1
|
Shanbhag S, Al-Sharabi N, Fritz-Wallace K, Kristoffersen EK, Bunæs DF, Romandini M, Mustafa K, Sanz M, Gruber R. Proteomic Analysis of Human Serum Proteins Adsorbed onto Collagen Barrier Membranes. J Funct Biomater 2024; 15:302. [PMID: 39452600 PMCID: PMC11508515 DOI: 10.3390/jfb15100302] [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: 09/19/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
Collagen barrier membranes are frequently used in guided tissue and bone regeneration. The aim of this study was to analyze the signature of human serum proteins adsorbed onto collagen membranes using a novel protein extraction method combined with mass spectrometry. Native porcine-derived collagen membranes (Geistlich Bio-Gide®, Wolhusen, Switzerland) were exposed to pooled human serum in vitro and, after thorough washing, subjected to protein extraction either in conjunction with protein enrichment or via a conventional surfactant-based method. The extracted proteins were analyzed via liquid chromatography with tandem mass spectrometry. Bioinformatic analysis of global profiling, gene ontology, and functional enrichment of the identified proteins was performed. Overall, a total of 326 adsorbed serum proteins were identified. The enrichment and conventional methods yielded similar numbers of total (315 vs. 309), exclusive (17 vs. 11), and major bone-related proteins (18 vs. 14). Most of the adsorbed proteins (n = 298) were common to both extraction groups and included several growth factors, extracellular matrix (ECM) proteins, cell adhesion molecules, and angiogenesis mediators involved in bone regeneration. Functional analyses revealed significant enrichment of ECM, exosomes, immune response, and cell growth components. Key proteins [transforming growth factor-beta 1 (TGFβ1), insulin-like growth factor binding proteins (IGFBP-5, -6, -7)] were exclusively detected with the enrichment-based method. In summary, native collagen membranes exhibited a high protein adsorption capacity in vitro. While both extraction methods were effective, the enrichment-based method showed distinct advantages in detecting specific bone-related proteins. Therefore, the use of multiple extraction methods is advisable in studies investigating protein adsorption on biomaterials.
Collapse
Affiliation(s)
- Siddharth Shanbhag
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, 5009 Bergen, Norway
- Department of Periodontology, Faculty of Dentistry, University of Oslo, 0455 Oslo, Norway
| | - Niyaz Al-Sharabi
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway
| | - Katarina Fritz-Wallace
- Proteomics Unit of University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway
| | - Einar K. Kristoffersen
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, 5009 Bergen, Norway
- Department of Clinical Medicine, University of Bergen, 5009 Bergen, Norway
| | - Dagmar Fosså Bunæs
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway
| | - Mario Romandini
- Department of Periodontology, Faculty of Dentistry, University of Oslo, 0455 Oslo, Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway
| | - Mariano Sanz
- Department of Periodontology, Faculty of Dentistry, University of Oslo, 0455 Oslo, Norway
- ETEP Research Group, University Complutense of Madrid, 28040 Madrid, Spain
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
2
|
Ahmed E, Mulay P, Ramirez C, Tirado-Mansilla G, Cheong E, Gormley AJ. Mapping Biomaterial Complexity by Machine Learning. Tissue Eng Part A 2024; 30:662-680. [PMID: 39135398 DOI: 10.1089/ten.tea.2024.0067] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
Biomaterials often have subtle properties that ultimately drive their bespoke performance. Given this nuanced structure-function behavior, the standard scientific approach of one experiment at a time or design of experiment methods is largely inefficient for the discovery of complex biomaterials. More recently, high-throughput experimentation coupled with machine learning methods has matured beyond expert users allowing scientists and engineers from diverse backgrounds to access these powerful data science tools. As a result, we now have the opportunity to strategically utilize all available data from high-throughput experiments to train efficacious models and map the structure-function behavior of biomaterials for their discovery. Herein, we discuss this necessary shift to data-driven determination of structure-function properties of biomaterials as we highlight how machine learning is leveraged in identifying physicochemical cues for biomaterials in tissue engineering, gene delivery, drug delivery, protein stabilization, and antifouling materials. We also discuss data-mining approaches that are coupled with machine learning to map biomaterial functions that reduce the load on experimental approaches for faster biomaterial discovery. Ultimately, harnessing the prowess of machine learning will lead to accelerated discovery and development of optimal biomaterial designs.
