1
|
Oliveira M, Angelova L, Avdeev G, Grenho L, Fernandes MH, Daskalova A. Femtosecond Laser-Engineered β-TCP Scaffolds: A Comparative Study of Green-Synthesized AgNPs vs. Ion Doping Against S. aureus for Bone Regeneration. Int J Mol Sci 2025; 26:4888. [PMID: 40430028 PMCID: PMC12112484 DOI: 10.3390/ijms26104888] [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: 04/08/2025] [Revised: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Implant-associated infections, particularly those linked to Staphylococcus aureus (S. aureus), continue to compromise the clinical success of β-tricalcium phosphate (β-TCP) implants despite their excellent biocompatibility and osteoconductivity. This investigation aims to tackle these challenges by integrating femtosecond (fs)-laser surface processing with two complementary strategies: ion doping and functionalization with green-synthesized silver nanoparticles (AgNPs). AgNPs were produced via fs-laser photoreduction using green tea leaf extract (GTLE), noted for its anti-inflammatory and antioxidant properties. Fs-laser processing was applied to modify β-TCP scaffolds by systematically varying scanning velocities, fluences, and patterns. Lower scanning velocities generated organized nanostructures with enhanced roughness and wettability, as confirmed by scanning electron microscopy (SEM), optical profilometry, and contact angle measurements, whereas higher laser energies induced significant phase transitions between hydroxyapatite (HA) and α-tricalcium phosphate (α-TCP), as revealed by X-ray diffraction (XRD). AgNP-functionalized scaffolds demonstrated markedly superior antibacterial activity against S. aureus compared to the ion-doped variants, attributed to the synergistic interplay of nanostructure-mediated surface disruption and AgNP-induced bactericidal mechanisms. Although ion-doped scaffolds exhibited limited direct antibacterial effects, they showed concentration-dependent activity in indirect assays, likely due to controlled ion release. Both strategies promoted osteogenic differentiation of human bone marrow mesenchymal stem cells (hBM-MSCs) under defined conditions, albeit with transient cytotoxicity at higher fluences and excessive ion doping. Overall, this approach holds promise for markedly improving antibacterial efficacy and osteogenic compatibility, potentially transforming bone regeneration therapies.
Collapse
Affiliation(s)
- Marco Oliveira
- Institute of Electronics, Bulgarian Academy of Sciences, 72 Tsarigradsko Chaussee Blvd, 1784 Sofia, Bulgaria; (M.O.); (L.A.)
| | - Liliya Angelova
- Institute of Electronics, Bulgarian Academy of Sciences, 72 Tsarigradsko Chaussee Blvd, 1784 Sofia, Bulgaria; (M.O.); (L.A.)
| | - Georgi Avdeev
- Institute of Physical Chemistry, Bulgarian Academy of Sciences, Akad. G. Bonchev Str., 1113 Sofia, Bulgaria;
| | - Liliana Grenho
- BoneLab—Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal; (L.G.); (M.H.F.)
- LAQV/REQUIMTE–Associated Laboratory for Green Chemistry, Research Group “Materials for Sustainability and Wellbeing”, University of Porto, 4160-007 Porto, Portugal
| | - Maria Helena Fernandes
- BoneLab—Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal; (L.G.); (M.H.F.)
- LAQV/REQUIMTE–Associated Laboratory for Green Chemistry, Research Group “Materials for Sustainability and Wellbeing”, University of Porto, 4160-007 Porto, Portugal
| | - Albena Daskalova
- Institute of Electronics, Bulgarian Academy of Sciences, 72 Tsarigradsko Chaussee Blvd, 1784 Sofia, Bulgaria; (M.O.); (L.A.)
| |
Collapse
|
2
|
Zhang H, Qiao W, Liu Y, Yao X, Zhai Y, Du L. Addressing the challenges of infectious bone defects: a review of recent advances in bifunctional biomaterials. J Nanobiotechnology 2025; 23:257. [PMID: 40158189 PMCID: PMC11954225 DOI: 10.1186/s12951-025-03295-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025] Open
Abstract
Infectious bone defects present a substantial clinical challenge due to the complex interplay between infection control and bone regeneration. These defects often result from trauma, autoimmune diseases, infections, or tumors, requiring a nuanced approach that simultaneously addresses infection and promotes tissue repair. Recent advances in tissue engineering and materials science, particularly in nanomaterials and nano-drug formulations, have led to the development of bifunctional biomaterials with combined osteogenic and antibacterial properties. These materials offer an alternative to traditional bone grafts, minimizing complications such as multiple surgeries, high antibiotic dosages, and lengthy recovery periods. This review examines the repair mechanisms in the infectious microenvironment and highlights various bifunctional biomaterials that foster both anti-infective and osteogenic processes. Emerging design strategies are also discussed to provide a forward-looking perspective on treating infectious bone defects with clinically significant outcomes.
Collapse
Affiliation(s)
- Huaiyuan Zhang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Wenyu Qiao
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yu Liu
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Xizhou Yao
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yonghua Zhai
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Longhai Du
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
3
|
Durán Hernández O, Baumer V, Marrero G, Karumanchi S, Prawel D. Fabrication and Characterization of Highly Porous Gyroid Scaffolds Composed of Deproteinized Bone Mineral. J Funct Biomater 2025; 16:119. [PMID: 40278227 PMCID: PMC12028333 DOI: 10.3390/jfb16040119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/14/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Current treatment methods for critical bone defects involve the implantation of large bone grafts, which are limited by tissue availability and failure to heal correctly with high complication rates. Bioengineered scaffolds have emerged, which deploy biodegradable, highly osteoconductive materials in porous structures to accommodate the high mass transport requirements of large bone defects. Ideal scaffold biomaterials require a balance between strength, composition, and osteoconduction, a balance which has yet to be discovered. Naturally derived materials like deproteinized bovine bone mineral (DBBM) have seen successful clinical use for decades as bone void fillers, but their granular or putty form lacks the interconnected porosity required to treat large defects. Leveraging the clinical success of DBBM, this paper presents the first fabrication of highly porous scaffolds composed of naturally derived, deproteinized bone mineral, for potential use in large bone defects. Ovine bone mineral powder was prepared from fresh ovine bone, fabricated into a photopolymeric slurry and 3D-printed using a photocasting process into 67% porous gyroid scaffolds. Ovine bone mineral composition, surface microstructure, compressive properties, and failure probability were evaluated and compared to gyroid scaffolds composed of tricalcium phosphate. Both scaffold types were similar, with characteristics in the low range of human cancellous bone.
Collapse
Affiliation(s)
- Otoniel Durán Hernández
- School of Materials Science and Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Vail Baumer
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Genesis Marrero
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Sreya Karumanchi
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - David Prawel
- School of Materials Science and Engineering, Colorado State University, Fort Collins, CO 80523, USA;
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
4
|
Hose D, Ray S, Rößler S, Thormann U, Schnettler R, de Veirman K, El Khassawna T, Heiss C, Hild A, Zahner D, Alagboso F, Henss A, Beck S, Emde-Rajaratnam M, Burhenne J, Bamberger J, Menu E, de Bruyne E, Gelinsky M, Kampschulte M, Rohnke M, Wenisch S, Vanderkerken K, Hanke T, Seckinger A, Alt V. Bortezomib-releasing silica-collagen xerogels for local treatment of osteolytic bone- and minimal residual disease in multiple myeloma. J Hematol Oncol 2024; 17:128. [PMID: 39695697 PMCID: PMC11657678 DOI: 10.1186/s13045-024-01636-4] [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: 09/19/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Accumulation of malignant plasma cells in the bone marrow causes lytic bone lesions in 80% of multiple myeloma patients. Frequently fracturing, they are challenging to treat surgically. Myeloma cells surviving treatment in the presumably protective environment of bone lesions impede their healing by continued impact on bone turnover and can explain regular progression of patients without detectable minimal residual disease (MRD). Locally applicable biomaterials could stabilize and foster healing of bone defects, simultaneously delivering anti-cancer compounds at systemically intolerable concentrations, overcoming drug resistance. METHODS We developed silica-collagen xerogels (sicXer) and bortezomib-releasing silica-collagen xerogels (boXer) for local treatment of osteolytic bone disease and MRD. In vitro and in vivo (tissue sections) release of bortezomib was assessed by ultrahigh-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS) and time-of-flight secondary ion mass spectrometry (ToF-SIMS). Material impact on bone formation was assessed in vitro regarding osteoclast/osteoblast numbers and activity. In vivo, drilling defects in a rat- and the 5T33-myeloma mouse model were treated by both materials and assessed by immunohistochemistry, UPLC-MS/MS, µCT, and ToF-SIMS. The material's anti-myeloma activity was assessed using ten human myeloma cell lines (HMCLs) and eight primary myeloma cell samples including four patients refractory to systemic bortezomib treatment. RESULTS sicXer and boXer show primary stability comparable to trabecular bone. Granule size and preparation method tailor degradation as indicated by release of the xerogel components (silica and collagen) and bortezomib into culture medium. In vitro, both materials reduce osteoclast activity and do not negatively interfere with osteoblast differentiation and function. The presumed resulting net bone formation with maintained basic remodeling properties was validated in vivo in a rat bone defect model, showing significantly enhanced bone formation for boXer compared to non-treated defects. Both materials induce myeloma cell apoptosis in all HMCLs and primary myeloma cell samples. In the 5T33-myeloma mouse model, both materials stabilized drilling defects and locally controlled malignant plasma cell growth. CONCLUSIONS The combination of stabilization of fracture-prone lesions, stimulation of bone healing, and anti-tumor effect suggest clinical testing of sicXer and boXer as part of a combined systemic/local treatment strategy in multiple myeloma and non-malignant diseases.
Collapse
Affiliation(s)
- Dirk Hose
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium.
| | - Seemun Ray
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | - Sina Rößler
- Institut für Werkstoffwissenschaft, Max-Bergmann-Zentrum für Biomaterialien, Technische Universität Dresden, Budapester Straße 27, 01069, Dresden, Germany
| | - Ulrich Thormann
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | | | - Kim de Veirman
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Thaqif El Khassawna
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | - Christian Heiss
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | - Anne Hild
- Klinische Anatomie und Experimentelle Chirurgie C/O Institut für Veterinär-Anatomie, -Histologie und -Embryologie, Justus-Liebig-Universität Gießen, Frankfurter Straße 98, 35392, Gießen, Germany
| | - Daniel Zahner
- Justus-Liebig-Universität Gießen, Ludwigstraße 23, 35392, Gießen, Germany
| | - Francisca Alagboso
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany
| | - Anja Henss
- I. Physikalisches Institut, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring 16, 35392, Gießen, Germany
| | - Susanne Beck
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Martina Emde-Rajaratnam
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Jürgen Burhenne
- Innere Medizin IX - Abteilung für Klinische Pharmakologie und Pharmakoepidemiologie, Medizinische Fakultät/Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Juliane Bamberger
- Labor Für Experimentelle Radiologie, Justus-Liebig-Universität Gießen, Carl-Maria-von-Weber-Straße 8, 35392, Gießen, Germany
| | - Eline Menu
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Elke de Bruyne
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Michael Gelinsky
- Zentrum für Translationale Knochen-, Gelenk- und Weichgewebeforschung, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Marian Kampschulte
- Labor Für Experimentelle Radiologie, Justus-Liebig-Universität Gießen, Carl-Maria-von-Weber-Straße 8, 35392, Gießen, Germany
| | - Marcus Rohnke
- Physikalisch-Chemisches Institut, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring 17, 35392, Gießen, Germany
| | - Sabine Wenisch
- Klinische Anatomie und Experimentelle Chirurgie C/O Institut für Veterinär-Anatomie, -Histologie und -Embryologie, Justus-Liebig-Universität Gießen, Frankfurter Straße 98, 35392, Gießen, Germany
| | - Karin Vanderkerken
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Thomas Hanke
- Institut für Werkstoffwissenschaft, Max-Bergmann-Zentrum für Biomaterialien, Technische Universität Dresden, Budapester Straße 27, 01069, Dresden, Germany
| | - Anja Seckinger
- Laboratory of Hematology and Immunology & Labor für Myelomforschung, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Belgium
| | - Volker Alt
- Experimentelle Unfallchirurgie (ForMED), Justus-Liebig-Universität Gießen, Aulweg 128, 35392, Gießen, Germany.
- Klinik und Poliklinik für Unfallchirurgie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
5
|
Safarova (Yantsen) Y, Nessipbekova A, Syzdykova A, Olzhayev F, Umbayev B, Kassenova A, Fadeeva IV, Askarova S, Rau JV. Strontium- and Copper-Doped Ceramic Granules in Bone Regeneration-Associated Cellular Processes. J Funct Biomater 2024; 15:352. [PMID: 39590555 PMCID: PMC11595051 DOI: 10.3390/jfb15110352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Pathological bone fracturing is an escalating problem driven by increasing aging and obesity. Bioceramics, particularly tricalcium-phosphate-based materials (TCP), are renowned for their exceptional biocompatibility, osteoconductivity, and ability to promote biomineralization. In the present study, we designed and characterized TCP porous granules doped with strontium (Sr) and copper (Cu) (CuSr TCP). Sr2+ ions were selected as Sr plays a crucial role in early bone formation, osteogenesis, and angiogenesis; Cu2+ ions possess antibacterial properties. MATERIALS The synthesized CuSr TCP granules were characterized by X-ray diffraction. Cytotoxicity and cell proliferation analyses' assays were performed through the lactate dehydrogenase (LDH) activity and CCK-8 viability tests in rat bone marrow-derived mesenchymal stem cells (BM-MSCs). Hemolytic activity was carried out with human red blood cells (RBCs). Early and late osteogenesis were assessed with alkaline phosphatase (ALP) and Alizarin Red S activity in human osteoblast progenitor cells and rat BM-MSCs. The influence of CuSr TCP on angiogenesis was investigated in human umbilical vein endothelial cells (HUVECs). RESULTS We have demonstrated that media enriched with CuSr TCP in concentrations ranging from 0.1 mg/mL to 1 mg/mL were not cytotoxic and did not significantly affect cell proliferation rate motility. Moreover, a concentration of 0.5 mg/mL showed a 2.5-fold increase in the migration potential of BM-MSCs. We also found that CuSr TCP-enriched media slightly increased early osteogenesis. We also found that Sr and Cu substitutions in TCP particles significantly enhanced the measured angiogenic parameters compared to control and unsubstituted TCP granules. CONCLUSION Our results demonstrate that TCP porous granules doped with Sr and Cu are biocompatible, promote osteodifferentiation and angiogenesis, and could be recommended for further in vivo studies.
