51
|
Zhang J, Deng F, Liu X, Ge Y, Zeng Y, Zhai Z, Ning C, Li H. Favorable osteogenic activity of iron doped in silicocarnotite bioceramic: In vitro and in vivo Studies. J Orthop Translat 2022; 32:103-111. [PMID: 35228992 PMCID: PMC8856950 DOI: 10.1016/j.jot.2021.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 01/22/2023] Open
Abstract
Background Methods Results Conclusion The translational potential of this article
Collapse
|
52
|
Yang Y, Kulkarni A, Soraru GD, Pearce JM, Motta A. 3D Printed SiOC(N) Ceramic Scaffolds for Bone Tissue Regeneration: Improved Osteogenic Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms222413676. [PMID: 34948473 PMCID: PMC8706922 DOI: 10.3390/ijms222413676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022] Open
Abstract
Bone tissue engineering has developed significantly in recent years as there has been increasing demand for bone substitutes due to trauma, cancer, arthritis, and infections. The scaffolds for bone regeneration need to be mechanically stable and have a 3D architecture with interconnected pores. With the advances in additive manufacturing technology, these requirements can be fulfilled by 3D printing scaffolds with controlled geometry and porosity using a low-cost multistep process. The scaffolds, however, must also be bioactive to promote the environment for the cells to regenerate into bone tissue. To determine if a low-cost 3D printing method for bespoke SiOC(N) porous structures can regenerate bone, these structures were tested for osteointegration potential by using human mesenchymal stem cells (hMSCs). This includes checking the general biocompatibilities under the osteogenic differentiation environment (cell proliferation and metabolism). Moreover, cell morphology was observed by confocal microscopy, and gene expressions on typical osteogenic markers at different stages for bone formation were determined by real-time PCR. The results of the study showed the pore size of the scaffolds had a significant impact on differentiation. A certain range of pore size could stimulate osteogenic differentiation, thus promoting bone regrowth and regeneration.
Collapse
Affiliation(s)
- Yuejiao Yang
- BIOtech, Center for Biomedical Technologies, University of Trento, Via Sommarive 9, 38123 Trento, Italy;
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
- Correspondence: (Y.Y.); (A.K.)
| | - Apoorv Kulkarni
- Glass & Ceramics Lab, Department of Industrial Engineering, University of Trento, Via Sommerive 9, 38123 Trento, Italy;
- Correspondence: (Y.Y.); (A.K.)
| | - Gian Domenico Soraru
- Glass & Ceramics Lab, Department of Industrial Engineering, University of Trento, Via Sommerive 9, 38123 Trento, Italy;
| | - Joshua M. Pearce
- Department of Electrical and Computer Engineering, Western University, 1151 Richmond St. N., London, ON N6A 5B9, Canada;
| | - Antonella Motta
- BIOtech, Center for Biomedical Technologies, University of Trento, Via Sommarive 9, 38123 Trento, Italy;
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
| |
Collapse
|
53
|
Williams DF. Biocompatibility pathways and mechanisms for bioactive materials: The bioactivity zone. Bioact Mater 2021; 10:306-322. [PMID: 34901548 PMCID: PMC8636667 DOI: 10.1016/j.bioactmat.2021.08.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 08/07/2021] [Indexed: 12/14/2022] Open
Abstract
This essay analyzes the scientific evidence that forms the basis of bioactive materials, covering the fundamental understanding of bioactivity phenomena and correlation with the mechanisms of biocompatibility of biomaterials. This is a detailed assessment of performance in areas such as bone-induction, cell adhesion, immunomodulation, thrombogenicity and antimicrobial behavior. Bioactivity is the modulation of biological activity by characteristics of the interfacial region that incorporates the material surface and the immediate local host tissue. Although the term ‘bioactive material’ is widely used and has a well understood general meaning, it would be useful now to concentrate on this interfacial region, considered as ‘the bioactivity zone’. Bioactivity phenomena are either due to topographical/micromechanical characteristics, or to biologically active species that are presented in the bioactivity zone. Examples of topographical/micromechanical effects are the modulation of the osteoblast – osteoclast balance, nanotopographical regulation of cell adhesion, and bactericidal nanostructures. Regulation of bioactivity by biologically active species include their influence, especially of metal ions, on signaling pathways in bone formation, the role of cell adhesion molecules and bioactive peptides in cell attachment, macrophage polarization by immunoregulatory molecules and antimicrobial peptides. While much experimental data exists to demonstrate the potential of such phenomena, there are considerable barriers to their effective clinical translation. This essay shows that there is solid scientific evidence of the existence of bioactivity mechanisms that are associated with some types of biomaterials, especially when the material is modified in a manner designed to specifically induce that activity.
Collapse
Affiliation(s)
- David F Williams
- Wake Forest Institute of Regenerative Medicine, 391 Technology Way. Winston-Salem, North Carolina, 27101, USA
| |
Collapse
|
54
|
Kroschwald LM, Allerdt F, Bernhardt A, Rother S, Zheng K, Maqsood I, Halfter N, Heinemann C, Möller S, Schnabelrauch M, Hacker MC, Rammelt S, Boccaccini AR, Hintze V. Artificial Extracellular Matrices Containing Bioactive Glass Nanoparticles Promote Osteogenic Differentiation in Human Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms222312819. [PMID: 34884623 PMCID: PMC8657909 DOI: 10.3390/ijms222312819] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 02/07/2023] Open
Abstract
The present study analyzes the capacity of collagen (coll)/sulfated glycosaminoglycan (sGAG)-based surface coatings containing bioactive glass nanoparticles (BGN) in promoting the osteogenic differentiation of human mesenchymal stroma cells (hMSC). Physicochemical characteristics of these coatings and their effects on proliferation and osteogenic differentiation of hMSC were investigated. BGN were stably incorporated into the artificial extracellular matrices (aECM). Oscillatory rheology showed predominantly elastic, gel-like properties of the coatings. The complex viscosity increased depending on the GAG component and was further elevated by adding BGN. BGN-containing aECM showed a release of silicon ions as well as an uptake of calcium ions. hMSC were able to proliferate on coll and coll/sGAG coatings, while cellular growth was delayed on aECM containing BGN. However, a stimulating effect of BGN on ALP activity and calcium deposition was shown. Furthermore, a synergistic effect of sGAG and BGN was found for some donors. Our findings demonstrated the promising potential of aECM and BGN combinations in promoting bone regeneration. Still, future work is required to further optimize the BGN/aECM combination for increasing its combined osteogenic effect.
Collapse
Affiliation(s)
- Lysann M. Kroschwald
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital “Carl Gustav Carus”, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany; (L.M.K.); (A.B.)
| | - Felix Allerdt
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, D-01069 Dresden, Germany; (F.A.); (S.R.); (N.H.); (C.H.)
| | - Anne Bernhardt
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital “Carl Gustav Carus”, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany; (L.M.K.); (A.B.)
| | - Sandra Rother
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, D-01069 Dresden, Germany; (F.A.); (S.R.); (N.H.); (C.H.)
| | - Kai Zheng
- Institute of Biomaterials, University of Erlangen-Nuremberg, D-91058 Erlangen, Germany; (K.Z.); (A.R.B.)
| | - Iram Maqsood
- Institute for Pharmacy, Pharmaceutical Technology, University Leipzig, D-04317 Leipzig, Germany;
- Riphah Institute of Pharmaceutical Sciences (RIPS), Riphah International University (RIU), Lahore 54000, Pakistan
| | - Norbert Halfter
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, D-01069 Dresden, Germany; (F.A.); (S.R.); (N.H.); (C.H.)
| | - Christiane Heinemann
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, D-01069 Dresden, Germany; (F.A.); (S.R.); (N.H.); (C.H.)
| | - Stephanie Möller
- Biomaterials Department, INNOVENT e.V., D-07745 Jena, Germany; (S.M.); (M.S.)
| | | | - Michael C. Hacker
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, D-40225 Düsseldorf, Germany;
| | - Stefan Rammelt
- University Centre for Orthopaedics, Plastic and Trauma Surgery, University Hospital Carl Gustav Carus, D-01307 Dresden, Germany;
| | - Aldo R. Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, D-91058 Erlangen, Germany; (K.Z.); (A.R.B.)
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, D-01069 Dresden, Germany; (F.A.); (S.R.); (N.H.); (C.H.)
- Correspondence:
| |
Collapse
|
55
|
Silicified collagen scaffold induces semaphorin 3A secretion by sensory nerves to improve in-situ bone regeneration. Bioact Mater 2021; 9:475-490. [PMID: 34820584 PMCID: PMC8586786 DOI: 10.1016/j.bioactmat.2021.07.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/12/2021] [Accepted: 07/17/2021] [Indexed: 12/14/2022] Open
Abstract
Sensory nerves promote osteogenesis through the release of neuropeptides. However, the potential application and mechanism in which sensory nerves promote healing of bone defects in the presence of biomaterials remain elusive. The present study identified that new bone formation was more abundantly produced after implantation of silicified collagen scaffolds into defects created in the distal femur of rats. The wound sites were accompanied by extensive nerve innervation and angiogenesis. Sensory nerve dysfunction by capsaicin injection resulted in significant inhibition of silicon-induced osteogenesis in the aforementioned rodent model. Application of extracellular silicon in vitro induced axon outgrowth and increased expression of semaphorin 3 A (Sema3A) and semaphorin 4D (Sema4D) in the dorsal root ganglion (DRG), as detected by the upregulation of signaling molecules. Culture medium derived from silicon-stimulated DRG cells promoted proliferation and differentiation of bone marrow mesenchymal stem cells and endothelial progenitor cells. These effects were inhibited by the use of Sema3A neutralizing antibodies but not by Sema4D neutralizing antibodies. Knockdown of Sema3A in DRG blocked silicon-induced osteogenesis and angiogenesis almost completely in a femoral defect rat model, whereas overexpression of Sema3A promoted the silicon-induced phenomena. Activation of “mechanistic target of rapamycin” (mTOR) pathway and increase of Sema3A production were identified in the DRG of rats that were implanted with silicified collagen scaffolds. These findings support the role of silicon in inducing Sema3A production by sensory nerves, which, in turn, stimulates osteogenesis and angiogenesis. Taken together, silicon has therapeutic potential in orthopedic rehabilitation. Nerve innervation, vascularization and tissue mineralization integrated into a single scaffold. Silicified collagen scaffolds has therapeutic potential in orthopedic rehabilitation. Silicified collagen scaffolds promote in-situ bone regeneration via sensory nerve innervation and semaphorin 3A production.