Collapse
Affiliation(s)
- Eman Ahmed
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Prajakatta Mulay
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Cesar Ramirez
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Gabriela Tirado-Mansilla
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Eugene Cheong
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Adam J Gormley
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
3
|
Sutthiwanjampa C, Kang SH, Kim MK, Hwa Choi J, Kim HK, Woo SH, Bae TH, Kim WJ, Kang SH, Park H. Tumor necrosis factor-α-treated human adipose-derived stem cells enhance inherent radiation tolerance and alleviate in vivo radiation-induced capsular contracture. J Adv Res 2024:S2090-1232(24)00295-9. [PMID: 39019109 DOI: 10.1016/j.jare.2024.07.011] [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: 03/01/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024] Open
Abstract
INTRODUCTION Post-mastectomy radiotherapy plays a crucial role in breast cancer treatment but can lead to an inflammatory response causing soft tissue damage, particularly radiation-induced capsular contracture (RICC), impacting breast reconstruction outcomes. Adipose-derived stem cells (ADSCs), known for their regenerative potential via paracrine capacity, exhibit inherent radiotolerance. The influence of tumor necrosis factor-alpha (TNF-α) on ADSCs has been reported to enhance the paracrine effect of ADSCs, promoting wound healing by modulating inflammatory responses. OBJECTIVE This study investigates the potential of TNF-α-treated human ADSCs (T-hASCs) on silicone implants to alleviate RICC, hypothesizing to enhance suppressive effects on RICC by modulating inflammatory responses in a radiation-exposed environment. METHODS In vitro, T-hASCs were cultured on various surfaces to assess viability after exposure to radiation up to 20 Gy. In vivo, T-hASC and non-TNF-α-treated hASC (C-hASCs)-coated membranes were implanted in mice before radiation exposure, and an evaluation of the RICC mitigation took place 4 and 8 weeks after implantation. In addition, the growth factors released from T-hASCs were assessed. RESULTS In vitro, hASCs displayed significant radiotolerance, maintaining consistent viability after exposure to 10 Gy. TNF-α treatment further enhanced radiation tolerance, as evidenced by significantly higher viability than C-hASCs at 20 Gy. In vivo, T-hASC-coated implants effectively suppressed RICC, reducing capsule thickness. T-hASCs exhibited remarkable modulation of the inflammatory response, suppressing M1 macrophage polarization while enhancing M2 polarization. The elevated secretion of vascular endothelial growth factor from T-hASCs is believed to induce macrophage polarization, potentially reducing RICC. CONCLUSION This study establishes T-hASCs as a promising strategy for ameliorating the adverse effects experienced by breast reconstruction patients after mastectomy and radiation therapy. The observed radiotolerance, anti-fibrotic effects, and immune modulation suggest the possibility of enhancing patient outcomes and quality of life. Further research and clinical trials are warranted for broader clinical uses.