Collapse
Affiliation(s)
- Yuliya Safarova (Yantsen)
- National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave. 53, 010000 Astana, Kazakhstan; (A.N.); (A.S.); (F.O.); (B.U.); (A.K.); (S.A.)
| | - Assem Nessipbekova
- National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave. 53, 010000 Astana, Kazakhstan; (A.N.); (A.S.); (F.O.); (B.U.); (A.K.); (S.A.)
| | - Aizhan Syzdykova
- National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave. 53, 010000 Astana, Kazakhstan; (A.N.); (A.S.); (F.O.); (B.U.); (A.K.); (S.A.)
| | - Farkhad Olzhayev
- National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave. 53, 010000 Astana, Kazakhstan; (A.N.); (A.S.); (F.O.); (B.U.); (A.K.); (S.A.)
| | - Bauyrzhan Umbayev
- National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave. 53, 010000 Astana, Kazakhstan; (A.N.); (A.S.); (F.O.); (B.U.); (A.K.); (S.A.)
| | - Aliya Kassenova
- National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave. 53, 010000 Astana, Kazakhstan; (A.N.); (A.S.); (F.O.); (B.U.); (A.K.); (S.A.)
| | - Inna V. Fadeeva
- A. A. Baikov Institute of Metallurgy and Material Science RAS, Leninskie, 49, 119334 Moscow, Russia;
| | - Sholpan Askarova
- National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave. 53, 010000 Astana, Kazakhstan; (A.N.); (A.S.); (F.O.); (B.U.); (A.K.); (S.A.)
| | - Julietta V. Rau
- Instituto di Struttura della Materia, Consiglio Nazionale delle Ricerche, ISM-CNR, Via del Fosso del Cavaliere 100, 00133 Rome, Italy;
- Department of Analytical, Physical and Colloid Chemistry, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Trubetskaya 8, Build. 2, 119048 Moscow, Russia
| |
Collapse
|
6
|
Shahbazi M, Jäger H, Ettelaie R, Chen J, Kashi PA, Mohammadi A. Dispersion strategies of nanomaterials in polymeric inks for efficient 3D printing of soft and smart 3D structures: A systematic review. Adv Colloid Interface Sci 2024; 333:103285. [PMID: 39216400 DOI: 10.1016/j.cis.2024.103285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/03/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Nanoscience-often summarized as "the future is tiny"-highlights the work of researchers advancing nanotechnology through incremental innovations. The design and innovation of new nanomaterials are vital for the development of next-generation three-dimensional (3D) printed structures characterized by low cost, high speed, and versatile capabilities, delivering exceptional performance in advanced applications. The integration of nanofillers into polymeric-based inks for 3D printing heralds a new era in additive manufacturing, allowing for the creation of custom-designed 3D objects with enhanced multifunctionality. To optimize the use of nanomaterials in 3D printing, effective disaggregation techniques and strong interfacial adhesion between nanofillers and polymer matrices are essential. This review provides an overview of the application of various types of nanomaterials used in 3D printing, focusing on their functionalization principles, dispersion strategies, and colloidal stability, as well as the methodologies for aligning nanofillers within the 3D printing framework. It discusses dispersive methods, synergistic dispersion, and in-situ growth, which have yielded smart 3D-printed structures with unique functionality for specific applications. This review also focuses on nanomaterial alignment in 3D printing, detailing methods that enhance selective deposition and orientation of nanofillers within established and customized printing techniques. By emphasizing alignment strategies, we explore their impact on the performance of 3D-printed composites and highlight potential applications that benefit from ordered nanoparticles. Through these continuing efforts, this review shows that the design and development of the new class of nanomaterials are crucial to developing the next generation of smart 3D printed architectures with versatile abilities for advanced structures with exceptional performance.
Collapse
Affiliation(s)
- Mahdiyar Shahbazi
- Institute of Material Technology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria.
| | - Henry Jäger
- Institute of Material Technology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria.
| | - Rammile Ettelaie
- Food Colloids and Bioprocessing Group, School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| | - Jianshe Chen
- Food Oral Processing Laboratory, School of Food Science & Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Peyman Asghartabar Kashi
- Faculty of Biosystem, College of Agricultural and Natural Resources Tehran University, Tehran, Iran
| | - Adeleh Mohammadi
- Department of Chemistry, University Hamburg, Institute of Food Chemistry, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| |
Collapse
|
7
|
Saito K, Inagaki Y, Uchihara Y, Okamoto M, Nishimura Y, Kawai A, Sugino T, Okamura K, Ogawa M, Kido A, Tanaka Y. MgO-enhanced β-TCP promotes osteogenesis in both in vitro and in vivo rat models. Sci Rep 2024; 14:19725. [PMID: 39183238 PMCID: PMC11345426 DOI: 10.1038/s41598-024-70512-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
Allogeneic bone grafts are used to treat bone defects in orthopedic surgery, but the osteogenic potential of artificial bones remains a challenge. In this study, we developed a β-tricalcium phosphate (β-TCP) formulation containing MgO, ZnO, SrO, and SiO2 and compared its bone-forming ability with that of β-TCP without biological elements. We prepared β-TCP discs with 60% porosity containing 1.0 wt% of these biological elements. β-TCP scaffolds were loaded with bone marrow-derived mesenchymal stem cells (BMSC) from 7-week-old male rats and cultured for 2 weeks. ALP activity and mRNA expression of osteogenic markers were evaluated. In addition, scaffolds were implanted subcutaneously in rats and analyzed after 7 weeks. In vitro, the MgO group showed lower Ca concentrations and higher osteogenic marker expression compared to controls. In vivo, the MgO group showed higher ALP activity compared to controls, and RT-qPCR analysis showed significant expression of BMP2 and VEGF. Histopathology, fluorescent immunostaining, and micro-CT also showed relatively better bone formation in the MgO group. β-TCP with MgO may enhance bone morphology in vitro and in vivo and improve the prognosis of patients with substantial and refractory bone defects.
Collapse
Affiliation(s)
- Kenichiro Saito
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Yusuke Inagaki
- Department of Rehabilitation Medicine, Nara Medical University, Kashihara, Nara, Japan.
| | - Yoshinobu Uchihara
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Masakazu Okamoto
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Yuki Nishimura
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Akihito Kawai
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Tatsuro Sugino
- Product Development Department, Olympus Terumo Biomaterials Corp., Shizuoka, Japan
| | - Kensuke Okamura
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Munehiro Ogawa
- Department of Sports Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Akira Kido
- Department of Rehabilitation Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Yasuhito Tanaka
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
8
|
Naguib G, Mously H, Mazhar J, Alkanfari I, Binmahfooz A, Zahran M, Hamed MT. Bond strength and surface roughness assessment of novel antimicrobial polymeric coated dental cement. DISCOVER NANO 2024; 19:123. [PMID: 39105979 PMCID: PMC11303365 DOI: 10.1186/s11671-024-04074-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 07/27/2024] [Indexed: 08/07/2024]
Abstract
Resin cement integrated with zein-incorporated magnesium oxide nanoparticles has previously been found to inhibit oral microbes and decrease bacterial biofilm. However, the bond strength and surface features of this biomaterial have yet to be investigated. The objective of this study was to evaluate the shear bond strength, mode of fracture, and surface roughness of resin cement modified with zein-incorporated magnesium oxide nanoparticles. Characterization of the cement was performed by X-ray diffraction, field emission scanning electron microscopy, and Fourier transform infrared spectroscopy. 126 human teeth were divided into 3 groups and cemented to lithium disilicate ceramic using resin cement with zein-incorporated magnesium oxide nanoparticles at concentrations of 0%, 1%, and 2% (n = 42). 21 samples of each group were subjected to the shear bond strength test, while the other 21 underwent thermocycling for 10,000 cycles before the test, after which all samples were evaluated for the mode of fracture. To assess surface roughness, resin cement disks were analyzed by a profilometer before and after undergoing thermocycling for 10,000 cycles. The shear bond strength of the cement with 1% and 2% nanoparticles was significantly higher than the control before thermocycling. The mode of fracture was found to be mainly adhesive with all groups, with the unmodified cement presenting the highest cohesive failure. There was no significant difference in surface roughness between the groups before or after thermocycling. The addition of zein-incorporated magnesium oxide nanoparticles to resin cement improved or maintained the shear bond strength and surface roughness of the resin cement.
Collapse
Affiliation(s)
- Ghada Naguib
- Department of Restorative Dentistry, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia.
- Department of Oral Biology, Cairo University School of Dentistry, Cairo, Egypt.
| | - Hisham Mously
- Department of Oral and Maxillofacial Prosthodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Ibrahim Alkanfari
- Department of Oral and Maxillofacial Prosthodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulelah Binmahfooz
- Department of Oral and Maxillofacial Prosthodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Zahran
- Department of Oral and Maxillofacial Prosthodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed T Hamed
- Department of Oral and Maxillofacial Prosthodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Fixed Prosthodontics, Cairo University School of Dentistry, Cairo, Egypt
| |
Collapse
|
9
|
Yang M, Cai X, Wang C, Wang Z, Xue F, Chu C, Bai J, Liu Q, Ni X. Highly Stable Amorphous (Pyro)phosphate Aggregates: Pyrophosphate as a Carrier for Bioactive Ions and Drugs in Bone Repair Applications. ACS OMEGA 2024; 9:23724-23740. [PMID: 38854518 PMCID: PMC11154929 DOI: 10.1021/acsomega.4c01660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Pyrophosphate is widely used as an iron supplement because of its excellent complexation and hydrolysis ability; however, there are few reports on the use of pyrophosphate in active ionophores for bone repair. In this research, we proposed a simple and efficient ultrasonic method to prepare magnesium-calcium (pyro)phosphate aggregates (AMCPs). Due to strong hydration, AMCPs maintain a stable amorphous form even at high temperatures (400 °C). By changing the molar ratio of calcium and magnesium ions, the content of calcium and magnesium ions can be customized. AMCPs had surface negativity and complexing ability that realized the controlled release of ions (Ca2+, Mg2+, and P) and drugs (such as doxorubicin) over a long period. Pyrophosphate gave it an excellent bacteriostatic effect. Increasingly released Mg2+ exhibited improved bioactivity though the content of Ca2+ decreased. While Mg2+ content was regulated to 15 wt %, it performed significantly enhanced stimulation on the proliferation, attachment, and differentiation (ALP activity, calcium nodules, and the related gene expression of osteogenesis) of mouse embryo osteoblast precursor cells (MC3T3-E1). Furthermore, the high content of Mg2+ also effectively promoted the proliferation, attachment, and migration of human umbilical vein endothelial cells (HUVECs) and the expression of angiogenic genes. In conclusion, pyrophosphate was an excellent carrier for bioactive ions, and the AMCPs we prepared had a variety of active functions for multiscenario bone repair applications.
Collapse
Affiliation(s)
- Mengmeng Yang
- School of Materials Science and Engineering, Southeast University, Nanjing, 211189 Jiangsu, China
- Center of Medical Physics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003 Jiangsu, China
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166 Jiangsu, China
| | - Xiang Cai
- School of Materials Science and Engineering, Southeast University, Nanjing, 211189 Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Nanjing, 211189 Jiangsu, China
| | - Cheng Wang
- School of Materials Science and Engineering, Southeast University, Nanjing, 211189 Jiangsu, China
| | - Zan Wang
- School of Materials Science and Engineering, Southeast University, Nanjing, 211189 Jiangsu, China
| | - Feng Xue
- School of Materials Science and Engineering, Southeast University, Nanjing, 211189 Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Nanjing, 211189 Jiangsu, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
| | - Chenglin Chu
- School of Materials Science and Engineering, Southeast University, Nanjing, 211189 Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Nanjing, 211189 Jiangsu, China
| | - Jing Bai
- School of Materials Science and Engineering, Southeast University, Nanjing, 211189 Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Nanjing, 211189 Jiangsu, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
- Jiangsu Key Laboratory for Light Metal Alloys, Nanjing 211212, China
| | - Qizhan Liu
- School of Materials Science and Engineering, Southeast University, Nanjing, 211189 Jiangsu, China
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166 Jiangsu, China
| | - Xinye Ni
- Center of Medical Physics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003 Jiangsu, China
| |
Collapse
|
10
|
Deng F, Han X, Ji Y, Jin Y, Shao Y, Zhang J, Ning C. Distinct mechanisms of iron and zinc metal ions on osteo-immunomodulation of silicocarnotite bioceramics. Mater Today Bio 2024; 26:101086. [PMID: 38765245 PMCID: PMC11098954 DOI: 10.1016/j.mtbio.2024.101086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/14/2024] [Accepted: 05/04/2024] [Indexed: 05/21/2024] Open
Abstract
The immunomodulatory of implants have drawn more and more attention these years. However, the immunomodulatory of different elements on the same biomaterials have been rarely investigated. In this work, two widely used biosafety elements, iron and zinc added silicocarnotite (Ca5(PO4)2SiO4, CPS) were applied to explore the routine of elements on immune response. The immune reactions over time of Fe-CPS and Zn-CPS were explored at genetic level and protein level, and the effects of their immune microenvironment with different time points on osteogenesis were also investigated in depth. The results confirmed that both Fe-CPS and Zn-CPS had favorable ability to secret anti-inflammatory cytokines. The immune microenvironment of Fe-CPS and Zn-CPS also could accelerate osteogenesis and osteogenic differentiation in vitro and in vivo. In terms of mechanism, RNA-seq analysis and Western-blot experiment revealed that PI3K-Akt signaling pathway and JAK-STAT signaling pathways were activated of Fe-CPS to promote macrophage polarization from M1 to M2, and its immune microenvironment induced osteogenic differentiation through the activation of Hippo signaling pathway. In comparison, Zn-CPS inhibited polarization of M1 macrophage via the up-regulation of Rap1 signaling pathway and complement and coagulation cascade pathway, while its osteogenic differentiation related pathway of immune environment was NF-κB signaling pathway.