Collapse
|
56
|
Li C, Yan T, Lou Z, Jiang Z, Shi Z, Chen Q, Gong Z, Wang B. Characterization and in vitro assessment of three-dimensional extrusion Mg-Sr codoped SiO 2-complexed porous microhydroxyapatite whisker scaffolds for biomedical engineering. Biomed Eng Online 2021; 20:116. [PMID: 34819108 PMCID: PMC8611959 DOI: 10.1186/s12938-021-00953-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Large bone defects have always been a great challenge for orthopedic surgeons. The use of a good bone substitute obtained by bone tissue engineering (BTE) may be an effective treatment method. Artificial hydroxyapatite, a commonly used bone defect filler, is the main inorganic component of bones. Because of its high brittleness, fragility, and lack of osteogenic active elements, its application is limited. Therefore, its fragility should be reduced, its osteogenic activity should be improved, and a more suitable scaffold should be constructed. METHODS In this study, a microhydroxyapatite whisker (mHAw) was developed, which was doped with the essential trace active elements Mg2+ and Sr2+ through a low-temperature sintering technique. After being formulated into a slurry, a bionic porous scaffold was manufactured by extrusion molding and freeze drying, and then SiO2 was used to improve the mechanical properties of the scaffold. The hydrophilicity, pore size, surface morphology, surface roughness, mechanical properties, and release rate of the osteogenic elements of the prepared scaffold were detected and analyzed. In in vitro experiments, Sprague-Dawley (SD) rat bone marrow mesenchymal stem cells (rBMSCs) were cultured on the scaffold to evaluate cytotoxicity, cell proliferation, spreading, and osteogenic differentiation. RESULTS Four types of scaffolds were obtained: mHAw-SiO2 (SHA), Mg-doped mHAw-SiO2 (SMHA), Sr-doped mHAw-SiO2 (SSHA), and Mg-Sr codoped mHAw-SiO2 (SMSHA). SHA was the most hydrophilic (WCA 5°), while SMHA was the least (WCA 8°); SMHA had the smallest pore size (247.40 ± 23.66 μm), while SSHA had the largest (286.20 ± 19.04 μm); SHA had the smallest Young's modulus (122.43 ± 28.79 MPa), while SSHA had the largest (188.44 ± 47.89 MPa); and SHA had the smallest compressive strength (1.72 ± 0.29 MPa), while SMHA had the largest (2.47 ± 0.25 MPa). The osteogenic active elements Si, Mg, and Sr were evenly distributed and could be sustainably released from the scaffolds. None of the scaffolds had cytotoxicity. SMSHA had the highest supporting cell proliferation and spreading rate, and its ability to promote osteogenic differentiation of rBMSCs was also the strongest. CONCLUSIONS These composite porous scaffolds not only have acceptable physical and chemical properties suitable for BTE but also have higher osteogenic bioactivity and can possibly serve as potential bone repair materials.
Collapse
Affiliation(s)
- Chengyong Li
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China
| | - Tingting Yan
- Faculty of Materials Science and Engineering, Kunming University of Science and Technology, Kunming, 650093, China
| | - Zhenkai Lou
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China
| | - Zhimin Jiang
- Faculty of Materials Science and Engineering, Kunming University of Science and Technology, Kunming, 650093, China
| | - Zhi Shi
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China
| | - Qinghua Chen
- Faculty of Materials Science and Engineering, Kunming University of Science and Technology, Kunming, 650093, China
| | - Zhiqiang Gong
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China
| | - Bing Wang
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
57
|
Zhu G, Zhang T, Chen M, Yao K, Huang X, Zhang B, Li Y, Liu J, Wang Y, Zhao Z. Bone physiological microenvironment and healing mechanism: Basis for future bone-tissue engineering scaffolds. Bioact Mater 2021; 6:4110-4140. [PMID: 33997497 PMCID: PMC8091181 DOI: 10.1016/j.bioactmat.2021.03.043] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/19/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023] Open
Abstract
Bone-tissue defects affect millions of people worldwide. Despite being common treatment approaches, autologous and allogeneic bone grafting have not achieved the ideal therapeutic effect. This has prompted researchers to explore novel bone-regeneration methods. In recent decades, the development of bone tissue engineering (BTE) scaffolds has been leading the forefront of this field. As researchers have provided deep insights into bone physiology and the bone-healing mechanism, various biomimicking and bioinspired BTE scaffolds have been reported. Now it is necessary to review the progress of natural bone physiology and bone healing mechanism, which will provide more valuable enlightenments for researchers in this field. This work details the physiological microenvironment of the natural bone tissue, bone-healing process, and various biomolecules involved therein. Next, according to the bone physiological microenvironment and the delivery of bioactive factors based on the bone-healing mechanism, it elaborates the biomimetic design of a scaffold, highlighting the designing of BTE scaffolds according to bone biology and providing the rationale for designing next-generation BTE scaffolds that conform to natural bone healing and regeneration.
Collapse
Affiliation(s)
- Guanyin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Ke Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Xinqi Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Bo Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yazhen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| |
Collapse
|
58
|
Cal F, Sezgin Arslan T, Derkus B, Kiran F, Cengiz U, Arslan YE. Synthesis of Silica-Based Boron-Incorporated Collagen/Human Hair Keratin Hybrid Cryogels with the Potential Bone Formation Capability. ACS APPLIED BIO MATERIALS 2021; 4:7266-7279. [PMID: 35006956 DOI: 10.1021/acsabm.1c00805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tissue engineering and regenerative medicine have evolved into a different concept, the so-called clinical tissue engineering. Within this context, the synthesis of next-generation inorganic-organic hybrid constructs without the use of chemical crosslinkers emerges with a great potential for treating bone defects. Here, we propose a sophisticated approach for synthesizing cost-effective boron (B)- and silicon (Si)-incorporated collagen/hair keratin (B-Si-Col-HK) cryogels with the help of sol-gel reactions. In this approach, collagen and hair keratin were engaged with a B-Si network using tetraethyl orthosilicate as a silica precursor, and the obtained cryogels were characterized in depth with attenuated total reflectance-Fourier transform infrared spectroscopy, solid-state NMR, X-ray diffraction, thermogravimetric analysis, porosity and swelling tests, Brunauer-Emmett-Teller and Barrett-Joyner-Halenda analyses, frequency sweep and temperature-dependent rheology, contact angle analysis, micromechanical tests, and scanning electron microscopy with energy dispersive X-ray analysis. In addition, the cell survival and osteogenic features of the cryogels were evaluated by the MTS test, live/dead assay, immuno/histochemistry, and quantitative real-time polymerase chain reaction analyses. We conclude that the B-Si-networked Col-HK cryogels having good mechanical durability and osteoinductive features would have the potential bone formation capability.
Collapse
Affiliation(s)
- Fatma Cal
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Tugba Sezgin Arslan
- Personalized Medicine and Biosensing Research (PMBR) Laboratory, Chemistry Department, Faculty of Science, Ankara University, Ankara 06560, Turkey
| | - Burak Derkus
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey.,Interdisciplinary Research Unit for Advanced Materials (INTRAM), Department of Chemistry, Ankara University, Ankara 06560, Turkey
| | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara 06100, Turkey
| | - Ugur Cengiz
- Surface Science Research Laboratory, Department of Chemical Engineering, Engineering Faculty, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| |
Collapse
|
59
|
Kamboj N, Ressler A, Hussainova I. Bioactive Ceramic Scaffolds for Bone Tissue Engineering by Powder Bed Selective Laser Processing: A Review. MATERIALS 2021; 14:ma14185338. [PMID: 34576562 PMCID: PMC8469313 DOI: 10.3390/ma14185338] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/02/2021] [Accepted: 09/12/2021] [Indexed: 02/07/2023]
Abstract
The implementation of a powder bed selective laser processing (PBSLP) technique for bioactive ceramics, including selective laser sintering and melting (SLM/SLS), a laser powder bed fusion (L-PBF) approach is far more challenging when compared to its metallic and polymeric counterparts for the fabrication of biomedical materials. Direct PBSLP can offer binder-free fabrication of bioactive scaffolds without involving postprocessing techniques. This review explicitly focuses on the PBSLP technique for bioactive ceramics and encompasses a detailed overview of the PBSLP process and the general requirements and properties of the bioactive scaffolds for bone tissue growth. The bioactive ceramics enclosing calcium phosphate (CaP) and calcium silicates (CS) and their respective composite scaffolds processed through PBSLP are also extensively discussed. This review paper also categorizes the bone regeneration strategies of the bioactive scaffolds processed through PBSLP with the various modes of functionalization through the incorporation of drugs, stem cells, and growth factors to ameliorate critical-sized bone defects based on the fracture site length for personalized medicine.
Collapse
Affiliation(s)
- Nikhil Kamboj
- Department of Mechanical and Industrial Engineering, Tallinn University of Technology, Ehitajate Tee 5, 19086 Tallinn, Estonia;
| | - Antonia Ressler
- Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev Trg 19, p.p.177, HR-10001 Zagreb, Croatia;
| | - Irina Hussainova
- Department of Mechanical and Industrial Engineering, Tallinn University of Technology, Ehitajate Tee 5, 19086 Tallinn, Estonia;
- Correspondence:
| |
Collapse
|
60
|
Shoushrah SH, Transfeld JL, Tonk CH, Büchner D, Witzleben S, Sieber MA, Schulze M, Tobiasch E. Sinking Our Teeth in Getting Dental Stem Cells to Clinics for Bone Regeneration. Int J Mol Sci 2021; 22:6387. [PMID: 34203719 PMCID: PMC8232184 DOI: 10.3390/ijms22126387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Dental stem cells have been isolated from the medical waste of various dental tissues. They have been characterized by numerous markers, which are evaluated herein and differentiated into multiple cell types. They can also be used to generate cell lines and iPSCs for long-term in vitro research. Methods for utilizing these stem cells including cellular systems such as organoids or cell sheets, cell-free systems such as exosomes, and scaffold-based approaches with and without drug release concepts are reported in this review and presented with new pictures for clarification. These in vitro applications can be deployed in disease modeling and subsequent pharmaceutical research and also pave the way for tissue regeneration. The main focus herein is on the potential of dental stem cells for hard tissue regeneration, especially bone, by evaluating their potential for osteogenesis and angiogenesis, and the regulation of these two processes by growth factors and environmental stimulators. Current in vitro and in vivo publications show numerous benefits of using dental stem cells for research purposes and hard tissue regeneration. However, only a few clinical trials currently exist. The goal of this review is to pinpoint this imbalance and encourage scientists to pick up this research and proceed one step further to translation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig- Strasse. 20, 53359 Rheinbach, Germany; (S.H.S.); (J.L.T.); (C.H.T.); (D.B.); (S.W.); (M.A.S.); (M.S.)