Collapse
Affiliation(s)
- Chanutchamon Sutthiwanjampa
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Seung Hyun Kang
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Mi Kyung Kim
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Departments of Pathology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Jin Hwa Choi
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Radiation Oncology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Han Koo Kim
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Soo Hyun Woo
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Tae Hui Bae
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Woo Joo Kim
- Department of Plastic Surgery, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong-si, Gyeonggi-do 14353, Republic of Korea
| | - Shin Hyuk Kang
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
4
|
Hadady H, Alam A, Khurana I, Mutreja I, Kumar D, Shankar MR, Dua R. Optimizing alkaline hydrothermal treatment for biomimetic smart metallic orthopedic and dental implants. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:31. [PMID: 38896291 PMCID: PMC11186882 DOI: 10.1007/s10856-024-06794-y] [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: 02/14/2024] [Accepted: 04/04/2024] [Indexed: 06/21/2024]
Abstract
Orthopedic and dental implant failure continues to be a significant concern due to localized bacterial infections. Previous studies have attempted to improve implant surfaces by modifying their texture and roughness or coating them with antibiotics to enhance antibacterial properties for implant longevity. However, these approaches have demonstrated limited effectiveness. In this study, we attempted to engineer the titanium (Ti) alloy surface biomimetically at the nanometer scale, inspired by the cicada wing nanostructure using alkaline hydrothermal treatment (AHT) to simultaneously confer antibacterial properties and support the adhesion and proliferation of mammalian cells. The two modified Ti surfaces were developed using a 4 h and 8 h AHT process in 1 N NaOH at 230 °C, followed by a 2-hour post-calcination at 600 °C. We found that the control plates showed a relatively smooth surface, while the treatment groups (4 h & 8 h AHT) displayed nanoflower structures containing randomly distributed nano-spikes. The results demonstrated a statistically significant decrease in the contact angle of the treatment groups, which increased wettability characteristics. The 8 h AHT group exhibited the highest wettability and significant increase in roughness 0.72 ± 0.08 µm (P < 0.05), leading to more osteoblast cell attachment, reduced cytotoxicity effects, and enhanced relative survivability. The alkaline phosphatase activity measured in all different groups indicated that the 8 h AHT group exhibited the highest activity, suggesting that the surface roughness and wettability of the treatment groups may have facilitated cell adhesion and attachment and subsequently increased secretion of extracellular matrix. Overall, the findings indicate that biomimetic nanotextured surfaces created by the AHT process have the potential to be translated as implant coatings to enhance bone regeneration and implant integration.
Collapse
Affiliation(s)
- Hanieh Hadady
- Polymer & Material Science Research, Department of Innovation & Technology Research, American Dental Association Science & Research Institute, L.L.C., Gaithersburg, MD, USA
| | - Arefin Alam
- Polymer & Material Science Research, Department of Innovation & Technology Research, American Dental Association Science & Research Institute, L.L.C., Gaithersburg, MD, USA
| | - Indu Khurana
- Department of Economics and Business, Hampden-Sydney College, Hampden-, Sydney, VA, USA
| | - Isha Mutreja
- Minnesota Dental Research Center for Biomaterials and Biomechanics, Department of Restorative Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Dhiraj Kumar
- Division of Pediatric Dentistry, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Mamilla Ravi Shankar
- Department of Mechanical Engineering, Indian Institute of Technology, Tirupati, AP, India
| | - Rupak Dua
- Polymer & Material Science Research, Department of Innovation & Technology Research, American Dental Association Science & Research Institute, L.L.C., Gaithersburg, MD, USA.
- Department of Chemical Engineering, Hampton University, Hampton, VA, USA.
| |
Collapse
|
5
|
Pecorini G, Braccini S, Simoni S, Corti A, Parrini G, Puppi D. Additive Manufacturing of Wet-Spun Poly(3-hydroxybutyrate-co-3-hydroxyvalerate)-Based Scaffolds Loaded with Hydroxyapatite. Macromol Biosci 2024; 24:e2300538. [PMID: 38534197 DOI: 10.1002/mabi.202300538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/20/2024] [Indexed: 03/28/2024]
Abstract
Tissue engineering represents an advanced therapeutic approach for the treatment of bone tissue defects. Polyhydroxyalkanoates are a promising class of natural polymers in this context thanks to their biocompatibility, processing versatility, and mechanical properties. The aim of this study is the development by computer-aided wet-spinning of novel poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV)-based composite scaffolds for bone engineering. In particular, PHBV scaffolds are loaded with hydroxyapatite (HA), an osteoinductive ceramic, in order to tailor their biological activity and mechanical properties. PHBV blending with poly(lactide-co-glycolide) (PLGA) is also explored to increase the processing properties of the polymeric mixture used for composite scaffold fabrication. Different HA percentages, up to 15% wt., can be loaded into the PHBV or PHBV/PLGA scaffolds without compromising their interconnected porous architecture, as well as the polymer morphological and thermal properties, as demonstrated by scanning electron microscopy, thermogravimetric analysis, and differential scanning calorimetry. In addition, HA loading results in increased scaffold compressive stiffness to levels comparable to those of trabecular bone tissue, as well as in higher in vitro MC3T3-E1 cell viability and production of mineralized extracellular matrix, in comparison to what observed for unloaded scaffolds. The observed mechanical and biological properties suggest the suitability of the developed scaffolds for bone engineering.