Collapse
Affiliation(s)
- Fanyan Deng
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Frontiers Science Center of Biomimetic Catalysis and Shanghai Engineering Research Center of Green Energy Chemical Engineering, Shanghai Normal University, Shanghai, China
| | - Xianzhuo Han
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China
| | - Yingqi Ji
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Ying Jin
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Frontiers Science Center of Biomimetic Catalysis and Shanghai Engineering Research Center of Green Energy Chemical Engineering, Shanghai Normal University, Shanghai, China
| | - Yiran Shao
- SHNU-YAPENG Joint Lab of Tissue Repair Materials, Shanghai Yapeng Biological Technology Co., Ltd, Shanghai, China
| | - Jingju Zhang
- Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Department of Orthodontics, School & Hospital of Stomatology, Shanghai, China
| | - Congqin Ning
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Frontiers Science Center of Biomimetic Catalysis and Shanghai Engineering Research Center of Green Energy Chemical Engineering, Shanghai Normal University, Shanghai, China
| |
Collapse
|
11
|
Rstakyan V, Mkhitaryan L, Baghdasaryan L, Ghaltaghchyan T, Karabekian Z, Sevoyan G, Aghayan M, Rodríguez MA. Stereolithography of ceramic scaffolds for bone tissue regeneration: Influence of hydroxyapatite/silica ratio on mechanical properties. J Mech Behav Biomed Mater 2024; 152:106421. [PMID: 38280269 DOI: 10.1016/j.jmbbm.2024.106421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
In this paper, the results obtained in the development of ceramic resin feedstock for stereolithography are shown. Hydroxyapatite and silica are used as source of ceramic. Hydroxyapatite is extracted from bovine bone, which enhances bioactivity of ceramic scaffold. The influence of hydroxyapatite amount in polymer-based slurry on the viscosity and printability of feedstock is explored. Hydroxyapatite and silica containing scaffolds are successfully obtained by stereolithography. Influence of hydroxyapatite/silica ratio on the bioactivity, biodegradability and mechanical properties of the scaffolds is also studied. It was observed that higher concentrations of hydroxyapatite led to improved mechanical strength of the scuffolds but increased viscosity of the slurry, affecting printability. Cell viability assays and cell visualization experiments indicated that the scaffolds not cause significant cell toxicity.
Collapse
Affiliation(s)
- Viktorya Rstakyan
- A.B. Nalbandyan Institute of Chemical Physics, National Academy of Sciences of the Republic of Armenia, P. Sevak 5/2, Yerevan, 0014, Armenia.
| | - Liana Mkhitaryan
- A.B. Nalbandyan Institute of Chemical Physics, National Academy of Sciences of the Republic of Armenia, P. Sevak 5/2, Yerevan, 0014, Armenia
| | - Lilit Baghdasaryan
- A.B. Nalbandyan Institute of Chemical Physics, National Academy of Sciences of the Republic of Armenia, P. Sevak 5/2, Yerevan, 0014, Armenia
| | - Tsovinar Ghaltaghchyan
- A.B. Nalbandyan Institute of Chemical Physics, National Academy of Sciences of the Republic of Armenia, P. Sevak 5/2, Yerevan, 0014, Armenia
| | - Zaruhi Karabekian
- Orbeli Institute of Physiology, National Academy of Sciences of the Republic of Armenia, Orbeli Bros. 22, Yerevan, 0028, Armenia
| | - Gohar Sevoyan
- Orbeli Institute of Physiology, National Academy of Sciences of the Republic of Armenia, Orbeli Bros. 22, Yerevan, 0028, Armenia
| | - Marina Aghayan
- A.B. Nalbandyan Institute of Chemical Physics, National Academy of Sciences of the Republic of Armenia, P. Sevak 5/2, Yerevan, 0014, Armenia; FACT Industries, Õismäe tee 124, 13513, Tallinn, Estonia
| | - Miguel A Rodríguez
- Instituto de Cerámica y Vidrio (CSIC), C/ Kelsen 5, 28904, Madrid, Spain
| |
Collapse
|
12
|
Qi L, Zhao T, Yan J, Ge W, Jiang W, Wang J, Gholipourmalekabadi M, Lin K, Wang X, Zhang L. Advances in magnesium-containing bioceramics for bone repair. BIOMATERIALS TRANSLATIONAL 2024; 5:3-20. [PMID: 39220661 PMCID: PMC11362349 DOI: 10.12336/biomatertransl.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/21/2024] [Accepted: 03/23/2024] [Indexed: 09/04/2024]
Abstract
Reconstruction of bone defects or fractures caused by ageing, trauma and tumour resection is still a great challenge in clinical treatment. Although autologous bone graft is considered as gold standard, the source of natural bone is limited. In recent years, regenerative therapy based on bioactive materials has been proposed for bone reconstruction. Specially, numerous studies have indicated that bioactive ceramics including silicate and phosphate bioceramics exhibit excellent osteoinductivity and osteoconductivity, further promote bone regeneration. In addition, magnesium (Mg) element, as an indispensable mineral element, plays a vital role in promoting bone mineralisation and formation. In this review, different types of Mg-containing bioceramics including Mg-containing calcium phosphate-based bioceramics (such as Mg-hydroxyapatite, Mg-biphasic calcium phosphate), Mg-containing calcium silicate-based bioceramics (such as Mg2SiO4, Ca2MgSi2O7 and Mg-doped bioglass), Mg-based biocements, Mg-containing metal/polymer-bioceramic composites were systematacially summarised. Additionally, the fabrication technologies and their materiobiological effects were deeply discussed. Clinical applications and perspectives of magnesium-containing bioceramics for bone repair are highlighted. Overall, Mg-containing bioceramics are regarded as regenerative therapy with their optimised performance. Furthermore, more in-depth two-way researches on their performance and structure are essential to satisfy their clinical needs.
Collapse
Affiliation(s)
- Lei Qi
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Tong Zhao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jinge Yan
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Weiwen Ge
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Weidong Jiang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jing Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Mazaher Gholipourmalekabadi
- Department’s Education Deputy, Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| | - Kaili Lin
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xiuhui Wang
- Institute of Translational Medicine, Shanghai University; Organoid Research Center, Shanghai University, Shanghai, China
| | - Lei Zhang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
13
|
Szczodra A, Houaoui A, Salminen T, Hannula M, Gobbo VA, Ghanavati S, Miettinen S, Massera J. Pore graded borosilicate bioactive glass scaffolds: in vitro dissolution and cytocompatibility. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:17. [PMID: 38507150 PMCID: PMC10954867 DOI: 10.1007/s10856-024-06791-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 03/05/2024] [Indexed: 03/22/2024]
Abstract
3D borosilicate bioactive glass (1393B20 and B12.5MgSr) scaffolds were prepared by robocasting, with and without a dense layer at the top. Pore graded scaffolds are promising as they allow for membrane deposition and could limit the risk of soft tissue infiltration. In vitro dissolution was studied in tris(hydroxymethyl)aminomethane (TRIS) and Simulated Body Fluid (SBF). 1393B20 scaffolds dissolved faster than B12.5MgSr in TRIS whereas they dissolved slower in SBF. The difference in dissolution profiles, as a function of the medium used, is assigned to the different rates of precipitation of hydroxyapatite (HA). While the precipitation of calcium phosphate (CaP) in the form of HA, first sign of bioactivity, was confirmed by ICP, FTIR-ATR and SEM-EDX analysis for both compositions, 1393B20 was found to precipitate HA at a faster rate. The presence of a dense top layer did not significantly impact the dissolution rate and CaP precipitation. In vitro cell culture was performed using human adipose-derived stem cells (hADSCs). Prior to cell plating, a preincubation of 3 days was found optimum to prevent burst ion release. In direct contact, cells proliferate and spread on the scaffolds while maintaining characteristic spindle morphology. Cell plated on 1393B20 scaffolds showed increased viability when compared to cell plated on B12.5MgSr. The lower cell viability, when testing B12.5MgSr, was assigned to the depletion of Ca2+ ions from culture medium and higher pH. Static cell culture leads to believe that the scaffold produced from the 1393B20 glass composition are promising in bone regeneration applications.
Collapse
Affiliation(s)
- Agata Szczodra
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland.
| | - Amel Houaoui
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Turkka Salminen
- Tampere University, Faculty of Engineering and Natural Sciences, Tampere, Finland
| | - Markus Hannula
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
| | | | - Sonya Ghanavati
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Susanna Miettinen
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
- Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Jonathan Massera
- Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
| |
Collapse
|
14
|
Jo Y, Majumdar U, Bose S. Vitamin D3 Release from MgO Doped 3D Printed TCP Scaffolds for Bone Regeneration. ACS Biomater Sci Eng 2024; 10:1676-1685. [PMID: 38386843 PMCID: PMC11186521 DOI: 10.1021/acsbiomaterials.3c01779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Regenerating bone tissue in critical-sized craniofacial bone defects remains challenging and requires the implementation of innovative bone implants with early stage osteogenesis and blood vessel formation. Vitamin D3 is incorporated into MgO-doped 3D-printed scaffolds for defect-specific and patient-specific implants in low load-bearing areas. This novel bone implant also promotes early stage osteogenesis and blood vessel development. Our results show that vitamin D3-loaded MgO-doped 3D-printed scaffolds enhance osteoblast cell proliferation 1.3-fold after being cultured for 7 days. Coculture studies on osteoblasts derived from human mesenchymal stem cells (hMSCs) and osteoclasts derived from monocytes show the upregulation of genes related to osteoblastogenesis and the downregulation of RANK-L, which is essential for osteoclastogenesis. Release of vitamin D3 also inhibits osteoclast differentiation by 1.9-fold after a 21-day culture. After 6 weeks, vitamin D3 release from MgO-doped 3D-printed scaffolds enhances the new bone formation, mineralization, and angiogenic potential. The multifunctional 3D-printed scaffolds can improve early stage osteogenesis and blood vessel formation in craniofacial bone defects.
Collapse
Affiliation(s)
- Yongdeok Jo
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Ujjayan Majumdar
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| |
Collapse
|
15
|
Wu J, Cheng X, Wu J, Chen J, Pei X. The development of magnesium-based biomaterials in bone tissue engineering: A review. J Biomed Mater Res B Appl Biomater 2024; 112:e35326. [PMID: 37861271 DOI: 10.1002/jbm.b.35326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/15/2023] [Accepted: 08/23/2023] [Indexed: 10/21/2023]
Abstract
Bone regeneration is a vital clinical challenge in massive or complicated bone defects. Recently, bone tissue engineering has come to the fore to meet the demand for bone repair with various innovative materials. However, the reported materials usually cannot satisfy the requirements, such as ideal mechanical and osteogenic properties, as well as biocompatibility at the same time. Mg-based biomaterials have considerable potential in bone tissue engineering owing to their excellent mechanical strength and biosafety. Moreover, the biocompatibility and osteogenic activity of Mg-based biomaterials have been the research focuses in recent years. The main limitation faced in the applications of Mg-based biomaterials is rapid degradation, which can produce excessive Mg2+ and hydrogen, affecting the healing of the bone defect. In order to overcome the limitations, researchers have explored several ways to improve the properties of Mg-based biomaterials, including alloying, surface modification with coatings, and synthesizing other composite materials to control the degradation rate upon implantation. This article reviewed the osteogenic mechanism and requirement for appropriate degradation rate and focused on current progress in the biomedical use of Mg-based biomaterials to inspire more clinical applications of Mg in bone regeneration in the future.
Collapse
Affiliation(s)
- Jiaxin Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinting Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jicenyuan Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
16
|
Soares Í, Sotelo L, Erceg I, Jean F, Lasgorceix M, Leriche A, Sikirić MD, Marušić K, Christiansen S, Daskalova A. Improvement of Metal-Doped β-TCP Scaffolds for Active Bone Substitutes via Ultra-Short Laser Structuring. Bioengineering (Basel) 2023; 10:1392. [PMID: 38135983 PMCID: PMC10741177 DOI: 10.3390/bioengineering10121392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 12/24/2023] Open
Abstract
Various efforts have been made to develop antibacterial biomaterials capable of also sustaining bone remodulation to be used as bone substitutes and reduce patient infection rates and related costs. In this work, beta-tricalcium phosphate (β-TCP) was chosen due to its known biocompatibility and use as a bone substitute. Metal dopants were incorporated into the crystal structure of the β-TCP, and disks were produced from this material. Magnesium and strontium, as well as copper and silver, were chosen as dopants to improve the osteogenic and antibacterial properties, respectively. The surface of the β-TCP samples was further modified using a femtosecond laser system. Grid and line patterns were produced on the plates' surface via laser ablation, creating grooves with depths lower than 20 μm and widths between 20 and 40 μm. Raman and FTIR analysis confirmed that laser ablation did not result in the degradation or phase change of the materials, making it suitable for surface patterning. Laser ablation resulted in increased hydrophilicity of the materials, as the control samples (non-ablated samples) have WCA values ranging from 70° to 93° and become, upon laser ablation, superwicking surfaces. Confocal measurements show an increase in specific surface area of 50% to 200% compared to the control. Overall, the results indicate the potential of laser ablation to improve the surface characteristics of β-TCP, which may lead to an improvement in the antibacterial and osteogenic properties of the produced materials.
Collapse
Affiliation(s)
- Íris Soares
- Laboratory of Micro and Nano-Photonics, Institute of Electronics, Bulgarian Academy of Sciences, 72 Tsarigradsko Chaussee Blvd, 1784 Sofia, Bulgaria
| | - Lamborghini Sotelo
- Institute for Nanotechnology and Correlative Microscopy vV INAM, Äußere Nürnberger Str. 62, 91301 Forcheim, Germany; (L.S.); (S.C.)