| |
Collapse
|
61
|
Dasgupta S, Mondal S, Ray S, Singh YP, Maji K. Hydroxyapatite-collagen nanoparticles reinforced polyanhydride based injectable paste for bone substitution: effect of dopant addition in vitro. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1312-1336. [PMID: 33874849 DOI: 10.1080/09205063.2021.1916867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The present study focuses on the synthesis and characterization of hydroxyapatite-collagen nanoparticles incorporated polyanhydride paste and investigating its bone regeneration capacity in vitro. Photocrosslinkable polyanhydride paste was prepared after synthesizing methacrylate derivatives of 1,6-bis(p-carboxyphenoxy)hexane (MCPH) and sebacic acid dimethacrylate (MSA). These multifunctional monomers, namely 45 wt% MSA, 45 wt% MCPH in addition to 10 wt% poly(ethylene glycol)diacrylate (PEGDA) were photopolymerized under ultraviolet light (365 nm) to produce highly crosslinked polyanhydride networks using camphroquinone (CQ)/ethyl 4-(dimethylamino)benzoate [4-EDMAB] for light initiated crosslinking and benzoyl peroxide (BPO)/dimethyl toludine (DMT) for chemically initiated crosslinking. Separately, using the co-precipitation process, (1 wt%) Si, (1 wt%) Sr, and (0.5 + 0.5) wt% Si/Sr was doped into hydroxyapatite-collagen nanoparticles in size range between 50 and 70 nm. Si, Sr, and both Si/Sr doped hydroxyapatite-collagen nanoparticles to the extent 10 wt% were added to polyanhydride monomer mixture containing 40 wt% MSA, 40 wt% MCPH and 10 wt% PEGDA and subsequently photopolymerized as previously mentioned. Incorporation of hydroxyapatite-collagen nanoparticles to the extent of 10 wt% into polyanhydride matrix enhanced compressive strength of the hardened paste from 30 to 49 MPa. Mesenchymal stem cells obtained from the human umbilical cord were cultured onto pure polyanhydride and hydroxyapatite-collagen added scaffold to assess their cellular proliferation and differentiation capacity to bone cell. MTT assay showed that mesenchymal stem cell proliferation was significantly higher in Si/Sr binary doped hydroxyapatite-collagen-polyanhydride sample as compared to other samples. Again from immunocytochemistry study using confocal images suggested that expression of osteocalcin, a marker indicating differentiation into osteoblast, was the highest in Si/Sr binary doped hydroxyapatite-collagen-polyanhydride sample against the other samples studied in this case. This study thus summarizes the development of photocurable biocomposites containing polyanhydride and Si, Sr doped hydroxyapatite-collagen nanoparticles that exhibited tremendous promise to regenerate bone tissues in complex-shaped musculoskeletal defect sites.
Collapse
Affiliation(s)
- Sudip Dasgupta
- Department of Ceramic Engineering, NIT Rourkela, Rourkela, Odisha, India
| | - Soumini Mondal
- Department of Ceramic Engineering, NIT Rourkela, Rourkela, Odisha, India
| | - Sambit Ray
- Department of Ceramic Engineering, NIT Rourkela, Rourkela, Odisha, India
| | | | - Kanchan Maji
- Department of Biotechnology and Medical Engineering, NIT Rourkela, Rourkela, Odisha, India
| |
Collapse
|
62
|
Šalandová M, van Hengel IAJ, Apachitei I, Zadpoor AA, van der Eerden BCJ, Fratila‐Apachitei LE. Inorganic Agents for Enhanced Angiogenesis of Orthopedic Biomaterials. Adv Healthc Mater 2021; 10:e2002254. [PMID: 34036754 PMCID: PMC11469191 DOI: 10.1002/adhm.202002254] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/30/2021] [Indexed: 01/02/2023]
Abstract
Aseptic loosening of a permanent prosthesis remains one of the most common reasons for bone implant failure. To improve the fixation between implant and bone tissue as well as enhance blood vessel formation, bioactive agents are incorporated into the surface of the biomaterial. This study reviews and compares five bioactive elements (copper, magnesium, silicon, strontium, and zinc) with respect to their effect on the angiogenic behavior of endothelial cells (ECs) when incorporated on the surface of biomaterials. Moreover, it provides an overview of the state-of-the-art methodologies used for the in vitro assessment of the angiogenic properties of these elements. Two databases are searched using keywords containing ECs and copper, magnesium, silicon, strontium, and zinc. After applying the defined inclusion and exclusion criteria, 59 articles are retained for the final assessment. An overview of the angiogenic properties of five bioactive elements and the methods used for assessment of their in vitro angiogenic potential is presented. The findings show that silicon and strontium can effectively enhance osseointegration through the simultaneous promotion of both angiogenesis and osteogenesis. Therefore, their integration onto the surface of biomaterials can ultimately decrease the incidence of implant failure due to aseptic loosening.
Collapse
Affiliation(s)
- Monika Šalandová
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Ingmar A. J. van Hengel
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Iulian Apachitei
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Amir A. Zadpoor
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Bram C. J. van der Eerden
- Department of Internal MedicineErasmus Medical CenterDoctor Molewaterplein 40Rotterdam3015 GDThe Netherlands
| | - Lidy E. Fratila‐Apachitei
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| |
Collapse
|
63
|
Wu WY, Chou PL, Yang JC, Chien CT. Silicon-containing water intake confers antioxidant effect, gastrointestinal protection, and gut microbiota modulation in the rodents. PLoS One 2021; 16:e0248508. [PMID: 33788857 PMCID: PMC8011764 DOI: 10.1371/journal.pone.0248508] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/26/2021] [Indexed: 01/17/2023] Open
Abstract
We explored the effects of silicon-containing water (BT) intake on gastrointestinal function and gut microbiota. BT was obtained by pressuring tap water through silicon minerals (mullite, Al6Si2O13) column. BT decreased H2O2 chemiluminescence counts, indicating its antioxidant activity. Four weeks of BT drinking increased H2O2 scavenging activity and glutathione peroxidase activity of plasma. BT drinking did not affect the body weight but significantly reduced the weight of feces and gastrointestinal motility. BT drinking significantly suppressed pylorus ligation enhanced gastric juice secretion, gastric reactive oxygen species amount, erythrocyte extravasation, IL-1β production by infiltrating leukocyte, and lipid peroxidation within gastric mucosa. Data from 16S rRNA sequencing revealed BT drinking significantly increased beneficial flora including Ruminococcaceae UCG-005, Prevotellaceae NK3B31, Weissella paramesenteroides, Lactobacillus reuteri, and Lactobacillus murinus and decreased harmful flora including Mucispirillum, Rodentibacter, and Staphylococcus aureus. This study pioneerly provided scientific evidences for the potential effects of water-soluble forms of silicon intake on antioxidant activity, gastrointestinal function, and gut microbiota modulation.
Collapse
Affiliation(s)
- Wei-Yi Wu
- Department of Life Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Pei-Li Chou
- Department of Life Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jyh-Chin Yang
- Department of Internal Medicine, Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail: (CTC); (JCY)
| | - Chiang-Ting Chien
- Department of Life Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
- * E-mail: (CTC); (JCY)
| |
Collapse
|
64
|
Feng KC, Wu YJ, Wang CY, Tu CS, Lin YL, Chen CS, Lai PL, Huang YT, Chen PY. Enhanced mechanical and biological performances of CaO-MgO-SiO 2 glass-ceramics via the modulation of glass and ceramic phases. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112060. [PMID: 33947554 DOI: 10.1016/j.msec.2021.112060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/26/2021] [Accepted: 03/20/2021] [Indexed: 12/23/2022]
Abstract
This work reports a new CaO-MgO-SiO2 (CMS) bioactive glass-ceramic, using ZrO2 as a nucleus to modulate the ratios of glass and ceramic phases as a function of sintering temperature. Mg-rich bioactive CMS glass-ceramics exhibit advantages regarding mechanical strength (flexural strength ~190 MPa and compressive strength ~555 MPa), in-vitro and in-vivo biocompatibilities, and bone ingrowth. The high mechanical strengths could be attributed to the CaMgSi2O6 glass-ceramic and lower porosity. X-ray absorption spectra indicate an increased SiO covalent bond via the development of CaMgSi2O6 glass-ceramics. From the in-vitro cytotoxicity and BMSC differentiation assays, the CMS samples sintered above 800 °C exhibited better cell attachment and differentiation, possibly due to structural stability, appropriate pore, and ion release to boost osteogenesis. Compared to hydroxyapatite (HA) ceramics, the CMS glass-ceramics display higher mechanical strengths, biocompatibility, and osteoconductivity. An in-vivo experiment demonstrated a fine bone-ingrowth profile around the CMS implant. This study may further the application of CMS glass-ceramics in bone implants.
Collapse
Affiliation(s)
- Kuei-Chih Feng
- Research Center for Intelligent Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Mechanical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Yu-Jie Wu
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Chi-Yun Wang
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan
| | - Chi-Shun Tu
- Research Center for Intelligent Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Physics, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Yu-Ling Lin
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Cheng-Sao Chen
- Department of Mechanical Engineering, Hwa Hsia University of Technology, New Taipei City 23567, Taiwan
| | - Po-Liang Lai
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan
| | - Yu-Tzu Huang
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan.
| | - Pin-Yi Chen
- Research Center for Intelligent Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Mechanical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan.
| |
Collapse
|
65
|
Rondanelli M, Faliva MA, Peroni G, Gasparri C, Perna S, Riva A, Petrangolini G, Tartara A. Silicon: A neglected micronutrient essential for bone health. Exp Biol Med (Maywood) 2021; 246:1500-1511. [PMID: 33715532 DOI: 10.1177/1535370221997072] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Bone matrix is predominantly made up of collagen, and in vitro and in animal models studies have shown that silicon is linked to glycosaminoglycans and plays an important role in the formation of cross-links between collagen and proteoglycans, determining the beneficial effects on strength, composition, and mechanical properties of bone. However, there are still no precise indications regarding a possible role of silicon on bone health in humans. Given this background, the aim of this narrative review was to consider the effectiveness of silicon dietary intake and silicon dietary supplementation (alone or with other micronutrients), in order to suggest a daily dosage of Si supplementation, on bone mineral density in humans. This review included eight eligible studies: four regarding dietary intake and four considering supplementation with silicon alone or with other nutrients. Despite the number of studies considered being low, the number of subjects studied is high (10012) and the results are interesting. Although to date the available scientific evidences are not considered valid enough to allow to establish an adequate level of Silicon intake, based on extrapolations from the data obtained with studies on animal and human models, it has been suggested that an adequate intake in order to promote beneficial effects for bone could be considered to be around 25 mg silicon/day. As for silicon dietary supplements, it has been shown that the combined treatment with orthosilicic acid (6 mg), calcium, and vitamin D has a potentially beneficial effect on femoral BMD compared to only use of calcium and vitamin D.