Collapse
Affiliation(s)
- Gianni Pecorini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | - Simona Braccini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | - Stefano Simoni
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | - Andrea Corti
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| | | | - Dario Puppi
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM Pisa, Via Moruzzi 13, Pisa, 56124, Italy
| |
Collapse
|
6
|
Cai F, Blanquer A, Costa MB, Schweiger L, Sarac B, Greer AL, Schroers J, Teichert C, Nogués C, Spieckermann F, Eckert J. Hierarchical Surface Pattern on Ni-Free Ti-Based Bulk Metallic Glass to Control Cell Interactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310364. [PMID: 38109153 PMCID: PMC11475312 DOI: 10.1002/smll.202310364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Ni-free Ti-based bulk metallic glasses (BMGs) are exciting materials for biomedical applications because of their outstanding biocompatibility and advantageous mechanical properties. The glassy nature of BMGs allows them to be shaped and patterned via thermoplastic forming (TPF). This work demonstrates the versatility of the TPF technique to create micro- and nano-patterns and hierarchical structures on Ti40Zr10Cu34Pd14Sn2 BMG. Particularly, a hierarchical structure fabricated by a two-step TPF process integrates 400 nm hexagonal close-packed protrusions on 2.5 µm square protuberances while preserving the advantageous mechanical properties from the as-cast material state. The correlations between thermal history, structure, and mechanical properties are explored. Regarding biocompatibility, Ti40Zr10Cu34Pd14Sn2 BMGs with four surface topographies (flat, micro-patterned, nano-patterned, and hierarchical-structured surfaces) are investigated using Saos-2 cell lines. Alamar Blue assay and live/dead analysis show that all tested surfaces have good cell proliferation and viability. Patterned surfaces are observed to promote the formation of longer filopodia on the edge of the cytoskeleton, leading to star-shaped and dendritic cell morphologies compared with the flat surface. In addition to potential implant applications, TPF-patterned Ti-BMGs enable a high level of order and design flexibility on the surface topography, expanding the available toolbox for studying cell behavior on rigid and ordered surfaces.
Collapse
Affiliation(s)
- Fei‐Fan Cai
- Department of Materials ScienceChair of Materials PhysicsMontanuniversität LeobenJahnstraße 12LeobenA‐8700Austria
- Erich Schmid Institute of Materials ScienceAustrian Academy of SciencesJahnstraße 12LeobenA‐8700Austria
| | - Andreu Blanquer
- Departament de Biologia Cel·lularFisiologia i ImmunologiaUniversitat Autònoma de BarcelonaCerdanyola del VallèsBellaterra08193Spain
| | - Miguel B. Costa
- Department of Materials Science & MetallurgyUniversity of CambridgeCambridgeCB3 0FSUK
| | - Lukas Schweiger
- Department of Materials ScienceChair of Materials PhysicsMontanuniversität LeobenJahnstraße 12LeobenA‐8700Austria
| | - Baran Sarac
- Erich Schmid Institute of Materials ScienceAustrian Academy of SciencesJahnstraße 12LeobenA‐8700Austria
| | - A. Lindsay Greer
- Department of Materials Science & MetallurgyUniversity of CambridgeCambridgeCB3 0FSUK
| | - Jan Schroers
- Department of Mechanical Engineering and Materials ScienceYale UniversityNew HavenCT 06511USA
| | - Christian Teichert
- Department PhysicsMechanics and Electrical EngineeringChair of PhysicsMontanuniversität LeobenFranz‐Josef‐Strasse 18LeobenA‐8700Austria
| | - Carme Nogués
- Departament de Biologia Cel·lularFisiologia i ImmunologiaUniversitat Autònoma de BarcelonaCerdanyola del VallèsBellaterra08193Spain
| | - Florian Spieckermann
- Department of Materials ScienceChair of Materials PhysicsMontanuniversität LeobenJahnstraße 12LeobenA‐8700Austria
| | - Jürgen Eckert
- Department of Materials ScienceChair of Materials PhysicsMontanuniversität LeobenJahnstraße 12LeobenA‐8700Austria
- Erich Schmid Institute of Materials ScienceAustrian Academy of SciencesJahnstraße 12LeobenA‐8700Austria
| |
Collapse
|
7
|
Hsu YH, Chou YC, Chen CL, Yu YH, Lu CJ, Liu SJ. Development of novel hybrid 3D-printed degradable artificial joints incorporating electrospun pharmaceutical- and growth factor-loaded nanofibers for small joint reconstruction. BIOMATERIALS ADVANCES 2024; 159:213821. [PMID: 38428121 DOI: 10.1016/j.bioadv.2024.213821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/04/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