- Friedrich-Alexander University Erlangen-Nürnberg, Staudstraße 7, 91058 Erlangen, Germany
| | - Ina Erceg
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Äußere Nürnberger Str. 62, 91301 Forcheim, Germany;
| | - Florian Jean
- University Polytechnique Hauts-de-France, INSA Hauts-de-France, CERAMATHS—Laboratoire de Matériaux Céramiques et de Mathématiques, F-59313 Valenciennes, France; (F.J.); (M.L.); (A.L.)
| | - Marie Lasgorceix
- University Polytechnique Hauts-de-France, INSA Hauts-de-France, CERAMATHS—Laboratoire de Matériaux Céramiques et de Mathématiques, F-59313 Valenciennes, France; (F.J.); (M.L.); (A.L.)
| | - Anne Leriche
- University Polytechnique Hauts-de-France, INSA Hauts-de-France, CERAMATHS—Laboratoire de Matériaux Céramiques et de Mathématiques, F-59313 Valenciennes, France; (F.J.); (M.L.); (A.L.)
| | - Maja Dutour Sikirić
- Laboratory for Biocolloids and Surface Chemistry, Division of Physical Chemistry, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| | - Katarina Marušić
- Radiation Chemistry and Dosimetry Laboratory, Division of Materials Chemistry, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia;
| | - Silke Christiansen
- Institute for Nanotechnology and Correlative Microscopy vV INAM, Äußere Nürnberger Str. 62, 91301 Forcheim, Germany; (L.S.); (S.C.)
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, Äußere Nürnberger Str. 62, 91301 Forcheim, Germany;
- Frei Iniverssität Berlin, Arnimalle 14, 14995 Berlin, Germany
| | - Albena Daskalova
- Laboratory of Micro and Nano-Photonics, Institute of Electronics, Bulgarian Academy of Sciences, 72 Tsarigradsko Chaussee Blvd, 1784 Sofia, Bulgaria
| |
Collapse
|
17
|
Bhattacharjee A, Bose S. Multifunctional polydopamine - Zn 2+-curcumin coated additively manufactured ceramic bone grafts with enhanced biological properties. BIOMATERIALS ADVANCES 2023; 153:213487. [PMID: 37400297 PMCID: PMC10699649 DOI: 10.1016/j.bioadv.2023.213487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/08/2023] [Accepted: 05/27/2023] [Indexed: 07/05/2023]
Abstract
The lack of site-specific chemotherapeutic agents after osteosarcoma surgeries often induces severe side effects. We propose the utilization of curcumin as an alternative natural chemo-preventive drug for tumor-specific delivery systems with 3D printed tricalcium phosphate (TCP) based artificial bone grafts. The poor bioavailability and hydrophobic nature of curcumin restrict its clinical use. We have used polydopamine (PDA) coating with Zn2+ functionalization to enhance the curcumin release in the biological medium. The obtained PDA-Zn2+ complex is characterized by X-ray photoelectron spectroscopy (XPS). The presence of PDA-Zn2+ coating leads to ~2 times enhancement in curcumin release. We have computationally predicted and validated the optimized surface composition by a novel multi-objective optimization method. The experimental validation of the predicted compositions indicates that the PDA-Zn2+ coated curcumin immobilized delivery system leads to a ~12 folds decrease in osteosarcoma viability on day 11 as compared to only TCP. The osteoblast viability shows ~1.4 folds enhancement. The designed surface shows the highest ~90 % antibacterial efficacy against gram-positive and gram-negative bacteria. This unique strategy of curcumin delivery with PDA-Zn2+ coating is expected to find application in low-load bearing critical-sized tumor-resection sites.
Collapse
Affiliation(s)
- Arjak Bhattacharjee
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, USA
| | - Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
18
|
Naguib GH, Abd El-Aziz GS, Almehmadi A, Bayoumi A, Mira AI, Hassan AH, Hamed MT. Evaluation of the time-dependent osteogenic activity of glycerol incorporated magnesium oxide nanoparticles in induced calvarial defects. Heliyon 2023; 9:e18757. [PMID: 37593643 PMCID: PMC10432181 DOI: 10.1016/j.heliyon.2023.e18757] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/06/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
Introduction Magnesium-based biomaterials have been explored for their potential as bone healing materials, as a result of their outstanding biodegradability and biocompatibility. These characteristics make magnesium oxide nanoparticles (MgO NPs) a promising material for treating bone disorders. The purpose of this investigation is to assess the osteogenic activity of newly-developed locally administered glycerol-incorporated MgO NPs (GIMgO NPs) in rabbits' calvarial defects. Materials and methods Characterization of GIMgO was done by X-ray Diffraction (XRD) and Fourier Transform Infrared Spectroscopy (FTIR). Bilateral calvarial defects were created in eighteen New Zealand Rabbits, of which they were divided into 3 groups with time points corresponding to 2, 4, and 6 weeks postoperatively (n = 6). One defect was implanted with absorbable gel foam impregnated with GIMgO NPs while the other was implanted with gel foam soaked with glycerol (the control). The defects were assessed using histological, Micro-Computed Tomography (Micro-CT), and histometric evaluation. Results The characterization of the GIMgO nanogel revealed the presence of MgO NPs and glycerol as well as the formation of the crystalline phase of the MgO NPs within the nanogel sample. The histological and micro-CT analysis showed time-dependent improvement of healing activity in the calvarial defects implanted with GIMgO NPs when compared to the control. Furthermore, the histometric analysis demonstrated a marked increase in the total area of new bone, connective tissue, new bone area and volume in the GIMgO NPs implanted site. Statistically, the amount of new bone formation was more significant at 6 weeks than at 2 and 4 weeks postoperatively in the calvarial defects implanted with GIMgO NPs as compared to the control. Conclusion The locally applied GIMgO NPs demonstrated efficacy in promoting bone formation, with more significant effects observed over an extended period. These findings suggest its suitability for clinical use as a therapeutic alternative to enhance bone healing.
Collapse
Affiliation(s)
- Ghada H. Naguib
- Department of Restorative Dentistry, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Oral Biology, Cairo University School of Dentistry, Cairo, Egypt
| | - Gamal S. Abd El-Aziz
- Department of Clinical Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmad Almehmadi
- Department of Oral Biology, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amr Bayoumi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulghani I. Mira
- Department of Restorative Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mohamed T. Hamed
- Department of Oral and Maxillofacial Prosthodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Fixed Prosthodontics, Cairo University School of Dentistry, Cairo, Egypt
| |
Collapse
|
19
|
Jo Y, Sarkar N, Bose S. In vitro biological evaluation of epigallocatechin gallate (EGCG) release from three-dimensional printed (3DP) calcium phosphate bone scaffolds. J Mater Chem B 2023; 11:5503-5513. [PMID: 36637404 PMCID: PMC11132590 DOI: 10.1039/d2tb02210a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Three-dimensional printed (3DP) tricalcium phosphate (TCP) scaffolds can guide bone regeneration, especially for patient-specific defect repair applications in low-load bearing sites. Epigallocatechin gallate (EGCG), a green tea compound, has gained attention as a safer alternative treatment for bone disorders. The 3DP TCP scaffold is designed for localized EGCG delivery, which can enhance in vitro osteogenic ability, anti-osteoclastogenic activity, vascularization formation, and chemoprevention. In the cocultures of human bone marrow-derived mesenchymal stem cells (hMSCs) and monocytes (THP-1), EGCG release enhances osteogenic differentiation of hMSCs at day 16 compared to the control; this is indicated by a 2.8- and 4.0-fold upregulation of Runt-related transcription factor 2 (Runx2) and bone gamma-carboxyglutamic acid-containing protein (BGLAP), the early and late osteoblast differentiation marker expressions. However, EGCG significantly downregulates the receptor activator of nuclear factor-κB ligand (RANKL) expression by 7.0-fold, indicating that EGCG suppresses RANKL-induced osteoclast maturation. EGCG also stimulates endothelial tube formation at as early as 3 hours when human umbilical vein endothelial cells (HUVECs) grow on Matrigel. It reduces human osteosarcoma MG-63 cell viability by 66% compared to the control at day 11. An in vitro release kinetics study demonstrates that EGCG shows a ∼64% release within a day followed by a sustained release in the physiological environment (pH 7.4) because its phenolic hydroxyl groups are easily deprotonated at physiological pH. These findings contribute to developing a multifunctional scaffold for the treatment of low load-bearing patient-specific bone defects after trauma and tumor excision.
Collapse
Affiliation(s)
- Yongdeok Jo
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, USA.
| | - Naboneeta Sarkar
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, USA.
| | - Susmita Bose
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, USA.
| |
Collapse
|
20
|
Amaral SS, Lima BSDS, Avelino SOM, Spirandeli BR, Campos TMB, Thim GP, Trichês EDS, Prado RFD, Vasconcellos LMRD. β-TCP/S53P4 Scaffolds Obtained by Gel Casting: Synthesis, Properties, and Biomedical Applications. Bioengineering (Basel) 2023; 10:bioengineering10050597. [PMID: 37237667 DOI: 10.3390/bioengineering10050597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
The objective of this study was to investigate the osteogenic and antimicrobial effect of bioactive glass S53P4 incorporated into β-tricalcium phosphate (β-TCP) scaffolds in vitro and the bone neoformation in vivo. β-TCP and β-TCP/S53P4 scaffolds were prepared by the gel casting method. Samples were morphologically and physically characterized through X-ray diffraction (XRD) and scanning electron microscope (SEM). In vitro tests were performed using MG63 cells. American Type Culture Collection reference strains were used to determine the scaffold's antimicrobial potential. Defects were created in the tibia of New Zealand rabbits and filled with experimental scaffolds. The incorporation of S53P4 bioglass promotes significant changes in the crystalline phases formed and in the morphology of the surface of the scaffolds. The β-TCP/S53P4 scaffolds did not demonstrate an in vitro cytotoxic effect, presented similar alkaline phosphatase activity, and induced a significantly higher protein amount when compared to β-TCP. The expression of Itg β1 in the β-TCP scaffold was higher than in the β-TCP/S53P4, and there was higher expression of Col-1 in the β-TCP/S53P4 group. Higher bone formation and antimicrobial activity were observed in the β-TCP/S53P4 group. The results confirm the osteogenic capacity of β-TCP ceramics and suggest that, after bioactive glass S53P4 incorporation, it can prevent microbial infections, demonstrating to be an excellent biomaterial for application in bone tissue engineering.
Collapse
Affiliation(s)
- Suelen Simões Amaral
- Institute of Science and Technology, São Paulo State University (UNESP), 777 Eng. Francisco José Longo Avenue, São José dos Campos 12245-000, SP, Brazil
| | - Beatriz Samara de Sousa Lima
- Institute of Science and Technology, São Paulo State University (UNESP), 777 Eng. Francisco José Longo Avenue, São José dos Campos 12245-000, SP, Brazil
| | - Sarah Oliveira Marco Avelino
- Institute of Science and Technology, São Paulo State University (UNESP), 777 Eng. Francisco José Longo Avenue, São José dos Campos 12245-000, SP, Brazil
| | - Bruno Roberto Spirandeli
- Bioceramics Laboratory, Federal University of São Paulo (UNIFESP), 330 Talim St, São José dos Campos 12231-280, SP, Brazil
| | - Tiago Moreira Bastos Campos
- Division of Fundamental Sciences, Technological Institute of Aeronautics (ITA), 50 Mal. Eduardo Gomes Plaza, São José dos Campos 12228-900, SP, Brazil
| | - Gilmar Patrocínio Thim
- Division of Fundamental Sciences, Technological Institute of Aeronautics (ITA), 50 Mal. Eduardo Gomes Plaza, São José dos Campos 12228-900, SP, Brazil
| | - Eliandra de Sousa Trichês
- Bioceramics Laboratory, Federal University of São Paulo (UNIFESP), 330 Talim St, São José dos Campos 12231-280, SP, Brazil
| | - Renata Falchete do Prado
- Institute of Science and Technology, São Paulo State University (UNESP), 777 Eng. Francisco José Longo Avenue, São José dos Campos 12245-000, SP, Brazil
| | - Luana Marotta Reis de Vasconcellos
- Institute of Science and Technology, São Paulo State University (UNESP), 777 Eng. Francisco José Longo Avenue, São José dos Campos 12245-000, SP, Brazil
| |
Collapse
|
21
|
Nandi SK, Roy M, Bandyopadhyay A, Bose S. In vivo biocompatibility of SrO and MgO doped brushite cements. J Biomed Mater Res B Appl Biomater 2023; 111:599-609. [PMID: 36254886 PMCID: PMC9852027 DOI: 10.1002/jbm.b.35177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/13/2022] [Accepted: 09/27/2022] [Indexed: 01/22/2023]
Abstract
The addition of dopants in biomaterials has emerged as a critical regulator of bone formation and regeneration due to their imminent role in the biological process. The present work evaluated the role of strontium (Sr) and magnesium (Mg) dopants in brushite cement (BrC) on in vivo bone healing performance in a rabbit model. Pure, 1 wt% SrO (Sr-BrC), 1 wt% MgO (Mg-BrC), and a binary composition of 1.0 wt% SrO + 1.0 wt% MgO (Sr + Mg-BrC) BrCs were implanted into critical-sized tibial defects in rabbits for up to 4 months. The in vivo bone healing of three doped and pure BrC samples was examined and compared using sequential radiological examination, histological evaluations, and fluorochrome labeling studies. The results indicated excellent osseous tissue formation for Sr-BrC and Sr + Mg-BrC and moderate bone regeneration for Mg-BrC compared to pure BrC. Our findings indicated that adding small amounts of SrO, MgO, and binary dopants to the BrC can significantly influence new bone formation for bone tissue engineering.