Collapse
Affiliation(s)
- Mariangela Rondanelli
- IRCCS Mondino Foundation, Pavia 27100, Italy.,Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, Pavia 27100, Italy
| | - Milena A Faliva
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ''Istituto Santa Margherita'', University of Pavia, Pavia 27100, Italy
| | - Gabriella Peroni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ''Istituto Santa Margherita'', University of Pavia, Pavia 27100, Italy
| | - Clara Gasparri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ''Istituto Santa Margherita'', University of Pavia, Pavia 27100, Italy
| | - Simone Perna
- Department of Biology, College of Science, University of Bahrain, Sakhir 32038, Bahrain
| | | | | | - Alice Tartara
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ''Istituto Santa Margherita'', University of Pavia, Pavia 27100, Italy
| |
Collapse
|
66
|
Sahvieh S, Oryan A, Hassanajili S, Kamali A. Role of bone 1stem cell-seeded 3D polylactic acid/polycaprolactone/hydroxyapatite scaffold on a critical-sized radial bone defect in rat. Cell Tissue Res 2021; 383:735-750. [PMID: 32924069 DOI: 10.1007/s00441-020-03284-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/14/2020] [Indexed: 01/07/2023]
Abstract
Osteoconductive biomaterials were used to find the most reliable materials in bone healing. Our focus was on the bone healing capacity of the stem cell-loaded and unloaded PLA/PCL/HA scaffolds. The 3D scaffold of PLA/PCL/HA was characterized by scanning electron microscopy (SEM), rheology, X-ray diffraction (XRD), and Fourier transform-infrared (FT-IR) spectroscopy. Bone marrow stem cells (BMSCs) have multipotential differentiation into osteoblasts. Forty Wistar male rats were used to organize four experimental groups: control, autograft, scaffold, and BMSCs-loaded scaffold groups. qRT-PCR showed that the BMSCs-loaded scaffold had a higher expression level of CD31 and osteogenic markers compared with the control group (P < 0.05). Radiology and computed tomography (CT) scan evaluations showed significant improvement in the BMSCs-loaded scaffold compared with the control group (P < 0.001). Biomechanical estimation demonstrated significantly higher stress (P < 0.01), stiffness (P < 0.001), and ultimate load (P < 0.01) in the autograft and BMSCs-loaded scaffold groups compared with the untreated group and higher strain was seen in the control group than the other groups (P < 0.01). Histomorphometric and immunohistochemical (IHC) investigations showed significantly improved regeneration scores in the autograft and BMSCs-loaded scaffold groups compared with the control group (P < 0.05). Also, there was a significant difference between the scaffold and control groups in all tests (P < 0.05). The results depicted that our novel approach will allow to develop PLA/PCL/HA 3D scaffold in bone healing via BMSC loading.
Collapse
Affiliation(s)
- Sonia Sahvieh
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ahmad Oryan
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Shadi Hassanajili
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz, Iran
| | - Amir Kamali
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
67
|
Hydroxyapatite Based Materials for Bone Tissue Engineering: A Brief and Comprehensive Introduction. CRYSTALS 2021. [DOI: 10.3390/cryst11020149] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hydroxyapatite (HA) is widely used in bone tissue engineering for its bioactivity and biocompatibility, and a growing number of researchers are exploring ways to improve the physical properties and biological functions of hydroxyapatite. Up to now, HA has been used as inorganic building blocks for tissue engineering or as nanofillers to blend with polymers, furthermore, various methods such as ion doping or surface modification have been also reported to prepare functionalized HA. In this review, we try to give a brief and comprehensive introduction about HA-based materials, including ion-doped HA, HA/polymer composites and surface modified HA and their applications in bone tissue engineering. In addition, the prospective of HA is also discussed. This review may be helpful for researchers to get a general understanding about the development of hydroxyapatite based materials.
Collapse
|
68
|
Yilmaz HD, Cengiz U, Arslan YE, Kiran F, Ceylan A. From a plant secretion to the promising bone grafts: Cryogels of silicon-integrated quince seed mucilage by microwave-assisted sol-gel reaction. J Biosci Bioeng 2021; 131:420-433. [PMID: 33454223 DOI: 10.1016/j.jbiosc.2020.11.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/06/2020] [Accepted: 11/25/2020] [Indexed: 01/13/2023]
Abstract
Design and fabrication of biologically active cryogels using novel biopolymer(s) are still of great importance at regenerating bone defects such as traumatic bone injuries, maxillofacial surgery, osteomyelitis, and osteoporosis. Nowadays, plant mucilage, an herbal biomaterial, has been drawn attention by scientists due to their marvelous potential to fabricate 3-dimensional (3D) physical constructs for the field of regenerative medicine. Herein, a 3D cryogel from silicon-integrated quince seed mucilage (QSM) is constructed using microwave-assisted sol-gel reaction, characterized in-depth by attenuated total reflectance Fourier transform-infrared spectroscopy (ATR-FTIR), solid-state silicon cross-polarization magic-angle nuclear magnetic resonance (29Si-CP-MAS NMR), X-ray diffraction (XRD), thermogravimetric analysis (TGA), differential scanning calorimeter (DSC), micro-mechanical testing, porosity, and swelling tests, contact angle measurements, Brunauer-Emmet-Teller and Barret-Joyner-Halenda (BET-BJH) analysis, enzymatic biodegradation test, and field emission-scanning electron microscopy-energy dispersive X-ray spectroscopy (FE-SEM-EDX) mapping. The osteobiologic capacity of the cryogels is determined using human adipose-derived mesenchymal stem cells (hAMSCs) under in vitro conditions. Osteogenic differentiation of hAMSCs on both QSM and silica-modified QSM (Si-QSM) cryogels is analyzed by histochemistry, immunohistochemistry, and quantitative-real time (q-RT) PCR techniques. The results obtained from in vitro experiments demonstrate that the upregulation of osteogenesis-related genes in Si-QSM cryogels presents a stronger and earlier development over QSM cryogels throughout the culture period, which in turn reveals the great potential of this novel Si-incorporated QSM cryogels for bone tissue engineering applications.
Collapse
Affiliation(s)
- Hilal Deniz Yilmaz
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Engineering Faculty, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey
| | - Ugur Cengiz
- Surface Science Research Laboratory, Department of Chemical Engineering, Engineering Faculty, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Engineering Faculty, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey.
| | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara 06100, Turkey
| | - Ahmet Ceylan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara 06110, Turkey
| |
Collapse
|
69
|
Han R, Buchanan F, Ford L, Julius M, Walsh P. A comparison of the degradation behaviour of 3D printed PDLGA scaffolds incorporating bioglass or biosilica. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111755. [DOI: 10.1016/j.msec.2020.111755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/21/2020] [Accepted: 11/21/2020] [Indexed: 10/22/2022]
|
70
|
Grzeskowiak RM, Schumacher J, Dhar MS, Harper DP, Mulon PY, Anderson DE. Bone and Cartilage Interfaces With Orthopedic Implants: A Literature Review. Front Surg 2020; 7:601244. [PMID: 33409291 PMCID: PMC7779634 DOI: 10.3389/fsurg.2020.601244] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/25/2020] [Indexed: 12/21/2022] Open
Abstract
The interface between a surgical implant and tissue consists of a complex and dynamic environment characterized by mechanical and biological interactions between the implant and surrounding tissue. The implantation process leads to injury which needs to heal over time and the rapidity of this process as well as the property of restored tissue impact directly the strength of the interface. Bleeding is the first and most relevant step of the healing process because blood provides growth factors and cellular material necessary for tissue repair. Integration of the implants placed in poorly vascularized tissue such as articular cartilage is, therefore, more challenging than compared with the implants placed in well-vascularized tissues such as bone. Bleeding is followed by the establishment of a provisional matrix that is gradually transformed into the native tissue. The ultimate goal of implantation is to obtain a complete integration between the implant and tissue resulting in long-term stability. The stability of the implant has been defined as primary (mechanical) and secondary (biological integration) stability. Successful integration of an implant within the tissue depends on both stabilities and is vital for short- and long-term surgical outcomes. Advances in research aim to improve implant integration resulting in enhanced implant and tissue interface. Numerous methods have been employed to improve the process of modifying both stability types. This review provides a comprehensive discussion of current knowledge regarding implant-tissue interfaces within bone and cartilage as well as novel approaches to strengthen the implant-tissue interface. Furthermore, it gives an insight into the current state-of-art biomechanical testing of the stability of the implants. Current knowledge reveals that the design of the implants closely mimicking the native structure is more likely to become well integrated. The literature provides however several other techniques such as coating with a bioactive compound that will stimulate the integration and successful outcome for the patient.
Collapse
Affiliation(s)
- Remigiusz M. Grzeskowiak
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Jim Schumacher
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Madhu S. Dhar
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - David P. Harper
- The Center for Renewable Carbon, Institute of Agriculture, University of Tennessee, Knoxville, TN, United States
| | - Pierre-Yves Mulon
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - David E. Anderson
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| |
Collapse
|
71
|
B P, U V. In Vitro bioactivity, biocompatibility and corrosion resistance of multi-ionic (Ce/Si) co-doped hydroxyapatite porous coating on Ti-6Al-4 V for bone regeneration applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111620. [PMID: 33321662 DOI: 10.1016/j.msec.2020.111620] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/26/2020] [Accepted: 10/06/2020] [Indexed: 12/27/2022]
Abstract
Dual-doped hydroxyapatite (Ce4+/Si4+ doped HAP) coating with admirable bacterial resistance and enriched bioactivity was fabricated via spin-coating technique. In this study, Ce/Si co-doped hydroxyapatite was coated on Ti-6Al-4 V substrates as a triple layer with extreme centrifugal force (2000 RPM, 3000 RPM and 4000 RPM) to improve the biological performance of the coating in terms of enhanced bone apposition. Further, the coated substrate was characterized by XRD, FTIR and SEM-EDS techniques. The contact angle of the coating was measured through the sessile drop method and in vitro biomineralization was carried out in SBF solution to predict the apatite formation on the surface of the coated implant. Pathogen restriction behaviour of the coating was studied using gram-negative and gram-positive bacteria such as Staphylococcus aureus, Bacillus subtilis, Escherichia coli and Pseudomonas aeruginosa respectively. Among these, gram-negative bacteria, Escherichia coli revealed greater inhibition than other bacteria. In vitro cell viability assay using MG-63 osteoblast cell was performed for the optimised coating acquired at 4000 RPM and the result showed excellent biocompatibility towards the cell line. Corrosion resistance behaviour of the coating using Polarization and EIS study exhibited excellent corrosion resistance. Therefore, based on the in vitro studies, the designed multifunctional coating can act as a potential biomaterial in the field of biomedical engineering.
