to effectively deliver the mRNAs into the cells by evading the immune responses, how researchers have developed certain strategies to increase the half-life, to successfully deliver the mRNAs to specific bone defect area and bring about effective bone regeneration.
Collapse
Affiliation(s)
- Arun Kumar Rajendran
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University Seoul 08826 Republic of Korea
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University Seoul 08826 Republic of Korea
- Institute for Engineering Research, Seoul National University Seoul 08826 Republic of Korea
| |
Collapse
|
45
|
Sukpaita T, Chirachanchai S, Pimkhaokham A, Ampornaramveth RS. Chitosan-Based Scaffold for Mineralized Tissues Regeneration. Mar Drugs 2021; 19:551. [PMID: 34677450 PMCID: PMC8540467 DOI: 10.3390/md19100551] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/20/2021] [Accepted: 09/26/2021] [Indexed: 12/20/2022] Open
Abstract
Conventional bone grafting procedures used to treat bone defects have several limitations. An important aspect of bone tissue engineering is developing novel bone substitute biomaterials for bone grafts to repair orthopedic defects. Considerable attention has been given to chitosan, a natural biopolymer primarily extracted from crustacean shells, which offers desirable characteristics, such as being biocompatible, biodegradable, and osteoconductive. This review presents an overview of the chitosan-based biomaterials for bone tissue engineering (BTE). It covers the basic knowledge of chitosan in terms of biomaterials, the traditional and novel strategies of the chitosan scaffold fabrication process, and their advantages and disadvantages. Furthermore, this paper integrates the relevant contributions in giving a brief insight into the recent research development of chitosan-based scaffolds and their limitations in BTE. The last part of the review discusses the next-generation smart chitosan-based scaffold and current applications in regenerative dentistry and future directions in the field of mineralized tissue regeneration.
Collapse
Affiliation(s)
- Teerawat Sukpaita
- Research Unit on Oral Microbiology and Immunology, Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Suwabun Chirachanchai
- Center of Excellence on Petrochemical and Materials Technology, Chulalongkorn University, Bangkok 10330, Thailand;
- Bioresources Advanced Materials (B2A), The Petroleum and Petrochemical College, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Atiphan Pimkhaokham
- Bioresources Advanced Materials (B2A), The Petroleum and Petrochemical College, Chulalongkorn University, Bangkok 10330, Thailand;
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | | |
Collapse
|
46
|
Wildemann B, Ignatius A, Leung F, Taitsman LA, Smith RM, Pesántez R, Stoddart MJ, Richards RG, Jupiter JB. Non-union bone fractures. Nat Rev Dis Primers 2021; 7:57. [PMID: 34354083 DOI: 10.1038/s41572-021-00289-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 11/09/2022]
Abstract
The human skeleton has remarkable regenerative properties, being one of the few structures in the body that can heal by recreating its normal cellular composition, orientation and mechanical strength. When the healing process of a fractured bone fails owing to inadequate immobilization, failed surgical intervention, insufficient biological response or infection, the outcome after a prolonged period of no healing is defined as non-union. Non-union represents a chronic medical condition not only affecting function but also potentially impacting the individual's psychosocial and economic well-being. This Primer provides the reader with an in-depth understanding of our contemporary knowledge regarding the important features to be considered when faced with non-union. The normal mechanisms involved in bone healing and the factors that disrupt the normal signalling mechanisms are addressed. Epidemiological considerations and advances in the diagnosis and surgical therapy of non-union are highlighted and the need for greater efforts in basic, translational and clinical research are identified.