Collapse
Affiliation(s)
- Samit K. Nandi
- Department of Veterinary Surgery and RadiologyWest Bengal University of Animal and Fishery SciencesKolkataIndia
| | - Mangal Roy
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials EngineeringWashington State UniversityPullmanWashingtonUSA
- Present address:
Metallurgical and Materials EngineeringIIT‐KharagpurKharagpurIndia
| | - Amit Bandyopadhyay
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials EngineeringWashington State UniversityPullmanWashingtonUSA
| | - Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials EngineeringWashington State UniversityPullmanWashingtonUSA
| |
Collapse
|
22
|
Francisco I, Basílio Â, Ribeiro MP, Nunes C, Travassos R, Marques F, Pereira F, Paula AB, Carrilho E, Marto CM, Vale F. Three-Dimensional Impression of Biomaterials for Alveolar Graft: Scoping Review. J Funct Biomater 2023; 14:76. [PMID: 36826875 PMCID: PMC9961517 DOI: 10.3390/jfb14020076] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/22/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
Craniofacial bone defects are one of the biggest clinical challenges in regenerative medicine, with secondary autologous bone grafting being the gold-standard technique. The development of new three-dimensional matrices intends to overcome the disadvantages of the gold-standard method. The aim of this paper is to put forth an in-depth review regarding the clinical efficiency of available 3D printed biomaterials for the correction of alveolar bone defects. A survey was carried out using the following databases: PubMed via Medline, Cochrane Library, Scopus, Web of Science, EMBASE, and gray literature. The inclusion criteria applied were the following: in vitro, in vivo, ex vivo, and clinical studies; and studies that assessed bone regeneration resorting to 3D printed biomaterials. The risk of bias of the in vitro and in vivo studies was performed using the guidelines for the reporting of pre-clinical studies on dental materials by Faggion Jr and the SYRCLE risk of bias tool, respectively. In total, 92 publications were included in the final sample. The most reported three-dimensional biomaterials were the PCL matrix, β-TCP matrix, and hydroxyapatite matrix. These biomaterials can be combined with different polymers and bioactive molecules such as rBMP-2. Most of the included studies had a high risk of bias. Despite the advances in the research on new three-dimensionally printed biomaterials in bone regeneration, the existing results are not sufficient to justify the application of these biomaterials in routine clinical practice.
Collapse
Affiliation(s)
- Inês Francisco
- Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Laboratory for Evidence-Based Sciences and Precision Dentistry, University of Coimbra, 3000-075 Coimbra, Portugal
| | - Ângela Basílio
- Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - Madalena Prata Ribeiro
- Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - Catarina Nunes
- Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - Raquel Travassos
- Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - Filipa Marques
- Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - Flávia Pereira
- Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - Anabela Baptista Paula
- Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Laboratory for Evidence-Based Sciences and Precision Dentistry, University of Coimbra, 3000-075 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-075 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3030-370 Coimbra, Portugal
- Institute of Integrated Clinical Practice, Faculty of Medicine, University of Coimbra, 3004-531 Coimbra, Portugal
| | - Eunice Carrilho
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Laboratory for Evidence-Based Sciences and Precision Dentistry, University of Coimbra, 3000-075 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-075 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3030-370 Coimbra, Portugal
- Institute of Integrated Clinical Practice, Faculty of Medicine, University of Coimbra, 3004-531 Coimbra, Portugal
| | - Carlos Miguel Marto
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Laboratory for Evidence-Based Sciences and Precision Dentistry, University of Coimbra, 3000-075 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-075 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3030-370 Coimbra, Portugal
- Institute of Integrated Clinical Practice, Faculty of Medicine, University of Coimbra, 3004-531 Coimbra, Portugal
- Institute of Experimental Pathology, Faculty of Medicine, University of Coimbra, 3004-531 Coimbra, Portugal
| | - Francisco Vale
- Institute of Orthodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Laboratory for Evidence-Based Sciences and Precision Dentistry, University of Coimbra, 3000-075 Coimbra, Portugal
| |
Collapse
|
23
|
Zhao C, Liu W, Zhu M, Wu C, Zhu Y. Bioceramic-based scaffolds with antibacterial function for bone tissue engineering: A review. Bioact Mater 2022; 18:383-398. [PMID: 35415311 PMCID: PMC8965760 DOI: 10.1016/j.bioactmat.2022.02.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/25/2022] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Bone defects caused by trauma, tumor, congenital abnormality and osteoarthritis, etc. have been substantially impacted the lives and health of human. Artificial bone implants, like bioceramic-based scaffolds, provide significant benefits over biological counterparts and are critical for bone repair and regeneration. However, it is highly probable that bacterial infections occur in the surgical procedures or on bioceramic-based scaffolds. Therefore, it is of great significance to obtain bioceramic-based scaffolds with integrative antibacterial and osteogenic functions for treating bone implant-associated infection and promoting bone repair. To fight against infection problems, bioceramic-based scaffolds with various antibacterial strategies are developed for bone repair and regeneration and also have made great progresses. This review summarizes recent progresses in bioceramic-based scaffolds with antibacterial function, which include drug-induced, ion-mediated, physical-activated and their combined antibacterial strategies according to specific antibacterial mechanism. Finally, the challenges and opportunities of antibacterial bioceramic-based scaffolds are discussed. Bioceramic-based scaffolds with antibacterial function (BSAF) are reviewed. BSAF have a great potential in treating bone infection and promoting bone repair. Antibacterial strategies of BSAF include drug, ion, physical and combined ways. The combined strategy may be the optimal approach in fighting bone infection. Limitations, challenges and perspectives of BSAF are discussed.
Collapse
Affiliation(s)
- Chaoqian Zhao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Weiye Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
| | - Min Zhu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, PR China
- Corresponding author. School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, PR China.
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
- Corresponding author. State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China.
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
- Corresponding author. State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China.
| |
Collapse
|
24
|
Yuan X, Lu T, He F, Wu T, Wang X, Ye J. 3D-plotted zinc silicate/β-tricalcium phosphate ceramic scaffolds enable fast osteogenesis by activating the p38 signaling pathway. J Mater Chem B 2022; 10:9639-9653. [PMID: 36377518 DOI: 10.1039/d2tb01868c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Biomaterials in combination with multiple bioactive ions could create a favorable microenvironment for bone remolding. Herein, zinc silicate/β-tricalcium phosphate (ZS/β-TCP) composite ceramic scaffolds with different amounts of ZS (5, 10, and 15 wt%) were constructed using a three-dimensional fiber deposition (3DF) technique. The physicochemical, osteogenic and angiogenic properties of these interconnected macroporous scaffolds were investigated systematically. Simultaneously, GeneChip, alkaline phosphatase (ALP), western blot (WB) and polymerase chain reaction (PCR) were utilized to elucidate the underlying mechanism of the enhancement in osteogenic differentiation. The results showed that the incorporation of ZS significantly improved the mechanical performance by more than 5 fold in comparison with the β-TCP ceramic scaffold (4.79 ± 0.99 MPa). The ZS modified β-TCP scaffolds greatly supported the cytoactivity, adhesion, proliferation of mouse bone marrow mesenchymal stem cells (mBMSCs) and human umbilical vein endothelial cells (HUVECs). The expression levels of osteogenic genes and proteins as well as angiogenic genes were markedly upregulated by the sustained release of bioactive ions (mainly Si and Zn) from the composite scaffolds. The 10ZS/β-TCP demonstrated the best overall performance in vitro. Moreover, the 10ZS/β-TCP displayed a high bone volume fraction, bone maturity and angiogenesis after implantation in the rat skull defects for 6 weeks. It was further verified that ZS/β-TCP scaffolds stimulated the osteogenic differentiation of mBMSCs by activating the p38 signaling pathway directly. The 10ZS/β-TCP ceramic scaffold holds great potential for the fast repair of bone defects, and deep understanding of the mechanism will facilitate the formulation of new strategies for bone repair.
Collapse
Affiliation(s)
- Xinyuan Yuan
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou 510641, China. .,National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Teliang Lu
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou 510641, China. .,National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China.,Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Fupo He
- School of Electromechanical Engineering, Guangdong University of Technology, Guangzhou 510006, China.
| | - Tingting Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Xiaolan Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jiandong Ye
- School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology, Guangzhou 510641, China. .,National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China.,Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510641, China
| |
Collapse
|
25
|
Biodegradable Mg-Sc-Sr Alloy Improves Osteogenesis and Angiogenesis to Accelerate Bone Defect Restoration. J Funct Biomater 2022; 13:jfb13040261. [PMID: 36547521 PMCID: PMC9787880 DOI: 10.3390/jfb13040261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022] Open
Abstract
Magnesium (Mg) and its alloys are considered to be biodegradable metallic biomaterials for potential orthopedic implants. While the osteogenic properties of Mg alloys have been widely studied, few reports focused on developing a bifunctional Mg implant with osteogenic and angiogenic properties. Herein, a Mg-Sc-Sr alloy was developed, and this alloy's angiogenesis and osteogenesis effects were evaluated in vitro for the first time. X-ray Fluorescence (XRF), X-ray diffraction (XRD), and metallography images were used to evaluate the microstructure of the developed Mg-Sc-Sr alloy. Human umbilical vein/vascular endothelial cells (HUVECs) were used to evaluate the angiogenic character of the prepared Mg-Sc-Sr alloy. A mix of human bone-marrow-derived mesenchymal stromal cells (hBM-MSCs) and HUVEC cell cultures were used to assess the osteogenesis-stimulating effect of Mg-Sc-Sr alloy through alkaline phosphatase (ALP) and Von Kossa staining. Higher ALP activity and the number of calcified nodules (27% increase) were obtained for the Mg-Sc-Sr-treated groups compared to Mg-treated groups. In addition, higher VEGF expression (45.5% increase), tube length (80.8% increase), and number of meshes (37.9% increase) were observed. The Mg-Sc-Sr alloy showed significantly higher angiogenesis and osteogenic differentiation than pure Mg and the control group, suggesting such a composition as a promising candidate in bone implants.
Collapse
|
26
|
In Vivo Application of Silica-Derived Inks for Bone Tissue Engineering: A 10-Year Systematic Review. Bioengineering (Basel) 2022; 9:bioengineering9080388. [PMID: 36004914 PMCID: PMC9404869 DOI: 10.3390/bioengineering9080388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
As the need for efficient, sustainable, customizable, handy and affordable substitute materials for bone repair is critical, this systematic review aimed to assess the use and outcomes of silica-derived inks to promote in vivo bone regeneration. An algorithmic selection of articles was performed following the PRISMA guidelines and PICO method. After the initial selection, 51 articles were included. Silicon in ink formulations was mostly found to be in either the native material, but associated with a secondary role, or to be a crucial additive element used to dope an existing material. The inks and materials presented here were essentially extrusion-based 3D-printed (80%), and, overall, the most investigated animal model was the rabbit (65%) with a femoral defect (51%). Quality (ARRIVE 2.0) and risk of bias (SYRCLE) assessments outlined that although a large majority of ARRIVE items were “reported”, most risks of bias were left “unclear” due to a lack of precise information. Almost all studies, despite a broad range of strategies and formulations, reported their silica-derived material to improve bone regeneration. The rising number of publications over the past few years highlights Si as a leverage element for bone tissue engineering to closely consider in the future.
Collapse
|
27
|
Zhou J, Nie Y, Jin C, Zhang JXJ. Engineering Biomimetic Extracellular Matrix with Silica Nanofibers: From 1D Material to 3D Network. ACS Biomater Sci Eng 2022; 8:2258-2280. [PMID: 35377596 DOI: 10.1021/acsbiomaterials.1c01525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biomaterials at nanoscale is a fast-expanding research field with which extensive studies have been conducted on understanding the interactions between cells and their surrounding microenvironments as well as intracellular communications. Among many kinds of nanoscale biomaterials, mesoporous fibrous structures are especially attractive as a promising approach to mimic the natural extracellular matrix (ECM) for cell and tissue research. Silica is a well-studied biocompatible, natural inorganic material that can be synthesized as morpho-genetically active scaffolds by various methods. This review compares silica nanofibers (SNFs) to other ECM materials such as hydrogel, polymers, and decellularized natural ECM, summarizes fabrication techniques for SNFs, and discusses different strategies of constructing ECM using SNFs. In addition, the latest progress on SNFs synthesis and biomimetic ECM substrates fabrication is summarized and highlighted. Lastly, we look at the wide use of SNF-based ECM scaffolds in biological applications, including stem cell regulation, tissue engineering, drug release, and environmental applications.
Collapse
Affiliation(s)
- Junhu Zhou
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Yuan Nie
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Congran Jin
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - John X J Zhang
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| |
Collapse
|
28
|
Effect of magnesium oxide nanoparticles, hydroxyapatite and hydrogel on regeneration of transverse fracture of distal radius. Int J Health Sci (Qassim) 2022. [DOI: 10.53730/ijhs.v6ns2.6206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Study's purpose of this study is to conduct synthesis and evaluate the effect of hydroxyapatite (HA) with hydrogel locally magnesium oxide nanoparticles (MgONPS) locally or intraperitoneally (IP) on the healing of the distal third radial fracture. Concentrations of MgONPs 200μg/ml, dissolved in 1 cc distilled water and the solution stirred by a stirrer for 10 min. HA 0.5 mg in 1ml hydrogel and the solution stirring at the vortex for 15 min. These materials were evaluated in vitro to ensure their suitability with the tissues. Seventy-five healthy adult male rabbits, aged about 1.5- 2 years old with average weighting 1.7- 2.3 Kg. B.W were used. Rabbits were divided into three groups randomly (n=25), group A (HA mixed hydrogel applied locally), group B (HA mixed with hydrogel and MgONPs applied locally) and group C (HA mixed hydrogel applied locally and MgONPs IP). Animals were anesthetized by i.m 40 mg/ kg B.W ketamine hydrochloride and 5mg/ kg B.W xylazine. A 5cm incision had made cranio-medially in the skin of the forelimb (right forelimb) and exposure radius and ulna. The macroscopic evaluation revealed that all groups at 2nd week showed bone reaction in different degrees.
Collapse
|
29
|
Depboylu FN, Korkusuz P, Yasa E, Korkusuz F. Smart Bioceramics for Orthopedic Applications. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2022:157-186. [DOI: 10.1007/978-981-16-7439-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
30
|
Rahimnejad M, Rezvaninejad R, Rezvaninejad R, França R. Biomaterials in bone and mineralized tissue engineering using 3D printing and bioprinting technologies. Biomed Phys Eng Express 2021; 7. [PMID: 34438382 DOI: 10.1088/2057-1976/ac21ab] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/26/2021] [Indexed: 12/29/2022]
Abstract
This review focuses on recently developed printable biomaterials for bone and mineralized tissue engineering. 3D printing or bioprinting is an advanced technology to design and fabricate complex functional 3D scaffolds, mimicking native tissue forin vivoapplications. We categorized the biomaterials into two main classes: 3D printing and bioprinting. Various biomaterials, including natural, synthetic biopolymers and their composites, have been studied. Biomaterial inks or bioinks used for bone and mineralized tissue regeneration include hydrogels loaded with minerals or bioceramics, cells, and growth factors. In 3D printing, the scaffold is created by acellular biomaterials (biomaterial inks), while in 3D bioprinting, cell-laden hydrogels (bioinks) are used. Two main classes of bioceramics, including bioactive and bioinert ceramics, are reviewed. Bioceramics incorporation provides osteoconductive properties and induces bone formation. Each biopolymer and mineral have its advantages and limitations. Each component of these composite biomaterials provides specific properties, and their combination can ameliorate the mechanical properties, bioactivity, or biological integration of the 3D printed scaffold. Present challenges and future approaches to address them are also discussed.