Collapse
Affiliation(s)
- Priyadarshini B
- Department of Chemistry, School of Advanced Sciences, VIT, Vellore 632 014, Tamil Nadu, India
| | - Vijayalakshmi U
- Department of Chemistry, School of Advanced Sciences, VIT, Vellore 632 014, Tamil Nadu, India.
| |
Collapse
|
72
|
Ferreira SA, Young G, Jones JR, Rankin S. Bioglass/carbonate apatite/collagen composite scaffold dissolution products promote human osteoblast differentiation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111393. [PMID: 33254998 DOI: 10.1016/j.msec.2020.111393] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/30/2022]
Abstract
OssiMend® Bioactive (Collagen Matrix Inc., NJ) is a three-component porous composite bone graft device of 45S5 Bioglass/carbonate apatite/collagen. Our in vitro studies showed that conditioned media of the dissolution products of OssiMend Bioactive stimulated primary human osteoblasts to form mineralized bone-like nodules in vitro in one week, in basal culture media (no osteogenic supplements). Osteoblast differentiation was followed by gene expression analysis and a mineralization assay. In contrast, the dissolution products from commercial OssiMend (Bioglass-free carbonate apatite/collagen scaffolds), or from 45S5 Bioglass particulate alone, did not induce the mineralization of the extracellular matrix, but did induce osteoblast differentiation to mature osteoblasts, evidenced by the strong upregulation of BGLAP and IBSP mRNA levels. The calcium ions and soluble silicon species released from 45S5 Bioglass particles and additional phosphorus release from OssiMend mediated the osteostimulatory effects. Medium conditioned with OssiMend Bioactive dissolution had a much higher concentration of phosphorus and silicon than media conditioned with OssiMend and 45S5 Bioglass alone. While OssiMend and OssiMend Bioactive led to calcium precipitation in cell culture media, OssiMend Bioactive produced a higher concentration of soluble silicon than 45S5 Bioglass and higher dissolution of phosphorus than OssiMend. These in vitro results suggest that adding 45S5 Bioglass to OssiMend produces a synergistic osteostimulation effect on primary human osteoblasts. In summary, dissolution products of a Bioglass/carbonate apatite/collagen composite scaffold (OssiMend® Bioactive) stimulate human osteoblast differentiation and mineralization of extracellular matrix in vitro without any osteogenic supplements. The mineralization was faster than for dissolution products of ordinary Bioglass.
Collapse
Affiliation(s)
- Silvia A Ferreira
- National Heart & Lung Institute, Imperial College London, London, UK.
| | - Gloria Young
- Department of Materials, Imperial College London, London, UK.
| | - Julian R Jones
- Department of Materials, Imperial College London, London, UK.
| | - Sara Rankin
- National Heart & Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
73
|
Colchicine mesoporous silica nanoparticles/hydrogel composite loaded cotton patches as a new encapsulator system for transdermal osteoarthritis management. Int J Biol Macromol 2020; 164:1149-1163. [DOI: 10.1016/j.ijbiomac.2020.07.133] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/02/2020] [Accepted: 07/11/2020] [Indexed: 01/01/2023]
|
74
|
Oryan A, Hassanajili S, Sahvieh S. Effectiveness of a biodegradable 3D polylactic acid/poly(ɛ-caprolactone)/hydroxyapatite scaffold loaded by differentiated osteogenic cells in a critical-sized radius bone defect in rat. J Tissue Eng Regen Med 2020; 15:150-162. [PMID: 33216449 DOI: 10.1002/term.3158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/31/2020] [Accepted: 11/09/2020] [Indexed: 11/06/2022]
Abstract
The effects of a scaffold made of polylactic acid, poly (ɛ-caprolactone) and hydroxyapatite by indirect 3D printing method with and without differentiated bone cells was tested on the regeneration of a critical radial bone defect in rat. The scaffold characterization and mechanical performance were determined by the rheology, scanning electron microscopy, energy-dispersive X-ray spectroscopy, X-ray diffraction, and Fourier transform infrared spectrometry. The defects were created in forty Wistar rats which were randomly divided into the untreated, autograft, scaffold cell-free, and differentiated bone cell-seeded scaffold groups (n = 10 in each group). The expression level of angiogenic and osteogenic markers, analyzed by quantitative real time-polymerase chain reaction (in vitro), significantly improved (p < 0.05) in the scaffold group compared to the untreated one. Radiology and computed tomography scan demonstrated a significant improvement in the cell-seeded scaffold group compared to the untreated one (p < 0.001). Biomechanical, histopathological, histomorphometric, and immunohistochemical investigations showed significantly better regeneration scores in the cell-seeded scaffold and autograft groups compared to the untreated group (p < 0.05). The cell-seeded scaffold and autograft groups did show comparable results on the 80th day post-treatment (p > 0.05), however, most results in the scaffold group were significantly higher than the untreated group (p < 0.05). Differentiated bone cells can enhance bone regeneration potential of the scaffold.
Collapse
Affiliation(s)
- Ahmad Oryan
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Shadi Hassanajili
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz, Iran
| | - Sonia Sahvieh
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
75
|
Cottrill E, Premananthan C, Pennington Z, Ehresman J, Theodore N, Sciubba DM, Witham T. Radiographic and clinical outcomes of silicate-substituted calcium phosphate (SiCaP) bone grafts in spinal fusion: Systematic review and meta-analysis. J Clin Neurosci 2020; 81:353-366. [PMID: 33222944 DOI: 10.1016/j.jocn.2020.09.073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/10/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
Pseudarthrosis continues to affect a nontrivial proportion of spine fusion patients. Given its ties to poorer patient outcomes and high reoperation rates, there remains great interest in interventions aimed at reducing the rates of nonunion. Recently, silicate-substituted calcium phosphate (SiCaP) bone grafts have been suggested to improve fusion rates, yet there exists no systematic review of the body of evidence for SiCaP grafts. Here, we present the first such review along with a meta-analysis of the effect of SiCaP bone grafts on fusion rates. Using the PubMed, Embase, and Web of Science databases, we queried the English-language literature for all studies examining the effect of SiCaPs on spinal fusion. Primary endpoints were: 1) radiographic fusion rate at last follow-up and 2) postoperative improvements in Visual Analog Scale (VAS) pain scores and Oswestry Disability Index (ODI) at last follow-up. Meta-analyses were performed for each endpoint using random effects. Ten articles (694 patients treated with SiCaP bone grafts) were included. Among SiCaP-treated patients, 93% achieved radiographic fusion (range: 79-100%), with comparable rates across subgroups. Meta-analysis of the three randomized controlled trials demonstrated no difference in fusion rates between SiCaP-treated patients and patients receiving grafts with recombinant human bone morphogenetic protein-2 (rhBMP-2) (OR: 1.11; p = 0.83). Patients treated with SiCaP bone grafts experienced significant improvements in VAS back pain (-3.3 points), VAS leg pain (-4.8 points), and ODI (-31.6 points) by last follow-up (p < 0.001 for each). Additional high-quality research is needed to evaluate the relative cost-effectiveness of SiCaP bone grafts in spinal fusion.
Collapse
Affiliation(s)
- Ethan Cottrill
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christine Premananthan
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zach Pennington
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeff Ehresman
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas Theodore
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel M Sciubba
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Timothy Witham
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
76
|
Reyes-Peces MV, Pérez-Moreno A, de-los-Santos DM, Mesa-Díaz MDM, Pinaglia-Tobaruela G, Vilches-Pérez JI, Fernández-Montesinos R, Salido M, de la Rosa-Fox N, Piñero M. Chitosan-GPTMS-Silica Hybrid Mesoporous Aerogels for Bone Tissue Engineering. Polymers (Basel) 2020; 12:polym12112723. [PMID: 33212958 PMCID: PMC7698430 DOI: 10.3390/polym12112723] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/13/2020] [Accepted: 11/14/2020] [Indexed: 01/10/2023] Open
Abstract
This study introduces a new synthesis route for obtaining homogeneous chitosan (CS)-silica hybrid aerogels with CS contents up to 10 wt%, using 3-glycidoxypropyl trimethoxysilane (GPTMS) as coupling agent, for tissue engineering applications. Aerogels were obtained using the sol-gel process followed by CO2 supercritical drying, resulting in samples with bulk densities ranging from 0.17 g/cm3 to 0.38 g/cm3. The textural analysis by N2-physisorption revealed an interconnected mesopore network with decreasing specific surface areas (1230-700 m2/g) and pore sizes (11.1-8.7 nm) by increasing GPTMS content (2-4 molar ratio GPTMS:CS monomer). In addition, samples exhibited extremely fast swelling by spontaneous capillary imbibition in PBS solution, presenting swelling capacities from 1.75 to 3.75. The formation of a covalent crosslinked hybrid structure was suggested by FTIR and confirmed by an increase of four hundred fold or more in the compressive strength up to 96 MPa. Instead, samples synthesized without GPTMS fractured at only 0.10-0.26 MPa, revealing a week structure consisted in interpenetrated polymer networks. The aerogels presented bioactivity in simulated body fluid (SBF), as confirmed by the in vitro formation of hydroxyapatite (HAp) layer with crystal size of approximately 2 µm size in diameter. In vitro studies revealed also non cytotoxic effect on HOB® osteoblasts and also a mechanosensitive response. Additionally, control cells grown on glass developed scarce or no stress fibers, while cells grown on hybrid samples showed a significant (p < 0.05) increase in well-developed stress fibers and mature focal adhesion complexes.
Collapse
Affiliation(s)
- María V. Reyes-Peces
- Department of Condensed Matter Physics 1, Faculty of Science, University of Cadiz, 11510 Cádiz, Spain; (M.V.R.-P.); (A.P.-M.); (N.d.l.R.-F.)
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (G.P.-T.); (J.I.V.-P.); (R.F.-M.); (M.S.)
| | - A. Pérez-Moreno
- Department of Condensed Matter Physics 1, Faculty of Science, University of Cadiz, 11510 Cádiz, Spain; (M.V.R.-P.); (A.P.-M.); (N.d.l.R.-F.)
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (G.P.-T.); (J.I.V.-P.); (R.F.-M.); (M.S.)
| | | | - María del Mar Mesa-Díaz
- Department of Chemical Engineering, Faculty of Science University of Cadiz, 11510 Cádiz, Spain;
- Instituto de Microscopía Electrónica y Materiales (IMEYMAT), University of Cadiz, 11510 Cádiz, Spain
| | - Gonzalo Pinaglia-Tobaruela
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (G.P.-T.); (J.I.V.-P.); (R.F.-M.); (M.S.)
- Department of Histology, SCIBM, Faculty of Medicine University of Cadiz, 11004 Cádiz, Spain
| | - Jose Ignacio Vilches-Pérez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (G.P.-T.); (J.I.V.-P.); (R.F.-M.); (M.S.)
- Department of Histology, SCIBM, Faculty of Medicine University of Cadiz, 11004 Cádiz, Spain
| | - Rafael Fernández-Montesinos
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (G.P.-T.); (J.I.V.-P.); (R.F.-M.); (M.S.)
- Department of Histology, SCIBM, Faculty of Medicine University of Cadiz, 11004 Cádiz, Spain
| | - Mercedes Salido
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (G.P.-T.); (J.I.V.-P.); (R.F.-M.); (M.S.)
- Department of Histology, SCIBM, Faculty of Medicine University of Cadiz, 11004 Cádiz, Spain
| | - Nicolás de la Rosa-Fox
- Department of Condensed Matter Physics 1, Faculty of Science, University of Cadiz, 11510 Cádiz, Spain; (M.V.R.-P.); (A.P.-M.); (N.d.l.R.-F.)