Small joint reconstruction remains challenging and can lead to prosthesis-related complications, mainly due to the suboptimal performance of the silicone materials used and adverse host reactions. In this study, we developed hybrid artificial joints using three-dimensional printing (3D printing) for polycaprolactone (PCL) and incorporated electrospun nanofibers loaded with drugs and biomolecules for small joint reconstruction. We evaluated the mechanical properties of the degradable joints and the drug discharge patterns of the nanofibers. Empirical data revealed that the 3D-printed PCL joints exhibited good mechanical and fatigue properties. The drug-eluting nanofibers sustainedly released teicoplanin, ceftazidime, and ketorolac in vitro for over 30, 19, and 30 days, respectively. Furthermore, the nanofibers released high levels of bone morphogenetic protein-2 and connective tissue growth factors for over 30 days. An in vivo animal test demonstrated that nanofiber-loaded joints released high concentrations of antibiotics and analgesics in a rabbit model for 28 days. The animals in the drug-loaded degradable joint group showed greater activity counts than those in the surgery-only group. The experimental data suggest that degradable joints with sustained release of drugs and biomolecules may be utilized in small joint arthroplasty.
Collapse
Affiliation(s)
- Yung-Heng Hsu
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan
| | - Ying-Chao Chou
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan
| | - Chao-Lin Chen
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Hsun Yu
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan
| | - Chia-Jung Lu
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shih-Jung Liu
- Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan.
| |
Collapse
|
8
|
Alsmael MA, Al-Khafaji AM. Evaluation of High-Performance Polyether Ether Ketone Polymer Treated with Piranha Solution and Epigallocatechin-3-Gallate Coating. BIOMED RESEARCH INTERNATIONAL 2024; 2024:1741539. [PMID: 38628498 PMCID: PMC11019569 DOI: 10.1155/2024/1741539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/27/2024] [Accepted: 03/23/2024] [Indexed: 04/19/2024]
Abstract
Background Dental implantation has become a standard procedure with high success rates, relying on achieving osseointegration between the implant surface and surrounding bone tissue. Polyether ether ketone (PEEK) is a promising alternative to traditional dental implant materials like titanium, but its osseointegration capabilities are limited due to its hydrophobic nature and reduced surface roughness. Objective The aim of the study is to increase the surface roughness and hydrophilicity of PEEK by treating the surface with piranha solution and then coating the surface with epigallocatechin-3-gallate (EGCG) by electrospraying technique. Materials and Methods The study includes four groups intended to investigate the effect of piranha treatment and EGCG coating: a control group of PEEK discs with no treatment (C), PEEK samples treated with piranha solution (P), a group of PEEK samples coated with EGCG (E), and a group of PEEK samples treated with piranha solution and coated with EGCG (PE). Surface roughness, wettability, and microhardness were assessed through statistical analysis. Results Piranha treatment increased surface roughness, while EGCG coating moderated it, resulting in an intermediate roughness in the PE group. EGCG significantly improved wettability, as indicated by the reduced contact angle. Microhardness increased by about 20% in EGCG-coated groups compared to noncoated groups. Statistical analysis confirmed significant differences between groups in all tests. Conclusion This study demonstrates the potential of EGCG coating to enhance the surface properties of PEEK as dental implants. The combined piranha and EGCG modification approach shows promise for improved osseointegration, although further vivo research is necessary. Surface modification techniques hold the key to optimizing biomaterial performance, bridging the gap between laboratory findings and clinical implementation in dental implantology.