Collapse
Affiliation(s)
- Maedeh Rahimnejad
- Biomedical Engineering Institute, Université de Montreal, Montreal, QC, Canada
| | - Raziyehsadat Rezvaninejad
- Department of Oral Medicine, Faculty of Dentistry, Hormozgan University of Medical Sciences, Hormozgan, Iran
| | | | - Rodrigo França
- Department of Restorative Dentistry, College of Dentistry, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
31
|
Zhongxing L, Shaohong W, Jinlong L, Limin Z, Yuanzheng W, Haipeng G, Jian C. Three-dimensional printed hydroxyapatite bone tissue engineering scaffold with antibacterial and osteogenic ability. J Biol Eng 2021; 15:21. [PMID: 34372891 PMCID: PMC8353754 DOI: 10.1186/s13036-021-00273-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/17/2021] [Indexed: 11/10/2022] Open
Abstract
The development of an effective scaffold for bone defect repair is an urgent clinical need. However, it is challenging to design a scaffold with efficient osteoinduction and antimicrobial activity for regeneration of bone defect. In this study, we successfully prepared a hydroxyapatite (HA) porous scaffold with a surface-specific binding of peptides during osteoinduction and antimicrobial activity using a three-dimensional (3D) printing technology. The HA binding domain (HABD) was introduced to the C-terminal of bone morphogenetic protein 2 mimetic peptide (BMP2-MP) and antimicrobial peptide of PSI10. The binding capability results showed that BMP2-MP and PSI10-containing HABD were firmly bound to the surface of HA scaffolds. After BMP2-MP and PSI10 were bound to the scaffold surface, no negative effect was observed on cell proliferation and adhesion. The gene expression and protein translation levels of type I collagen (COL-I), osteocalcin (OCN) and Runx2 have been significantly improved in the BMP2-MP/HABP group. The level of alkaline phosphatase significantly increased in the BMP2-MP/HABP group. The inhibition zone test against Staphylococcus aureus and Escherichia coli BL21 prove that the PSI10/HABP@HA scaffold has strong antibacterial ability than another group. These findings suggest that 3D-printed HA scaffolds with efficient osteoinduction and antimicrobial activity represent a promising biomaterial for bone defect reconstruction.
Collapse
Affiliation(s)
- Liu Zhongxing
- Department of Orthopedics, Affiliated Hospital of Chifeng University, Inner Mongolia, 024000, Chifeng, People's Republic of China
| | - Wu Shaohong
- Department of Stomatology, Affiliated Hospital of Chifeng University, Inner Mongolia, 024000, Chifeng, People's Republic of China
| | - Li Jinlong
- Department of Orthopedics, Affiliated Hospital of Chifeng University, Inner Mongolia, 024000, Chifeng, People's Republic of China.
| | - Zhang Limin
- Department of Ophthalmology, Affiliated Hospital of Chifeng University, Inner Mongolia, 024000, Chifeng, People's Republic of China
| | - Wang Yuanzheng
- Department of Orthopedics, Affiliated Hospital of Chifeng University, Inner Mongolia, 024000, Chifeng, People's Republic of China
| | - Gao Haipeng
- Department of Orthopedics, Affiliated Hospital of Chifeng University, Inner Mongolia, 024000, Chifeng, People's Republic of China
| | - Cao Jian
- Department of Orthopedics, Affiliated Hospital of Chifeng University, Inner Mongolia, 024000, Chifeng, People's Republic of China.
| |
Collapse
|
32
|
Sugimoto H, Inagaki Y, Furukawa A, Kira T, Kawasaki S, Uchihara Y, Akahane M, Tanaka Y. Silicate/zinc-substituted strontium apatite coating improves the osteoinductive properties of β-tricalcium phosphate bone graft substitute. BMC Musculoskelet Disord 2021; 22:673. [PMID: 34372804 PMCID: PMC8353809 DOI: 10.1186/s12891-021-04563-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
Background β-Tricalcium phosphate (β-TCP) is a popular synthetic bone graft substitute with excellent osteoconductive properties and bioabsorbability. However, its osteoinductive properties are inferior to those of autologous or allogeneic bone. Trace elements such as strontium (Sr), silica (Si), and zinc (Zn) have been reported to promote osteogenesis in materials. In this study, we aimed to determine whether a Si/Zn-substituted Sr apatite coating of β-TCP could enhance osteoinductive properties. Methods The apatite-coated β-TCP disks were prepared using nanoparticle suspensions of silicate-substituted Sr apatite (SrSiP) or silicate- and Zn-co-substituted Sr apatite (SrZnSiP). Bone marrow mesenchymal cells (BMSCs) from rat femur were cultured and subsequently seeded at a density of 1.0 × 106/cm2 onto apatite-coated and non-coated β-TCP disks. In vitro, the β-TCP disks were then placed in osteogenic medium, and lactate dehydrogenase (LDH) activity was measured from supernatants after culture for 2 days. Additionally, after culture for 14 days, the mRNA expression of genes encoding osteocalcin (OC), alkaline phosphatase (ALP), bone morphogenetic protein-2 (BMP-2), and vascular endothelial growth factor (VEGF) was evaluated by qRT-PCR. In vivo, the β-TCP disks were transplanted subcutaneously into rats that were sacrificed after 4 weeks. Then, the harvested disks were evaluated biochemically (ALP activity, OC content, mRNA expression of OC, ALP, BMP-2, and VEGF measured by qRT-PCR), radiologically, and histologically. Results Significantly higher mRNA expression of almost all evaluated osteogenic and angiogenic genes was observed in the SrZnSiP and SrSiP groups than in the non-coated group, with no significant cytotoxicity elicited by the apatite coating in vitro. Moreover, in vivo, the SrZnSiP and SrSiP groups showed significantly higher osteogenic and angiogenic gene expression and higher ALP activity and OC content than the non-coated group (P < 0.05). Radiological and histopathological findings revealed abundant bone formation in the apatite-coated group. Conclusions Our findings indicate that apatite coating of β-TCP improves osteoinductive properties without inducing significant cytotoxicity.
Collapse
Affiliation(s)
- Hironori Sugimoto
- Department of Orthopaedic Surgery, Nara Medical University, Shijocho 840, 634-8521, Kashihara, Nara, Japan
| | - Yusuke Inagaki
- Department of Orthopaedic Surgery, Nara Medical University, Shijocho 840, 634-8521, Kashihara, Nara, Japan.
| | - Akira Furukawa
- Department of Orthopaedic Surgery, Nara Medical University, Shijocho 840, 634-8521, Kashihara, Nara, Japan
| | - Tsutomu Kira
- Department of Orthopaedic Surgery, Nara Medical University, Shijocho 840, 634-8521, Kashihara, Nara, Japan
| | - Sachiko Kawasaki
- Department of Orthopaedic Surgery, Nara Medical University, Shijocho 840, 634-8521, Kashihara, Nara, Japan
| | - Yoshinobu Uchihara
- Department of Orthopaedic Surgery, Nara Medical University, Shijocho 840, 634-8521, Kashihara, Nara, Japan
| | - Manabu Akahane
- Department of Health and Welfare Services, National Institute of Public Health, 2-3-6 Minami, 351-0197, Wako, Saitama, Japan
| | - Yasuhito Tanaka
- Department of Orthopaedic Surgery, Nara Medical University, Shijocho 840, 634-8521, Kashihara, Nara, Japan
| |
Collapse
|
33
|
Kizalaite A, Grigoraviciute-Puroniene I, Asuigui DRC, Stoll SL, Cho SH, Sekino T, Kareiva A, Zarkov A. Dissolution-Precipitation Synthesis and Characterization of Zinc Whitlockite with Variable Metal Content. ACS Biomater Sci Eng 2021; 7:3586-3593. [PMID: 34318657 PMCID: PMC8396804 DOI: 10.1021/acsbiomaterials.1c00335] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/16/2021] [Indexed: 11/29/2022]
Abstract
In the present work, a series of zinc whitlockite (CaxZny(HPO4)2(PO4)12) powders was synthesized by a low-temperature dissolution-precipitation process for the first time. The phase conversion from calcium hydroxyapatite to zinc whitlockite occurred in an acidic medium in the presence of Zn2+ ions. Variable chemical composition of the synthesis products was achieved by changing Ca-to-Zn molar ratio in the reaction mixture. Investigation of the phase evolution as a function of time demonstrated that phase-pure zinc whitlockite powders can be synthesized in just 3 h. It is also demonstrated that single-phase products can be obtained when the Ca-to-Zn ratio in the reaction medium is in the range from 9 to 30. With higher or lower ratios, neighboring crystal phases such as scholzite or calcium hydroxyapatite were obtained. The morphology of the synthesized powders was found to be dependent on the chemical composition, transforming from hexagonal to rhombohedral plates with the increase of Zn content. Thermal stability studies revealed that the synthesized compounds were thermally unstable and decomposed upon heat treatment.
Collapse
Affiliation(s)
- Agne Kizalaite
- Institute
of Chemistry, Vilnius University, Naugarduko 24, LT-03225 Vilnius, Lithuania
| | | | - Dane Romar C. Asuigui
- Department
of Chemistry, Georgetown University, 37th and O Streets NW, Washington, D.C. 20057, United States
| | - Sarah L. Stoll
- Department
of Chemistry, Georgetown University, 37th and O Streets NW, Washington, D.C. 20057, United States
| | - Sung Hun Cho
- SANKEN
(The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Tohru Sekino
- SANKEN
(The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Aivaras Kareiva
- Institute
of Chemistry, Vilnius University, Naugarduko 24, LT-03225 Vilnius, Lithuania
| | - Aleksej Zarkov
- Institute
of Chemistry, Vilnius University, Naugarduko 24, LT-03225 Vilnius, Lithuania
| |
Collapse
|
34
|
Zhang H, Jiao C, Liu Z, He Z, Mengxing Ge, Zongjun Tian, Wang C, Wei Z, Shen L, Liang H. 3D-printed composite, calcium silicate ceramic doped with CaSO4·2H2O: Degradation performance and biocompatibility. J Mech Behav Biomed Mater 2021; 121:104642. [PMID: 34174680 DOI: 10.1016/j.jmbbm.2021.104642] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/31/2021] [Accepted: 06/05/2021] [Indexed: 12/22/2022]
Abstract
Calcium silicate is a common implant material with excellent mechanical strength and good biological activity. In recent years, the addition of strengthening materials to calcium silicate has been proven to promote bone tissue regeneration, but its degradation properties require further improvements. In this paper, calcium silicate was used as the matrix, and 10 wt% hydroxyapatite and 10 wt% strontium phosphate were added to im prove the biological activity of the scaffold. The effect of adding different amounts of calcium sulfate dihydrate (CaSO4·2H2O) on the degradation of the scaffold was explored. A porous ceramic scaffold was prepared by digital light processing (DLP) technology, and its performance was evaluated. Cell experiments showed that the addition of calcium sulfate improved cell proliferation and differentiation. Simulated body fluid (SBF) immersion tests showed that small amounts of apatite deposits appeared on the fourth day, larger deposits appeared on the 14th day, and degradation occurred on the surface after 28 days of immersion. Mechanical tests showed that the addition of 5 wt% CaSO4·2H2O improved the compressibility of the composite. After soaking in SBF for 14 days, it retained its compressive strength (11.8 MPa), which meets the requirements of cancellous bone, demonstrating its potential application value for bone repair.
Collapse
Affiliation(s)
- Hanxu Zhang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China; Jiangsu Key Laboratory of Digital Medical Equipment Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Chen Jiao
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China; Jiangsu Key Laboratory of Digital Medical Equipment Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Zibo Liu
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Zhijing He
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China; Jiangsu Key Laboratory of Digital Medical Equipment Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Mengxing Ge
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Zongjun Tian
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China; Jiangsu Key Laboratory of Digital Medical Equipment Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Changjiang Wang
- Department of Engineering and Design, University of Sussex, Brighton, BN1 9RH, United Kingdom
| | - Zhen Wei
- Jiangsu Pharmaceutical Association, Zhongshan East Road, 210002, Nanjing, China
| | - Lida Shen
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China; Jiangsu Key Laboratory of Digital Medical Equipment Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China.
| | - Huixin Liang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, 210008, China; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, 210016, China
| |
Collapse
|
35
|
Ramadas M, Ferreira JMF, Ballamurugan AM. Fabrication of three dimensional bioactive Sr 2+ substituted apatite scaffolds by gel-casting technique for hard tissue regeneration. J Tissue Eng Regen Med 2021; 15:577-585. [PMID: 33843156 DOI: 10.1002/term.3197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/23/2021] [Indexed: 11/08/2022]
Abstract
This study aimed to fabricate three-dimensional (3D) bioactive Sr2+ -substituted apatite (Sr-HAP) scaffolds prepared by gel-casting with polymer sponge infiltration technique. 3D Sr-HAP scaffolds were prepared as engineering constructs with interconnected porous structure with a pore size of 200-600 μm ranging from a 10 × 10 × 6 mm size was designed. The characterization of X-ray diffraction, field emission scanning electron microscopy, and energy dispersion spectroscopy was utilized in order to evaluate the crystalline phase, structure, and morphology in the interconnected porous of the synthesized Sr-HAP scaffold. The bioactive and biocompatible of the resultant Sr-HAP scaffolds were analyzed by using simulated body fluid solution. Furthermore, the cytotoxicity and proliferation of MG-63 cell lines on the scaffolds were examined in 24 h culture. Furthermore, in vivo experiments demonstrated that the tibia bone defect with 4 mm diameter in rabbits was successfully healed by Sr-HAP porous scaffold after 45 days implantation. The histological images indicated the improved cell proliferation and new bone formation occurred in the porous scaffold treated group. The results indicated that the fabricated Sr-HAP scaffold is a promising capacity to infuse bone regeneration and promote in vivo tissue repair.