- Instituto de Microscopía Electrónica y Materiales (IMEYMAT), University of Cadiz, 11510 Cádiz, Spain
| | - Manuel Piñero
- Department of Condensed Matter Physics 1, Faculty of Science, University of Cadiz, 11510 Cádiz, Spain; (M.V.R.-P.); (A.P.-M.); (N.d.l.R.-F.)
- Instituto de Microscopía Electrónica y Materiales (IMEYMAT), University of Cadiz, 11510 Cádiz, Spain
- Correspondence:
| |
Collapse
|
77
|
Arab‐Ahmadi S, Irani S, Bakhshi H, Atyabi F, Ghalandari B. Immobilization of carboxymethyl chitosan/laponite on polycaprolactone nanofibers as osteoinductive bone scaffolds. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.5128] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Samira Arab‐Ahmadi
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Hadi Bakhshi
- Department of Functional Polymer Systems Fraunhofer Institute for Applied Polymer Research, Geiselbergstraße 68 Potsdam Germany
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy Tehran University of Medical Sciences Tehran Iran
- Nanotechnology Research Centre, Faculty of Pharmacy Tehran University of Medical Sciences Tehran Iran
| | - Behafarid Ghalandari
- Department of Medical Nanotechnology, Applied Biophotonics Research Center, Science and Research Branch Islamic Azad University Tehran Iran
| |
Collapse
|
78
|
Mesa M. Chitosan and silica as dietary carriers: Potential application for β-galactosidase, silicon and calcium supplementation. Food Hydrocoll 2020. [DOI: 10.1016/j.foodhyd.2020.106067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
79
|
Ma R, Li Y, Wang J, Yang P, Wang K, Wang W. Incorporation of nanosized calcium silicate improved osteointegration of polyetheretherketone under diabetic conditions. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:98. [PMID: 33130931 DOI: 10.1007/s10856-020-06435-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/24/2020] [Indexed: 06/11/2023]
Abstract
Diabetes can impair osteoblastic functions and negatively interfere with osteointegration at the bone/implant interface. Previously, we prepared a nanosized calcium silicate (CS) incorporated-polyetheretherketone (PK) biocomposite (CS/PK) and found that the CS/PK composite exhibited enhanced osteoblast functions in vitro and osteointegration in vivo, but its bioperformance under diabetic conditions remained elusive. In this study, MC3T3-E1 cells incubated on CS/PK and PK samples were subjected to diabetic serum (DS) and normal serum (NS); cell attachment, morphology, spreading, proliferation, and osteogenic differentiation were compared to assess in vitro osteoblastic functions on the surfaces of different materials. An in vivo test was performed on diabetic rabbits implanted with CS/PK or PK implants into the cranial bone defect to assess the osteointegration ability of the implants. In vitro results showed that diabetes inhibited osteoblastic functions evidenced by impaired morphology and spreading, and decreased attachment, proliferation, and osteogenic differentiation compared with the findings under normal conditions. Notably, CS/PK ameliorated osteoblastic disfunction under diabetic conditions in vitro. In vivo results from micro-CT and histologic examinations revealed that rabbits with CS/PK implants exhibited improved osteointegration at the bone/implant interface under diabetic conditions compared with PK. Therefore, the CS/PK composite improved the impaired osteointegration induced by diabetes and is a promising orthopedic or craniofacial implant material that may obtain good clinical performance in diabetic patients.
Collapse
Affiliation(s)
- Rui Ma
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shanxi, China
| | - Yongwei Li
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shanxi, China
| | - Jialin Wang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shanxi, China
| | - Pei Yang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shanxi, China
| | - Kunzheng Wang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shanxi, China.
| | - Wei Wang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shanxi, China.
| |
Collapse
|
80
|
Apanasevich V, Papynov E, Plekhova N, Zinoviev S, Kotciurbii E, Stepanyugina A, Korshunova O, Afonin I, Evdokimov I, Shichalin O, Bardin A, Nevozhai V, Polezhaev A. Morphological Characteristics of the Osteoplastic Potential of Synthetic CaSiO 3/HAp Powder Biocomposite. J Funct Biomater 2020; 11:jfb11040068. [PMID: 32977458 PMCID: PMC7712391 DOI: 10.3390/jfb11040068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 02/06/2023] Open
Abstract
The study describes the influence of synthetic CaSiO3/HAp powder biocomposite on the process of regeneration in osseous tissue in the alveolar ridges in terms of the morphological characteristics of the osteoplastic potential. The authors investigated the osteoinduction and osteoconduction “in vivo” processes during bone tissue regeneration in the mandible defect area of an experimental animal (rabbit). The possibility of angiogenesis in the graft as an adaptation factor was studied in the process of bone tissue regeneration. The results of the histological study that included the qualitative parameters of bone tissue regeneration, the morphometric parameters (microarchitectonics) of the bone, the parameters of osteosynthesis (thickness of the osteoid plates), and resorption (volume density of the eroded surface) were presented. The results allowed the authors to characterize the possibility of the practical adaptation for synthetic powder biocomposite as an osteoplastic graft for the rehabilitation of osseous defects in dentistry.
Collapse
Affiliation(s)
- Vladimir Apanasevich
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| | - Evgeniy Papynov
- Institute of Chemistry, Far Eastern Branch of Russian Academy of Sciences, 159, Prosp. 100-letiya Vladivostoka, Vladivostok 690022, Russia;
- Far Eastern Federal University, 8, Sukhanova St., Vladivostok 690091, Russia;
- Correspondence:
| | - Nataliay Plekhova
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| | - Sergey Zinoviev
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| | - Evgeniy Kotciurbii
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| | - Alexandra Stepanyugina
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| | - Oksana Korshunova
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| | - Igor Afonin
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| | - Ivan Evdokimov
- Far Eastern Federal University, 8, Sukhanova St., Vladivostok 690091, Russia;
| | - Oleg Shichalin
- Institute of Chemistry, Far Eastern Branch of Russian Academy of Sciences, 159, Prosp. 100-letiya Vladivostoka, Vladivostok 690022, Russia;
- Far Eastern Federal University, 8, Sukhanova St., Vladivostok 690091, Russia;
| | - Artem Bardin
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| | - Vladimir Nevozhai
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| | - Alexandr Polezhaev
- Central Research Laboratory, Institute of Surgery, Pacific State Medical University, 2, Ostryakov Aven., Vladivostok 690990, Russia; (V.A.); (N.P.); (S.Z.); (E.K.); (A.S.); (O.K.); (I.A.); (A.B.); (V.N.); (A.P.)
| |
Collapse
|
81
|
Oryan A, Hassanajili S, Sahvieh S, Azarpira N. Effectiveness of mesenchymal stem cell-seeded onto the 3D polylactic acid/polycaprolactone/hydroxyapatite scaffold on the radius bone defect in rat. Life Sci 2020; 257:118038. [PMID: 32622947 DOI: 10.1016/j.lfs.2020.118038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE The importance of regeneration in large bone defects forces the orthopedic surgeons to search for a proper methodology. The present experiment evaluated the capability of polylactic acid/polycaprolactone/hydroxyapatite (PLA/PCL/HA) scaffold loaded with and without mesenchymal stem cells (MSCs) on bone regeneration. METHODS Fourier transform infrared spectrometry, X-ray diffraction, scanning electron microscopy, and rheology methodologies were used to characterize the scaffold. Forty Wistar rats were randomly divided into the four groups including the untreated defects as the control group and three other groups in which the bone defects were treated with autologous bones (autograft group), the PLA/PCL/HA scaffolds (PLA/PCL/HA group), and the MSCs-seeded scaffolds (MSCs-seeded PLA/PCL/HA group). RESULTS Based on the qRT-PCR results, significantly higher expression levels of osteocalcin, osteopontin, and CD31 were seen in the cell-seeded scaffold group compared to the control group (P < 0.05). The CT scanning and radiographic images depicted significantly more newly formed bonny tissue in the MSCs-loaded scaffold and autograft groups than the untreated group (P < 0.001). The immunohistochemistry, biomechanical, histopathologic, and histomorphometric evaluations demonstrated significantly improved regeneration in the autograft and MSCs-loaded scaffold groups compared to the non-treated group (P < 0.05). There were significant differences between the scaffold and untreated groups in all in vivo evaluations (P < 0.05). CONCLUSION The MSCs enhanced bone healing potential of the PLA/PCL/HA scaffold and the MSCs-seeded scaffold was comparable to the autograft as the golden treatment regimen (P > 0.05).
Collapse
Affiliation(s)
- A Oryan
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - S Hassanajili
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz, Iran
| | - S Sahvieh
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - N Azarpira
- Transplant Research Center, Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
82
|
Baranova J, Büchner D, Götz W, Schulze M, Tobiasch E. Tooth Formation: Are the Hardest Tissues of Human Body Hard to Regenerate? Int J Mol Sci 2020; 21:E4031. [PMID: 32512908 PMCID: PMC7312198 DOI: 10.3390/ijms21114031] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
With increasing life expectancy, demands for dental tissue and whole-tooth regeneration are becoming more significant. Despite great progress in medicine, including regenerative therapies, the complex structure of dental tissues introduces several challenges to the field of regenerative dentistry. Interdisciplinary efforts from cellular biologists, material scientists, and clinical odontologists are being made to establish strategies and find the solutions for dental tissue regeneration and/or whole-tooth regeneration. In recent years, many significant discoveries were done regarding signaling pathways and factors shaping calcified tissue genesis, including those of tooth. Novel biocompatible scaffolds and polymer-based drug release systems are under development and may soon result in clinically applicable biomaterials with the potential to modulate signaling cascades involved in dental tissue genesis and regeneration. Approaches for whole-tooth regeneration utilizing adult stem cells, induced pluripotent stem cells, or tooth germ cells transplantation are emerging as promising alternatives to overcome existing in vitro tissue generation hurdles. In this interdisciplinary review, most recent advances in cellular signaling guiding dental tissue genesis, novel functionalized scaffolds and drug release material, various odontogenic cell sources, and methods for tooth regeneration are discussed thus providing a multi-faceted, up-to-date, and illustrative overview on the tooth regeneration matter, alongside hints for future directions in the challenging field of regenerative dentistry.
Collapse
Affiliation(s)
- Juliana Baranova
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Avenida Professor Lineu Prestes 748, Vila Universitária, São Paulo 05508-000, Brazil;
| | - Dominik Büchner
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Straße 20, 53359 Rheinbach, NRW, Germany; (D.B.); (M.S.)
| | - Werner Götz
- Oral Biology Laboratory, Department of Orthodontics, Dental Hospital of the University of Bonn, Welschnonnenstraße 17, 53111 Bonn, NRW, Germany;
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Straße 20, 53359 Rheinbach, NRW, Germany; (D.B.); (M.S.)