Collapse
Affiliation(s)
- Mohammed A. Alsmael
- Department of Prosthodontics, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | | |
Collapse
|
9
|
Ling Y, Duan M, Lyu W, Yang J, Liu Y, Ren S, Wu W. Electrospun L-Lysine/Amorphous Calcium Phosphate Loaded Core-Sheath Nanofibers for Managing Oral Biofilm Infections and Promoting Periodontal Tissue Repairment. Int J Nanomedicine 2024; 19:2917-2938. [PMID: 38525010 PMCID: PMC10961091 DOI: 10.2147/ijn.s453702] [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: 01/16/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction Periodontitis, a chronic inflammatory disease prevalent worldwide, is primarily treated through GTR for tissue regeneration. The efficacy of GTR, however, remains uncertain due to potential infections and the intricate microenvironment of periodontal tissue. Herein, We developed a novel core-shell structure multifunctional membrane using a dual-drug-loaded coaxial electrospinning technique (Lys/ACP-CNF), contains L-lysine in the outer layer to aid in controlling biofilms after GTR regenerative surgery, and ACP in the inner layer to enhance osteogenic performance for accelerating alveolar bone repair. Methods The biocompatibility and cell adhesion were evaluated through CCK-8 and fluorescence imaging, respectively. The antibacterial activity was assessed using a plate counting assay. ALP, ARS, and RT-qPCR were used to examine osteogenic differentiation. Additionally, an in vivo experiment was conducted on a rat model with acute periodontal defect and infection. Micro-CT and histological analysis were utilized to analyze the in vivo alveolar bone regeneration. Results Structural and physicochemical characterization confirmed the successful construction of the core-shell fibrous structure. Additionally, the Lys/ACP-CNF showed strong antibacterial coaggregation effects and induced osteogenic differentiation of PDLSCs in vitro. The in vivo experiment confirmed that Lys/ACP-CNF promotes new bone formation. Conclusion Lys/ACP-CNF rapidly exhibited excellent antibacterial activity, protected PDLSCs from infection, and was conducive to osteogenesis, demonstrating its potential application for clinical periodontal GTR surgery.
Collapse
Affiliation(s)
- Yufeng Ling
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
| | - Menglu Duan
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
| | - Wen Lyu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
| | - Jie Yang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
- Department of Periodontology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
| | - Yu Liu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
- Department of Senior Specialist, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
| | - Shuangshuang Ren
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
- Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
| | - Wenlei Wu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
- Department of Senior Specialist, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
| |
Collapse
|
10
|
Ghosh R, Gupta S, Mehrotra S, Kumar A. Surface-Modified Diopside-Reinforced PCL Biopolymer Composites with Enhanced Interfacial Strength and Mechanical Properties for Orthopedic Applications. ACS APPLIED MATERIALS & INTERFACES 2024; 16:7670-7685. [PMID: 38310585 DOI: 10.1021/acsami.3c15637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The phase separation of ceramics in a biopolymer matrix makes it challenging to achieve satisfactory mechanical properties required for orthopedic applications. It has been found that silane coupling agents can modify the surface of the bioceramic phase by forming a molecular bridge between the polymer and the ceramic, resulting in improved interfacial strength and adhesion. Therefore, in the present study, silane-modified diopside (DI) ceramic and ε-polycaprolactone (PCL) biopolymer composites were fabricated by injection molding method. The silane modification of DI resulted in their uniform dispersion in the PCL matrix, whereas agglomeration was found in composites containing unmodified DI. The thermal stability of the silane-modified DI-containing composites also increased. The Young's modulus of the composite containing 50% w/w DI modified by 3% w/w silane increased by 103% compared to composites containing 50% w/w unmodified DI. The biodegradation of the unmodified composites was significantly high, indicating their weak interfacial strength with the PCL matrix (p ≤ 0.001). The osteoconductive behavior of the composites was also validated by in vitro cell-material studies. Overall, our findings supported that the silane-modified composites have improved surface roughness, mechanical, and osteoconductive properties compared to the unmodified composite and have the potential for orthopedic applications.