Collapse
Affiliation(s)
- Munusamy Ramadas
- Department of Nanoscience and Technology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Jose M F Ferreira
- Department of Ceramics and Glass Engineering CICECO, University of Aveiro, Aveiro, Portugal
| | | |
Collapse
|
36
|
Peng W, Ren S, Zhang Y, Fan R, Zhou Y, Li L, Xu X, Xu Y. MgO Nanoparticles-Incorporated PCL/Gelatin-Derived Coaxial Electrospinning Nanocellulose Membranes for Periodontal Tissue Regeneration. Front Bioeng Biotechnol 2021; 9:668428. [PMID: 33842452 PMCID: PMC8026878 DOI: 10.3389/fbioe.2021.668428] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/02/2021] [Indexed: 12/19/2022] Open
Abstract
Electrospinning technique has attracted considerable attention in fabrication of cellulose nanofibrils or nanocellulose membranes, in which polycaprolactone (PCL) could be used as a promising precursor to prepare various cellulose nanofibril membranes for periodontal tissue regeneration. Conventional bio-membranes and cellulose films used in guided tissue regeneration (GTR) can prevent the downgrowth of epithelial cells, fibroblasts, and connective tissue in the area of tooth root but have limitations related to osteogenic and antimicrobial properties. Cellulose nanofibrils can be used as an ideal drug delivery material to encapsulate and carry some drugs. In this study, magnesium oxide (MgO) nanoparticles-incorporated PCL/gelatin core-shell nanocellulose periodontal membranes were fabricated using coaxial electrospinning technique, which was termed as Coaxial-MgO. The membranes using single-nozzle electrospinning technique, namely Blending-MgO and Blending-Blank, were used as control. The morphology and physicochemical property of these nanocellulose membranes were characterized by scanning electron microscopy (SEM), energy-dispersive spectrum of X-ray (EDS), transmission electron microscopy (TEM), contact angle, and thermogravimetric analysis (TGA). The results showed that the incorporation of MgO nanoparticles barely affected the morphology and mechanical property of nanocellulose membranes. Coaxial-MgO with core-shell fiber structure had better hydrophilic property and sustainable release of magnesium ion (Mg2+). CCK-8 cell proliferation and EdU staining demonstrated that Coaxial-MgO membranes showed better human periodontal ligament stem cells (hPDLSCs) proliferation rates compared with the other group due to its gelatin shell with great biocompatibility and hydrophilicity. SEM and immunofluorescence assay results illustrated that the Coaxial-MgO scaffold significantly enhanced hPDLSCs adhesion. In vitro osteogenic and antibacterial properties showed that Coaxial-MgO membrane enhanced alkaline phosphatase (ALP) activity, formation of mineralized nodules, osteogenic-related genes [ALP, collagen type 1 (COL1), runt-related transcription factor 2 (Runx2)], and high antibacterial properties toward Escherichia coli (E. coli) and Actinobacillus actinomycetemcomitans (A. a) when compared with controls. Our findings suggested that MgO nanoparticles-incorporated coaxial electrospinning PCL-derived nanocellulose periodontal membranes might have great prospects for periodontal tissue regeneration.
Collapse
Affiliation(s)
- Wenzao Peng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Shuangshuang Ren
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yibo Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ruyi Fan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xuanwen Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
37
|
Porous Zirconia/Magnesia Ceramics Support Osteogenic Potential In Vitro. MATERIALS 2021; 14:ma14041049. [PMID: 33672374 PMCID: PMC7926502 DOI: 10.3390/ma14041049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/09/2021] [Accepted: 02/17/2021] [Indexed: 11/17/2022]
Abstract
Porous zirconia (ZrO2), magnesia (MgO) and zirconia/magnesia (ZrO2/MgO) ceramics were synthesised by sintering and designated as ZrO2(100), ZrO2(75)MgO(25), ZrO2(50)MgO(50), ZrO2(25)MgO(75), MgO(100) based on their composition. The ceramic samples were characterised by means of scanning electron microscopy, X-ray diffraction, energy-dispersive X-ray spectroscopy and atomic absorption spectrometry to explore the incorporation of Mg atoms into the zirconia lattice. The resulting porosity of the samples was calculated based on the composition and density. The final porosity of the cylinder-shaped ceramic samples ranged between 30 and 37%. The mechanical analysis exhibited that the Young modulus increased and the microstress decreased with increasing magnesia amount, with values ranging from 175 GPa for zirconia to 301 GPa for magnesia. The adhesion, viability, proliferation and osteogenic activity of MC3T3-E1 pre-osteoblastic cells cultured on the zirconia/magnesia ceramics was found to increase, with the magnesia-containing ceramics exhibiting higher values of calcium mineralisation. The results from the mechanical analysis, the ALP activity, the calcium and collagen production demonstrate that the zirconia/magnesia ceramics possess robust osteoinductive capacity, therefore holding great potential for bone tissue engineering.
Collapse
|
38
|
Lin H, Zhou Y, Lei Q, Lin D, Chen J, Wu C. Effect of inorganic phosphate on migration and osteogenic differentiation of bone marrow mesenchymal stem cells. BMC DEVELOPMENTAL BIOLOGY 2021; 21:1. [PMID: 33407089 PMCID: PMC7788862 DOI: 10.1186/s12861-020-00229-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/10/2020] [Indexed: 12/26/2022]
Abstract
Background Phosphate is the major ingredient of bone tissue, and is also an important component of commercial bone substitute materials, bone scaffolds, and implant surface coatings. With the dissolution of the bone substitute materials and the degradation by cells, local ion concentrations will change and affect bone tissue reconstruction. Bone marrow -derived mesenchymal stem cells (BM-MSCs) are main autologous cells to repair injured bone. When bone injure occurs, BM-MSCs migrate to the damaged area, differentiate into osteoblasts, and secrete bioactive factors to promote bone tissue repaired. This study aimed to investigate the effect of inorganic phosphate (Pi) at a series of concentration on migration and osteogenic differentiation of human bone marrow -derived mesenchymal stem cells(hBM-MSCs). Methods The culture of hBM-MSCs in mediums with different concentration of Pi from 2 mM to 10 mM were performed. HBM-MSCs migration were examined with transwell assays. HBM-MSCs proliferation were evaluated by cell counting kit-8 colorimetric method. Osteogenic genes expression were analyzed by real-time reverse transcriptase polymerase chain reaction. Mineralized nodules formation were demonstrated by Alizarin red staining. Result 4–10 mM Pi could effectively promote the migration of hBM-MSCs at 12 h and 18 h. There was no significant difference in the migration number of hBM-MSCs in Pi culture mediums at a concentration of 6, 8, and10mM. 2–10 mM Pi could promote the proliferation of hBM-MSCs to varying degrees in the observation period, while 4–10 mM Pi could promote the osteogenic differentiation and mineralization of hBM-MSCs. Conclusion The findings in our study showed 4-10 mM Pi could promote the migration, osteogenic differentiation, and mineralization of hBM-MSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s12861-020-00229-x.
Collapse
Affiliation(s)
- Hengzhang Lin
- Department of Stomatology, Fujian Provincial Governmental Hospital, Fuzhou, China
| | - Yong Zhou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Qun Lei
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| | - Dong Lin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jiang Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Chuhuo Wu
- Fujian Medical University, Fuzhou, China
| |
Collapse
|
39
|
Wu Z, Meng Z, Wu Q, Zeng D, Guo Z, Yao J, Bian Y, Gu Y, Cheng S, Peng L, Zhao Y. Biomimetic and osteogenic 3D silk fibroin composite scaffolds with nano MgO and mineralized hydroxyapatite for bone regeneration. J Tissue Eng 2020; 11:2041731420967791. [PMID: 33294153 PMCID: PMC7705190 DOI: 10.1177/2041731420967791] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/01/2020] [Indexed: 01/15/2023] Open
Abstract
Artificial bioactive materials have received increasing attention worldwide in clinical orthopedics to repair bone defects that are caused by trauma, infections or tumors, especially dedicated to the multifunctional composite effect of materials. In this study, a weakly alkaline, biomimetic and osteogenic, three-dimensional composite scaffold (3DS) with hydroxyapatite (HAp) and nano magnesium oxide (MgO) embedded in fiber (F) of silkworm cocoon and silk fibroin (SF) is evaluated comprehensively for its bone repair potential in vivo and in vitro experiments, particularly focusing on the combined effect between HAp and MgO. Magnesium ions (Mg2+) has long been proven to promote bone tissue regeneration, and HAp is provided with osteoconductive properties. Interestingly, the weak alkaline microenvironment from MgO may also be crucial to promote Sprague-Dawley (SD) rat bone mesenchymal stem cells (BMSCs) proliferation, osteogenic differentiation and alkaline phosphatase (ALP) activities. This SF/F/HAp/nano MgO (SFFHM) 3DS with superior biocompatibility and biodegradability has better mechanical properties, BMSCs proliferation ability, osteogenic activity and differentiation potential compared with the scaffolds adding HAp or MgO alone or neither. Similarly, corresponding meaningful results are also demonstrated in a model of distal lateral femoral defect in SD rat. Therefore, we provide a promising 3D composite scaffold for promoting bone regeneration applications in bone tissue engineering.
Collapse
Affiliation(s)
- Ziquan Wu
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Zhulong Meng
- Municipal Hospital Affiliated to Medical School of Taizhou University, Taizhou, Zhejiang, China
| | - Qianjin Wu
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Delu Zeng
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Zhengdong Guo
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jiangling Yao
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yangyang Bian
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yuntao Gu
- The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Shaowen Cheng
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Lei Peng
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China.,Key Laboratory of Emergency and Trauma of Hainan Medical University, Ministry of Education, Haikou, Hainan, China
| | - Yingzheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
40
|
Sarkar N, Bose S. Controlled release of soy isoflavones from multifunctional 3D printed bone tissue engineering scaffolds. Acta Biomater 2020; 114:407-420. [PMID: 32652224 DOI: 10.1016/j.actbio.2020.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 01/09/2023]
Abstract
Recent challenges in post-surgical bone tumor management have elucidated the need for a multifunctional scaffold, which can be used for residual tumor-cell suppression, defect repair, and simultaneous bone regeneration. In this perspective, 3D printing allows to create a wide variety of patient-specific implant with complex porous architecture and compatible mechanical strength to that of cancellous bone. Here, a multifunctional bone graft substitute is designed by incorporating the three primary soy isoflavones: genistein, daidzein, and glycitein onto a 3D printed (3DP) tricalcium phosphate (TCP) scaffolds with designed pores, endowing them with in vitro chemopreventive, bone-cell proliferating and immune-modulatory potential. The interconnected porosity and biodegradability of 3DP TCP ceramics have allowed controlled release kinetics of genistein, daidzein and glycitein in acidic and physiological buffer medium for 16 days, which is fitted with Korsmeyer-Peppas model. Presence of genistein, a well-known natural biomolecule shows a 90% reduction in vitro osteosarcoma (MG-63) cell viability and proliferation after 11 days. Meanwhile, daidzein, the other primary isoflavone, promotes in vitro cellular attachment and enhances viability and proliferation of human fetal osteoblast cell (hFOB). Furthermore, controlled release of genistein, daidzein, and glycitein from 3DP TCP scaffold demonstrates improved hFOB cell proliferation, viability, and differentiation in a dynamic flow-perfusion bioreactor, which is utilized to better simulate the clinical microenvironment. Finally, in vivo H&E staining confirms controlled co-delivery of genistein-daidzein-glycitein from 3DP scaffold carefully modulated neutrophil recruitment to the surgery site after 24 h of implantation in a rat distal femur model. These results advance our understanding towards multipronged therapeutic approaches utilizing synthetic bone graft substitutes as a drug delivery vehicle, and more importantly, demonstrate the feasibility of localized tumor cell suppression and bone cell proliferation for post-surgical defect repair application. STATEMENT OF SIGNIFICANCE: Designed multimodal porosity of 3D printed TCP scaffold allows a controlled and sustained release of soy isoflavones, genistein, daidzein and glycitein in both physiological and acidic pH. Presence of genistein shows 90% reduction in vitro bone cancer cell viability and proliferation. Meanwhile, controlled release of genistein, daidzein, and glycitein from 3DP TCP scaffolds demonstrate improved osteoblast cell proliferation, viability, and differentiation in static and dynamic flow-perfusion bioreactor. Furthermore, H&E staining at 24 h post-surgical specimens from rat distal femur model shows neutrophil recruitment at the surgery site is significantly decreased, suggesting the anti-inflammatory property of soy isoflavones. This work provides deeper understanding on the design of a multifunctional 3D printed patient-specific scaffold with enhanced in vitro chemopreventive, osteogenic and in vivo anti-inflammatory ability.
Collapse
|
41
|
Yang S, Liang L, Liu L, Yin Y, Liu Y, Lei G, Zhou K, Huang Q, Wu H. Using MgO nanoparticles as a potential platform to precisely load and steadily release Ag ions for enhanced osteogenesis and bacterial killing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111399. [PMID: 33321576 DOI: 10.1016/j.msec.2020.111399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/31/2020] [Accepted: 08/19/2020] [Indexed: 12/14/2022]
Abstract
Bio-functional fillers including bio-ceramic, degradable metallic and composite particles are commonly introduced into bone tissue engineering (BTE) scaffolds to endow the materials with specific biological functions for enhanced bone defect therapy. In this work, MgO nanoparticles (NPs) were employed as a potential platform for precise loading and sustained release of Ag+. The results showed that MgO NPs possessed strong adsorption capacity (almost 100%) towards Ag+ in AgNO3 solutions with different concentrations (0.1, 1 and 10 mM). After the adsorption of Ag+ in AgNO3 solutions, cube-shaped MgO NPs transformed to lamella-structured nano-composites (NCs) composed of Mg(OH)2 and Ag2O, which were referred as MgO-xAg (x = 0.1, 1 or 10) NCs depending on the employed concentration of AgNO3 solution. After being suspended in distilled water, as-prepared positively charged NCs underwent a fast degradation process during the initial 4 days. From day 4 and 14, steady release behaviors of Mg2+ and/or Ag+ from the NCs were noticed. With the lowest loading amount of Ag+, MgO-0.1Ag NCs did not exhibit significant modulatory effect on SaOS-2 cell response. On the contrary, MgO-10Ag NCs loaded with the highest amount of Ag+ showed significant cyto-toxicity towards SaOS-2 cells. With appropriate amount of Ag+ loading, MgO-1Ag NCs showed significantly stimulatory effects on SaOS-2 cell proliferation and differentiation. This is evidenced by the enhanced cell viability, alkaline phosphatase (ALP) activity and collagen (COL) production as well as the gene expressions of ALP, COL and osteoprotegerin (OPG) in MgO-1Ag group. Moreover, MgO-1Ag exhibited strong bactericidal capacity against both Escherichia coli and Staphylococcus aureus. Together, the results indicate that MgO could be employed as a potential platform for precise loading and sustained release of Ag+. MgO-1Ag NCs are promising to be used as bio-functional fillers in BTE scaffolds for simultaneously promoted osteogenesis and bacterial killing.