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Straße 20, 53359 Rheinbach, NRW, Germany; (D.B.); (M.S.)
| |
Collapse
|
83
|
An In Vivo Study in Rat Femurs of Bioactive Silicate Coatings on Titanium Dental Implants. J Clin Med 2020; 9:jcm9051290. [PMID: 32365687 PMCID: PMC7288333 DOI: 10.3390/jcm9051290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/10/2020] [Accepted: 04/26/2020] [Indexed: 12/16/2022] Open
Abstract
Silica-based ceramics have been proposed for coating purposes to enhance dental and orthopedic titanium (Ti) implant bioactivity. The aim of this study was to investigate the influence of sphene-based bioceramic (CaO.TiO2.SiO2) coatings on implant osseointegration in vivo. Sphene coatings were obtained from preceramic polymers and nano-sized active precursors and deposited by an automatic airbrush. Twenty customized Ti implants, ten sphene-coated and ten uncoated rough implants were implanted into the proximal femurs of ten Sprague-Dawley rats. Overall, cortical and cancellous bone-to-implant contact (BIC) were determined using micro-computed tomography (micro-CT) at 14 and 28 days. Moreover, peri-implant bone healing was histologically and histomorphometrically evaluated. The white blood cell count in the synovial fluid of the knee joints, if present, was also assessed. No difference in the BIC values was observed between the sphene-coated and uncoated implants, overall and in the two bone compartments (p > 0.05). Delamination of the coating occurred in three cases. Consistently with micro-CT data, the histological evaluation revealed no differences between the two groups. In addition, no synovial fluid could be collected on the test side, thus confirming sphene biocompatibility. In conclusion, sphene coating was found to be a suitable material for biomedical applications. Further studies are needed to improve coating adhesion to the implants.
Collapse
|
84
|
Li Y, Zhang X, Dai C, Yin Y, Gong L, Pan W, Huang R, Bu Y, Liao X, Guo K, Gao F. Bioactive Three-Dimensional Graphene Oxide Foam/Polydimethylsiloxane/Zinc Silicate Scaffolds with Enhanced Osteoinductivity for Bone Regeneration. ACS Biomater Sci Eng 2020; 6:3015-3025. [PMID: 33463276 DOI: 10.1021/acsbiomaterials.9b01931] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Nanocomposite scaffold materials have shown great prospect in promoting bone integration and bone regeneration. A three-dimensional graphene oxide foam/polydimethylsiloxane/zinc silicate (GF/PDMS/ZS) scaffold for bone tissue engineering was synthesized via dip coating and hydrothermal synthesis processes, resulting in the interconnected macroporous structure. The scaffold was characterized with scanning electron microscopy (SEM), X-ray diffraction (XRD), X-ray photoelectron spectroscopy (XPS), and thermogravimetric (TG) analysis. The result showed that scaffolds exhibiting a porous characteristic had organic-inorganic components similar to natural bone tissue. Moreover, the scaffolds possessed suitable pore size, high porosity, and good mechanical properties. In vitro experiments with mouse bone marrow mesenchymal stem cells (mBMSCs) revealed that the composite scaffold not only has great biocompatibility but also has the ability to induce mBMSC proliferation and preferential osteogentic differentiation. Thereafter, the expression of critical genes, ALP, RUNX2, VEGFA, and OPN, was activated. In vivo analysis of critical bone defect in rabbits demonstrated superior bone formation in defect sites in the GF/PDMS/ZS scaffold group at 12 weeks of post implantation without no significant inflammatory response. All the results validated that the GF/PDMS/ZS scaffold is a promising alternative for applications in bone regeneration.
Collapse
Affiliation(s)
- Yang Li
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China.,Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Xing Zhang
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Chengbai Dai
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China.,Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Yiming Yin
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China
| | - Ling Gong
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China
| | - Wenzhen Pan
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China.,Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Ruqi Huang
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China.,Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Yeyang Bu
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China.,Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Xianjiu Liao
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China
| | - Kaijin Guo
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China.,Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 221002, Xuzhou, China
| | - Fenglei Gao
- School of Pharmacy, Xuzhou Medical University, 221004, Xuzhou, China
| |
Collapse
|
85
|
Liang W, Gao M, Lou J, Bai Y, Zhang J, Lu T, Sun X, Ye J, Li B, Sun L, Heng BC, Zhang X, Deng X. Integrating silicon/zinc dual elements with PLGA microspheres in calcium phosphate cement scaffolds synergistically enhances bone regeneration. J Mater Chem B 2020; 8:3038-3049. [PMID: 32196049 DOI: 10.1039/c9tb02901j] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Integrating multiple pro-osteogenic factors into bone graft substitutes is a practical and effective approach to improve bone repair efficacy. Here, Si-Zn dual elements and PLGA microspheres were incorporated into calcium phosphate cement (CPC) scaffolds (PLGA/CPC-Si/Zn) as a novel strategy to synergistically enhance bone regeneration. The incorporation of PLGA microspheres and Si/Zn dual elements within CPC scaffolds improved the setting time, injectability and compressive strength. The PLGA/CPC-Si/Zn scaffolds displayed controlled sequential release of Si and Zn ions. In vitro, RAW 264.7 cells displayed the M2 phenotype with a high level of anti-inflammatory cytokines in response to PLGA/CPC-Si/Zn. The conditioned medium of RAW 264.7 cells cultured on the PLGA/CPC-Si/Zn scaffolds significantly enhanced the osteogenic differentiation of rat BMSCs. In a rat femur defect model, the implanted PLGA/CPC-Si/Zn scaffolds led to obvious new bone formation after 4 weeks, apparent bone ingrowth into the PLGA microspheres after 12 weeks, and was almost completely filled with mature new bone upon degradation of the PLGA microspheres at 24 weeks. These findings demonstrate that the PLGA/CPC-Si/Zn scaffolds promote osteogenesis by synergistically improving the immune microenvironment and biodegradability. Hence, integrating multiple trace elements together with degradable components within bone graft biomaterials can be an effective strategy for promoting bone regeneration.
Collapse
Affiliation(s)
- Weiwei Liang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Kondiah PJ, Kondiah PPD, Choonara YE, Marimuthu T, Pillay V. A 3D Bioprinted Pseudo-Bone Drug Delivery Scaffold for Bone Tissue Engineering. Pharmaceutics 2020; 12:E166. [PMID: 32079221 PMCID: PMC7076403 DOI: 10.3390/pharmaceutics12020166] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/27/2020] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
A 3D bioprinted pseudo-bone drug delivery scaffold was fabricated to display matrix strength, matrix resilience, as well as porous morphology of healthy human bone. Computer-aided design (CAD) software was employed for developing the 3D bioprinted scaffold. Further optimization of the scaffold was undertaken using MATLAB® software and artificial neural networks (ANN). Polymers employed for formulating the 3D scaffold comprised of polypropylene fumarate (PPF), free radical polymerized polyethylene glycol- polycaprolactone (PEG-PCL-PEG), and pluronic (PF127). Simvastatin was incorporated into the 3D bioprinted scaffolds to further promote bone healing and repair properties. The 3D bioprinted scaffold was characterized for its chemical, morphological, mechanical, and in vitro release kinetics for evaluation of its behavior for application as an implantable scaffold at the site of bone fracture. The ANN-optimized 3D bioprinted scaffold displayed significant properties as a controlled release platform, demonstrating drug release over 20 days. The 3D bioprinted scaffold further displayed formation as a pseudo-bone matrix, using a human clavicle bone model, induced with a butterfly fracture. The strength of the pseudo-bone matrix, evaluated for its matrix hardness (MH) and matrix resilience (MR), was evaluated to be as strong as original bone, having a 99% MH and 98% MR property, to healthy human clavicle bones.
Collapse
Affiliation(s)
| | | | | | | | - Viness Pillay
- 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, 7 York Road, Parktown 2193, South Africa; (P.J.K.); (P.P.D.K.); (Y.E.C.); (T.M.)
| |
Collapse
|
87
|
Ford NR, Xiong Y, Hecht KA, Squier TC, Rorrer GL, Roesijadi G. Optimizing the Design of Diatom Biosilica-Targeted Fusion Proteins in Biosensor Construction for Bacillus anthracis Detection. BIOLOGY 2020; 9:biology9010014. [PMID: 31936120 PMCID: PMC7168173 DOI: 10.3390/biology9010014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/24/2019] [Accepted: 01/04/2020] [Indexed: 11/16/2022]
Abstract
In vivo functionalization of diatom biosilica frustules by genetic manipulation requires careful consideration of the overall structure and function of complex fusion proteins. Although we previously had transformed Thalassiosira pseudonana with constructs containing a single domain antibody (sdAb) raised against the Bacillus anthracis Sterne strain, which detected an epitope of the surface layer protein EA1 accessible in lysed spores, we initially were unsuccessful with constructs encoding a similar sdAb that detected an epitope of EA1 accessible in intact spores and vegetative cells. This discrepancy limited the usefulness of the system as an environmental biosensor for B. anthracis. We surmised that to create functional biosilica-localized biosensors with certain constructs, the biosilica targeting and protein trafficking functions of the biosilica-targeting peptide Sil3T8 had to be uncoupled. We found that retaining the ER trafficking sequence at the N-terminus and relocating the Sil3T8 targeting peptide to the C-terminus of the fusion protein resulted in successful detection of EA1 with both sdAbs. Homology modeling of antigen binding by the two sdAbs supported the hypothesis that the rescue of antigen binding in the previously dysfunctional sdAb was due to removal of steric hindrances between the antigen binding loops and the diatom biosilica for that particular sdAb.
Collapse
Affiliation(s)
- Nicole R. Ford
- Marine Biotechnology Group, Pacific Northwest National Laboratory, Sequim, WA 98382, USA
- Correspondence:
| | - Yijia Xiong
- Department of Basic Medical Sciences, Western University of Health Sciences, Lebanon, OR 97355, USA
| | - Karen A. Hecht
- Marine Biotechnology Group, Pacific Northwest National Laboratory, Sequim, WA 98382, USA
| | - Thomas C. Squier
- Department of Basic Medical Sciences, Western University of Health Sciences, Lebanon, OR 97355, USA
| | - Gregory L. Rorrer
- School of Chemical Biological and Environmental Engineering, Oregon State University, Corvallis, OR 97331, USA
| | - Guritno Roesijadi
- Marine Biotechnology Group, Pacific Northwest National Laboratory, Sequim, WA 98382, USA
- School of Chemical Biological and Environmental Engineering, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
88
|
Liu J, Shen X, Tang S, Li H, Mei S, Zheng H, Sun Y, Zhao J, Kaewmanee R, Yang L, Gan Q, Wei J. Improvement of rBMSCs Responses to Poly(propylene carbonate) Based Biomaterial through Incorporation of Nanolaponite and Surface Treatment Using Sodium Hydroxide. ACS Biomater Sci Eng 2019; 6:329-339. [PMID: 33463218 DOI: 10.1021/acsbiomaterials.9b01137] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Poly(propylene carbonate) (PPC) has aroused extensive attention in the biomaterial field because of its excellent biocompatibility and appropriate degradability, but surface hydrophobicity and bioinertness limit its applications for bone repair and tissue engineering. In this study, a bioactive PPC/laponite (LAP) nanocomposite (PL) was prepared by a melt-blending method, and a microporous surface on PPC and PL (PT and PLT) was created by sodium hydroxide (NaOH) treatment. The results demonstrated that the surface roughness, hydrophilicity, surface energy, and degradability as well as protein adsorption of PLT were obviously improved compared with PPC. Moreover, the degradability of PLT was remarkably enhanced with a slight increase of pH values in Tris-HCl solution. Furthermore, adhesion and proliferation as well as osteogenic differentiation of rat bone marrow mesenchymal stem cells (rBMSCs) to PLT were significantly promoted compared with PPC. The results suggested that incorporating LAP into PPC obviously improved the surface performance of PL (with nanotopography), and surface treatment with NaOH further enhanced surface properties of PLT (with micronanotopography and hydrophilic groups), which significantly promoted responses of rBMSCs. In short, PLT displayed excellent cytocompatibility, which would have great potential for bone regeneration.