Collapse
Affiliation(s)
- Britt Wildemann
- Experimental Trauma Surgery, Department of Trauma, Hand and Reconstructive Surgery, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany. .,Julius Wolff Institute and BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University, Ulm, Baden Württemberg, Germany
| | - Frankie Leung
- Department of Orthopaedics and Traumatology, Queen Mary Hospital, the University of Hong Kong, Hong Kong, Hong Kong
| | - Lisa A Taitsman
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, USA
| | - R Malcolm Smith
- Orthopedic trauma service, University of Massachusetts Medical School, Worcester, MA, USA
| | - Rodrigo Pesántez
- Departamento de Ortopedia Y Traumatología Fundación Santa Fé de Bogotá - Universidad de los Andes, Bogotá, Colombia
| | | | | | - Jesse B Jupiter
- Department of Orthopaedic surgery, Massachussets General Hospital, Boston, MA, USA.
| |
Collapse
|
47
|
A Damage Model to Trabecular Bone and Similar Materials: Residual Resource, Effective Elasticity Modulus, and Effective Stress under Uniaxial Compression. Symmetry (Basel) 2021. [DOI: 10.3390/sym13061051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Experimental research of bone strength remains costly and limited for ethical and technical reasons. Therefore, to predict the mechanical state of bone tissue, as well as similar materials, it is desirable to use computer technology and mathematical modeling. Yet, bone tissue as a bio-mechanical object with a hierarchical structure is difficult to analyze for strength and rigidity; therefore, empirical models are often used, the disadvantage of which is their limited application scope. The use of new analytical solutions overcomes the limitations of empirical models and significantly improves the way engineering problems are solved. Aim of the paper: the development of analytical solutions for computer models of the mechanical state of bone and similar materials. Object of research: a model of trabecular bone tissue as a quasi-brittle material under uniaxial compression (or tension). The new ideas of the fracture mechanics, as well as the methods of mathematical modeling and the biomechanics of bone tissues were used in the work. Compression and tension are considered as asymmetric mechanical states of the material. Results: a new nonlinear function that simulates both tension and compression is justified, analytical solutions for determining the effective and apparent elastic modulus are developed, the residual resource function and the damage function are justified, and the dependences of the initial and effective stresses on strain are obtained. Using the energy criterion, it is proven that the effective stress continuously increases both before and after the extremum point on the load-displacement plot. It is noted that the destruction of bone material is more likely at the inflection point of the load-displacement curve. The model adequacy is explained by the use of the energy criterion of material degradation. The results are consistent with the experimental data available in the literature.
Collapse
|
48
|
Sharma A, Puri V, Kumar P, Singh I, Huanbutta K. Development and Evaluation of Rifampicin Loaded Alginate-Gelatin Biocomposite Microfibers. Polymers (Basel) 2021; 13:polym13091514. [PMID: 34066853 PMCID: PMC8125895 DOI: 10.3390/polym13091514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/28/2022] Open
Abstract
Various systematic phases such as inflammation, tissue proliferation, and phases of remodeling characterize the process of wound healing. The natural matrix system is suggested to maintain and escalate these phases, and for that, microfibers were fabricated employing naturally occurring polymers (biopolymers) such as sodium alginate, gelatin and xanthan gum, and reinforcing material such as nanoclay was selected. The fabrication of fibers was executed with the aid of extrusion-gelation method. Rifampicin, an antibiotic, has been incorporated into a biopolymeric solution. RF1, RF2, RF3, RF4 and RF5 were coded as various formulation batches of microfibers. The microfibers were further characterized by different techniques such as SEM, DSC, XRD, and FTIR. Mechanical properties and physical evaluations such as entrapment efficiency, water uptake and in vitro release were also carried out to explain the comparative understanding of the formulation developed. The antimicrobial activity and whole blood clotting of fabricated fibers were additionally executed, hence they showed significant results, having excellent antimicrobial properties; they could be prominent carriers for wound healing applications.
Collapse
Affiliation(s)
- Ameya Sharma
- Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, India; (A.S.); (V.P.)
- Chitkara University School of Pharmacy, Chitkara University, Solan 174103, India
| | - Vivek Puri
- Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, India; (A.S.); (V.P.)
- Chitkara University School of Pharmacy, Chitkara University, Solan 174103, India
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, India; (A.S.); (V.P.)
- Correspondence: (I.S.); (K.H.)
| | - Kampanart Huanbutta
- Faculty of Pharmaceutical Sciences, Burapha University, 169, Saensook, Muang, Chonburi 20131, Thailand
- Correspondence: (I.S.); (K.H.)
| |
Collapse
|