Collapse
Affiliation(s)
- Si Yang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China
| | - Luxin Liang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China
| | - Ling Liu
- Hepatobiliary and Pancreatic Surgery Department, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Yong Yin
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China; School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Yong Liu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Kun Zhou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Qianli Huang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China.
| | - Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China.
| |
Collapse
|
42
|
Effect of Bone Morphogenic Protein-2-Loaded Mesoporous Strontium Substitution Calcium Silicate/Recycled Fish Gelatin 3D Cell-Laden Scaffold for Bone Tissue Engineering. Processes (Basel) 2020. [DOI: 10.3390/pr8040493] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bone has a complex hierarchical structure with the capability of self-regeneration. In the case of critical-sized defects, the regeneration capabilities of normal bones are severely impaired, thus causing non-union healing of bones. Therefore, bone tissue engineering has since emerged to solve problems relating to critical-sized bone defects. Amongst the many biomaterials available on the market, calcium silicate-based (CS) cements have garnered huge interest due to their versatility and good bioactivity. In the recent decade, scientists have attempted to modify or functionalize CS cement in order to enhance the bioactivity of CS. Reports have been made that the addition of mesoporous nanoparticles onto scaffolds could enhance the bone regenerative capabilities of scaffolds. For this study, the main objective was to reuse gelatin from fish wastes and use it to combine with bone morphogenetic protein (BMP)-2 and Sr-doped CS scaffolds to create a novel BMP-2-loaded, hydrogel-based mesoporous SrCS scaffold (FGSrB) and to evaluate for its composition and mechanical strength. From this study, it was shown that such a novel scaffold could be fabricated without affecting the structural properties of FGSr. In addition, it was proven that FGSrB could be used for drug delivery to allow stable localized drug release. Such modifications were found to enhance cellular proliferation, thus leading to enhanced secretion of alkaline phosphatase and calcium. The above results showed that such a modification could be used as a potential alternative for future bone tissue engineering research.
Collapse
|
43
|
The Calcium Channel Affect Osteogenic Differentiation of Mesenchymal Stem Cells on Strontium-Substituted Calcium Silicate/Poly-ε-Caprolactone Scaffold. Processes (Basel) 2020. [DOI: 10.3390/pr8020198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There had been a paradigm shift in tissue engineering studies over the past decades. Of which, part of the hype in such studies was based on exploring for novel biomaterials to enhance regeneration. Strontium ions have been reported by others to have a unique effect on osteogenesis. Both in vitro and in vivo studies had demonstrated that strontium ions were able to promote osteoblast growth, and yet at the same time, inhibit the formation of osteoclasts. Strontium is thus considered an important biomaterial in the field of bone tissue engineering. In this study, we developed a Strontium-calcium silicate scaffold using 3D printing technology and evaluated for its cellular proliferation capabilities by assessing for protein quantification and mineralization of Wharton’s Jelly mesenchymal stem cells. In addition, verapamil (an L-type of calcium channel blocker, CCB) was used to determine the mechanism of action of strontium ions. The results found that the relative cell proliferation rate on the scaffold was increased between 20% to 60% within 7 days of culture, while the CCB group only had up to approximately 10% proliferation as compared with the control specimen. Besides, the CCB group had downregulation and down expressions of all downstream cell signaling proteins (ERK and P38) and osteogenic-related protein (Col I, OPN, and OC). Furthermore, CCB was found to have 3–4 times lesser calcium deposition and quantification after 7 and 14 days of culture. These results effectively show that the 3D printed strontium-contained scaffold could effectively stimulate stem cells to undergo bone differentiation via activation of L-type calcium channels. Such results showed that strontium-calcium silicate scaffolds have high development potential for bone tissue engineering.
Collapse
|
44
|
Regulation of Osteogenic Markers at Late Stage of Osteoblast Differentiation in Silicon and Zinc Doped Porous TCP. J Funct Biomater 2019; 10:jfb10040048. [PMID: 31694210 PMCID: PMC6963500 DOI: 10.3390/jfb10040048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/24/2019] [Accepted: 10/22/2019] [Indexed: 11/17/2022] Open
Abstract
Calcium phosphates (CaPs) are one of the most widely used synthetic materials for bone grafting applications in the orthopedic industry. Recent trends in synthetic bone graft applications have shifted towards the incorporation of metal trace elements that extend the performance of CaPs to have osteoinductive properties. The objective of this study is to investigate the effects of silicon (Si) and zinc (Zn) dopants in highly porous tricalcium phosphate (TCP) scaffolds on late-stage osteoblast cell differentiation markers. In this study, an oil emulsion method is utilized to fabricate highly porous SiO2 doped β-TCP (Si-TCP) and ZnO doped β-TCP (Zn-TCP) scaffolds through the incorporation of 0.5 wt.% SiO2 and 0.25 wt.% ZnO, respectively, to the β-TCP scaffold. Reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) is utilized to analyze the mRNA expression of osteoprotegerin (OPG), receptor activator of nuclear kappa beta ligand (RANKL), bone morphogenetic protein 2 (BMP2), and runt-related transcription factor 2 (Runx2) at the later stage of osteoblast differentiation, day 21 and day 28. Results show that the addition of Si and Zn to the β-TCP structure inhibited the β to α-TCP phase transformation and enhance the density without affecting the dissolution properties. Normal BMP-2 and Runx2 transcriptions are observed in both Si-TCP and Zn-TCP scaffolds at the initial time point, as demonstrated by RT-qPCR. Moreover, the addition of both Si and Zn positively regulate the osteoprotegerin: receptor activator of nuclear factor k-β ligand (OPG:RANKL) ratio at 21-days for Si-TCP and Zn-TCP scaffolds. These results demonstrate the effects of Si and Zn doped porous β-TCP scaffolds on the upregulation of osteoblast marker gene expression including OPG, RANKL, BMP-2, and Runx2, indicating the role of trace elements on the effective regulation of late-stage osteoblast cell differentiation markers.
Collapse
|
45
|
Basu S, Basu B. Unravelling Doped Biphasic Calcium Phosphate: Synthesis to Application. ACS APPLIED BIO MATERIALS 2019; 2:5263-5297. [DOI: 10.1021/acsabm.9b00488] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Subhadip Basu
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
46
|
Vu AA, Bose S. Vitamin D 3 Release from Traditionally and Additively Manufactured Tricalcium Phosphate Bone Tissue Engineering Scaffolds. Ann Biomed Eng 2019; 48:1025-1033. [PMID: 31168676 DOI: 10.1007/s10439-019-02292-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/16/2019] [Indexed: 12/17/2022]
Abstract
Bone is a randomized, complex porous network which researchers have tried to mimic within bone tissue engineering scaffolds. The objective of this study was to understand the effects of random and controlled scaffold porosity on the release kinetics of vitamin D3 to determine if a designed porous structure was comparable in effectiveness on osteoblast proliferation to the randomized essence of natural bone. In this study, porous tricalcium phosphate (TCP) scaffolds were prepared by fugitive material removal method using naphthalene and 3D printing to model random and controlled porosity, respectively. Scaffold comparison was made based on open pore volume percentage of which naphthalene scaffolds had 45.8 ± 1.5% and 3D printed scaffolds had 48.9 ± 2.5%, Comparative analysis of traditional bioceramic processing to additive manufacturing is limited especially regarding drug release kinetics. Results showed the naphthalene scaffold surface area was only 0.3% that of 3D printed scaffolds due to the lower open pore interconnectivity. This increase in surface area produced higher release of drug and osteoblast proliferation in 3D printed scaffolds comparatively. By 11 days, osteoblast proliferation was enhanced by 64% from scaffolds manufactured using 3D printing compared to traditional processing. Understanding the effects of processing methods of TCP scaffolds on the release kinetics of vitamin D3 and the system effects on cells can aid in low load bearing applications for bone tissue engineering.
Collapse
Affiliation(s)
- Ashley A Vu
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164, USA
| | - Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164, USA.
| |
Collapse
|
47
|
Chiu YC, Shie MY, Lin YH, Lee AKX, Chen YW. Effect of Strontium Substitution on the Physicochemical Properties and Bone Regeneration Potential of 3D Printed Calcium Silicate Scaffolds. Int J Mol Sci 2019; 20:E2729. [PMID: 31163656 PMCID: PMC6600364 DOI: 10.3390/ijms20112729] [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: 04/23/2019] [Revised: 06/02/2019] [Accepted: 06/02/2019] [Indexed: 12/18/2022] Open
Abstract
In this study, we synthesized strontium-contained calcium silicate (SrCS) powder and fabricated SrCS scaffolds with controlled precise structures using 3D printing techniques. SrCS scaffolds were shown to possess increased mechanical properties as compared to calcium silicate (CS) scaffolds. Our results showed that SrCS scaffolds had uniform interconnected macropores (~500 µm) with a compressive strength 2-times higher than that of CS scaffolds. The biological behaviors of SrCS scaffolds were assessed using the following characteristics: apatite-precipitating ability, cytocompatibility, proliferation, and osteogenic differentiation of human mesenchymal stem cells (MSCs). With CS scaffolds as controls, our results indicated that SrCS scaffolds demonstrated good apatite-forming bioactivity with sustained release of Si and Sr ions. The in vitro tests demonstrated that SrCS scaffolds possessed excellent biocompatibility which in turn stimulated adhesion, proliferation, and differentiation of MSCs. In addition, the SrCS scaffolds were able to enhance MSCs synthesis of osteoprotegerin (OPG) and suppress macrophage colony-stimulating factor (M-CSF) thus disrupting normal bone homeostasis which led to enhanced bone formation over bone resorption. Implanted SrCS scaffolds were able to promote new blood vessel growth and new bone regeneration within 4 weeks after implantation in critical-sized rabbit femur defects. Therefore, it was shown that 3D printed SrCS scaffolds with specific controllable structures can be fabricated and SrCS scaffolds had enhanced mechanical property and osteogenesis behavior which makes it a suitable potential candidate for bone regeneration.
Collapse
Affiliation(s)
- Yung-Cheng Chiu
- School of Medicine, China Medical University, Taichung 40447, Taiwan.
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Ming-You Shie
- School of Dentistry, China Medical University, Taichung 40447, Taiwan.
- 3D Printing Medical Research Center, China Medical University Hospital, Taichung 40447, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 40447, Taiwan.
| | - Yen-Hong Lin
- 3D Printing Medical Research Center, China Medical University Hospital, Taichung 40447, Taiwan.
- The Ph.D. Program for Medical Engineering and Rehabilitation Science, China Medical University, Taichung 40447, Taiwan.
| | - Alvin Kai-Xing Lee
- School of Medicine, China Medical University, Taichung 40447, Taiwan.
- 3D Printing Medical Research Center, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Yi-Wen Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40447, Taiwan.
- 3D Printing Medical Research Institute, Asia University, Taichung 40447, Taiwan.
| |
Collapse
|
48
|
Ke D, Banerjee D, Bose S. In Vitro Characterizations of Si 4+ and Zn 2+ Doped Plasma Sprayed Hydroxyapatite Coatings Using Osteoblast and Osteoclast Coculture. ACS Biomater Sci Eng 2019; 5:1302-1310. [PMID: 33405648 DOI: 10.1021/acsbiomaterials.8b01553] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Osteoporosis is one of the most commonly identified bone disorders, which leads to an enhanced risk of bone fracture, especially for the older population. Hydroxyapatite (HA) coated titanium (Ti) alloys have been used widespread for load bearing applications like hip or knee replacements owing to their compositional similarity to natural bone; however, incorporation of osteoinductivity is still a challenge. The objective of this study is to evaluate the effects of SiO2 and ZnO as dopants in HA coated Ti alloys on cellular osteoporotic conditions mimicked by an in vitro osteoblast and osteoclast coculture model. HA, Si-HA, and Zn-HA coatings showed adhesive bond strengths of 25.7 ± 1.9 MPa, 23.8 ± 2.3 MPa, and 22.9 ± 3.5 MPa, respectively. To study the effects of doped HA coatings on the simulated osteoporotic cellular condition, human mesenchymal stem cells (hMSCs) and monocytes (THP-1) were seeded simultaneously in a ratio of 1:4, respectively. Gene expressions studies were carried out with marker genes showing that the presence of the dopants in the HA coating enhanced osteoblast proliferation along with diminishing cell growth of osteoclasts. This study demonstrates the promising effects of SiO2 and ZnO in plasma sprayed HA coatings on alleviating osteoporosis cellular conditions, which can potentially be used for load-bearing implants in aging patients whose bone resorption is more dominant than bone formation.
Collapse
Affiliation(s)
- Dongxu Ke
- W. M. Keck Biomedical Materials Research Laboratory School of Mechanical and Materials Engineering Washington State University, 980 E College Avenue, Pullman, Washington 99164-2920, United States
| | - Dishary Banerjee
- W. M. Keck Biomedical Materials Research Laboratory School of Mechanical and Materials Engineering Washington State University, 980 E College Avenue, Pullman, Washington 99164-2920, United States
| | - Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory School of Mechanical and Materials Engineering Washington State University, 980 E College Avenue, Pullman, Washington 99164-2920, United States
| |
Collapse
|