Collapse
Affiliation(s)
- Jingyuan Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, No.130, Meilong Road, Shanghai 200237, China
| | - Xuening Shen
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, No.130, Meilong Road, Shanghai 200237, China
| | - Songchao Tang
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, No.130, Meilong Road, Shanghai 200237, China
| | - Hong Li
- College of Physical Science and Technology, Sichuan University, No. 17, South Renmin Road, Chengdu 610041, China
| | - Shiqi Mei
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, No.130, Meilong Road, Shanghai 200237, China
| | - Han Zheng
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, No.130, Meilong Road, Shanghai 200237, China
| | - Yupeng Sun
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, No.130, Meilong Road, Shanghai 200237, China
| | - Jun Zhao
- Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, No. 639, Manufacturing Bureau Road, Shanghai 200011, China
| | - Rames Kaewmanee
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, No.130, Meilong Road, Shanghai 200237, China
| | - Lili Yang
- Department of Orthopaedic Surgery, Changzheng Hospital, The Second Military Medical University, No. 415, Fengyang Road, Shanghai 200003, China
| | - Qi Gan
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, No.130, Meilong Road, Shanghai 200237, China
| | - Jie Wei
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, No.130, Meilong Road, Shanghai 200237, China
| |
Collapse
|
89
|
Witzler M, Büchner D, Shoushrah SH, Babczyk P, Baranova J, Witzleben S, Tobiasch E, Schulze M. Polysaccharide-Based Systems for Targeted Stem Cell Differentiation and Bone Regeneration. Biomolecules 2019; 9:E840. [PMID: 31817802 PMCID: PMC6995597 DOI: 10.3390/biom9120840] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/22/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023] Open
Abstract
Bone tissue engineering is an ever-changing, rapidly evolving, and highly interdisciplinary field of study, where scientists try to mimic natural bone structure as closely as possible in order to facilitate bone healing. New insights from cell biology, specifically from mesenchymal stem cell differentiation and signaling, lead to new approaches in bone regeneration. Novel scaffold and drug release materials based on polysaccharides gain increasing attention due to their wide availability and good biocompatibility to be used as hydrogels and/or hybrid components for drug release and tissue engineering. This article reviews the current state of the art, recent developments, and future perspectives in polysaccharide-based systems used for bone regeneration.
Collapse
Affiliation(s)
- Markus Witzler
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (M.W.); (D.B.); (S.H.S.); (P.B.); (S.W.); (E.T.)
| | - Dominik Büchner
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (M.W.); (D.B.); (S.H.S.); (P.B.); (S.W.); (E.T.)
| | - Sarah Hani Shoushrah
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (M.W.); (D.B.); (S.H.S.); (P.B.); (S.W.); (E.T.)
| | - Patrick Babczyk
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (M.W.); (D.B.); (S.H.S.); (P.B.); (S.W.); (E.T.)
| | - Juliana Baranova
- Laboratory of Neurosciences, Department of Biochemistry, Institute of Chemistry–USP, University of São Paulo, Avenida Professor Lineu Prestes 748, Vila Universitaria, São Paulo, SP 05508-000, Brazil;
| | - Steffen Witzleben
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (M.W.); (D.B.); (S.H.S.); (P.B.); (S.W.); (E.T.)
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (M.W.); (D.B.); (S.H.S.); (P.B.); (S.W.); (E.T.)
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (M.W.); (D.B.); (S.H.S.); (P.B.); (S.W.); (E.T.)
| |
Collapse
|
90
|
Brunello G, Elsayed H, Biasetto L. Bioactive Glass and Silicate-Based Ceramic Coatings on Metallic Implants: Open Challenge or Outdated Topic? MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2929. [PMID: 31510062 PMCID: PMC6766230 DOI: 10.3390/ma12182929] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 12/19/2022]
Abstract
The overall success and long-term life of the medical implants are decisively based on the convenient osseointegration at the hosting tissue-implant interface. Therefore, various surface modifications and different coating approaches have been utilized to the implants to enhance the bone formation and speed up the interaction with the surrounding hosting tissues, thereby enabling the successful fixation of implants. In this review, we will briefly present the main metallic implants and discuss their biocompatibility and osseointegration ability depending on their chemical and mechanical properties. In addition, as the main goal of this review, we explore the main properties of bioactive glasses and silica-based ceramics that are used as coating materials for both orthopedic and dental implants. The current review provides an overview of these bioactive coatings, with a particular emphasis on deposition methods, coating adhesion to the substrates and apatite formation ability tested by immersion in Simulated Body Fluid (SBF). In vitro and in vivo performances in terms of biocompatibility, biodegradability and improved osseointegration are examined as well.
Collapse
Affiliation(s)
- Giulia Brunello
- Department of Management and Engineering, University of Padova, Stradella San Nicola 3, 36100 Vicenza, Italy.
- Department of Neurosciences, Section of Dentistry, University of Padova, Via Giustiniani 2, 35128 Padova, Italy.
| | - Hamada Elsayed
- Department of Industrial Engineering, University of Padova, Via F. Marzolo 9, 35131 Padova, Italy.
- Ceramics Department, National Research Centre, El-Bohous Street, Cairo 12622, Egypt.
| | - Lisa Biasetto
- Department of Management and Engineering, University of Padova, Stradella San Nicola 3, 36100 Vicenza, Italy.
| |
Collapse
|
91
|
Skwira A, Szewczyk A, Prokopowicz M. The Effect of Polydimethylsiloxane-Ethylcellulose Coating Blends on the Surface Characterization and Drug Release of Ciprofloxacin-Loaded Mesoporous Silica. Polymers (Basel) 2019; 11:E1450. [PMID: 31487861 PMCID: PMC6780097 DOI: 10.3390/polym11091450] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/02/2019] [Accepted: 09/02/2019] [Indexed: 12/22/2022] Open
Abstract
In this study, we obtained novel solid films composed of ciprofloxacin-loaded mesoporous silica materials (CIP-loaded MCM-41) and polymer coating blends. Polymer coating blends were composed of ethylcellulose (EC) with various levels of polydimethylsiloxane (PDMS, 0, 1, 2% (v/v)). The solid films were prepared via the solvent-evaporation molding method and characterized by using scanning electron microscopy (SEM), optical profilometry, and wettability analyses. The solid-state of CIP present in the solid films was studied using X-ray diffraction (XRD) and differential scanning calorimetry (DSC). The release profiles of CIP were examined as a function of PDMS content in solid films. The surface morphology analysis of solid films indicated the progressive increase in surface heterogeneity and roughness with increasing PDMS content. The contact angle study confirmed the hydrophobicity of all solid films and significant impact of both PDMS and CIP-loaded MCM-41 on surface wettability. DSC and XRD analysis confirmed the presence of amorphous/semi-crystalline CIP in solid films. The Fickian diffusion-controlled drug release was observed for the CIP-loaded MCM-41 coated with PDMS-free polymer blend, whereas zero-order drug release was noticed for the CIP-loaded MCM-41 coated with polymer blends enriched with PDMS. Both the release rate and initial burst of CIP decreased with increasing PDMS content.
Collapse
Affiliation(s)
- Adrianna Skwira
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Hallera 107, Gdańsk 80-416, Poland
| | - Adrian Szewczyk
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Hallera 107, Gdańsk 80-416, Poland
| | - Magdalena Prokopowicz
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Hallera 107, Gdańsk 80-416, Poland.
| |
Collapse
|
92
|
Witzler M, Ottensmeyer PF, Gericke M, Heinze T, Tobiasch E, Schulze M. Non-Cytotoxic Agarose/Hydroxyapatite Composite Scaffolds for Drug Release. Int J Mol Sci 2019; 20:E3565. [PMID: 31330875 PMCID: PMC6678963 DOI: 10.3390/ijms20143565] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022] Open
Abstract
Healing of large bone defects requires implants or scaffolds that provide structural guidance for cell growth, differentiation, and vascularization. In the present work, an agarose-hydroxyapatite composite scaffold was developed that acts not only as a 3D matrix, but also as a release system. Hydroxyapatite (HA) was incorporated into the agarose gels in situ in various ratios by a simple procedure consisting of precipitation, cooling, washing, and drying. The resulting gels were characterized regarding composition, porosity, mechanical properties, and biocompatibility. A pure phase of carbonated HA was identified in the scaffolds, which had pore sizes of up to several hundred micrometers. Mechanical testing revealed elastic moduli of up to 2.8 MPa for lyophilized composites. MTT testing on Lw35human mesenchymal stem cells (hMSCs) and osteosarcoma MG-63 cells proved the biocompatibility of the scaffolds. Furthermore, scaffolds were loaded with model drug compounds for guided hMSC differentiation. Different release kinetic models were evaluated for adenosine 5'-triphosphate (ATP) and suramin, and data showed a sustained release behavior over four days.
Collapse
Affiliation(s)
- Markus Witzler
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany
- Institute of Organic Chemistry and Macromolecular Chemistry, Center of Excellence of Polysaccharide Research, Friedrich Schiller University of Jena, Humboldtstr. 10, 07743 Jena, Germany
| | - Patrick Frank Ottensmeyer
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany
| | - Martin Gericke
- Institute of Organic Chemistry and Macromolecular Chemistry, Center of Excellence of Polysaccharide Research, Friedrich Schiller University of Jena, Humboldtstr. 10, 07743 Jena, Germany
| | - Thomas Heinze
- Institute of Organic Chemistry and Macromolecular Chemistry, Center of Excellence of Polysaccharide Research, Friedrich Schiller University of Jena, Humboldtstr. 10, 07743 Jena, Germany
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany.
| |
Collapse
|