Collapse
Affiliation(s)
- Rupita Ghosh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Sneha Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Shreya Mehrotra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Centre of Excellence for Orthopedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| |
Collapse
|
11
|
Majidi M, Pakzad S, Salimi M, Azadbakht A, Hajighasemlou S, Amoupour M, Nokhbedehghan Z, Bonakdar S, Sineh Sepehr K, Pal Singh Chauhan N, Gholipourmalekabadi M. Macrophage cell morphology-imprinted substrates can modulate mesenchymal stem cell behaviors and macrophage M1/M2 polarization for wound healing applications. Biotechnol Bioeng 2023; 120:3638-3654. [PMID: 37668186 DOI: 10.1002/bit.28546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 09/06/2023]
Abstract
Mesenchymal stem cells and macrophages (MQ) are two very important cells involved in the normal wound healing process. It is well understood that topological cues and mechanical factors can lead to different responses in stem cells and MQ by influencing their shape, cytoskeleton proliferation, migration, and differentiation, which play an essential role in the success or failure of biomaterial implantation and more importantly wound healing. On the other hand, the polarization of MQ from proinflammatory (M1) to prohealing (M2) phenotypes has a critical role in the acceleration of wound healing. In this study, the morphology of different MQ subtypes (M0, M1, and M2) was imprinted on a silicon surface (polydimethylsiloxane [PDMS]) to prepare a nano-topography cell-imprinted substrate with the ability to induce anti-inflammatory effects on the mouse adipose-derived stem cells (ADSCs) and RAW264.7 monocyte cell line (MO). The gene expression profiles and flow cytometry of MQ revealed that the cell shape microstructure promoted the MQ phenotypes according to the specific shape of each pattern. The ELISA results were in agreement with the gene expression profiles. The ADSCs on the patterned PDMS exhibited remarkably different shapes from no-patterned PDMS. The MOs grown on M2 morphological patterns showed a significant increase in expression and section of anti-inflammatory cytokine compared with M0 and M1 patterns. The ADSCs homing in niches heavily deformed the cytoskeletal, which is probably why the gene expression and phenotype unexpectedly changed. In conclusion, wound dressings with M2 cell morphology-induced surfaces are suggested as excellent anti-inflammatory and antiscarring dressings.
Collapse
Affiliation(s)
- Mohammad Majidi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saeedreza Pakzad
- Food and Drug Laboratory Research Center, Food and Drug Administration, Iran Ministry of Health and Medical Education, Tehran, Iran
| | - Maryam Salimi
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolnaser Azadbakht
- Department of Biomedical Engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Saieh Hajighasemlou
- Food and Drug Administration, Iran Ministry of Health and Medical Education, Tehran, Iran
| | - Moein Amoupour
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Nokhbedehghan
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Koushan Sineh Sepehr
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Mazaher Gholipourmalekabadi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Chen X, Li L, Chen L, Shao W, Chen Y, Fan X, Liu Y, Tang C, Ding S, Xu X, Zhou G, Feng X. Tea polyphenols coated sodium alginate-gelatin 3D edible scaffold for cultured meat. Food Res Int 2023; 173:113267. [PMID: 37803580 DOI: 10.1016/j.foodres.2023.113267] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 10/08/2023]
Abstract
This study aimed to use edible scaffolds as a platform for animal stem cell expansion, thus constructing block-shaped cell culture meat. The tea polyphenols (TP)-coated 3D scaffolds were constructed of sodium alginate (SA) and gelatin (Gel) with good biocompatibility and mechanical support. Initially, the physicochemical properties and mechanical properties of SA-Gel-TP scaffolds were measured, and the biocompatibility of the scaffolds was evaluated by C2C12 cells. SEM results showed that the scaffold had a porous laminar structure with TP particles attached to the surface, while FT-IR results also demonstrated the encapsulation of TP coating on the scaffold. In addition, the porosity of all scaffolds was higher than 40% and the degradation rate during the incubation cycle was less than 40% and the S2-G1-TP0.1-3 h scaffold has excellent cell adhesion and extension. Subsequently, we inoculated rabbit skeletal muscle myoblasts (RbSkMC) on the scaffold and induced differentiation. The results showed good adhesion and extension behavior of RbSkMC on S2-G1-TP0.1-3 h scaffolds with high expression of myogenic differentiation proteins and genes, and SEM results confirmed the formation of myotubes. Additionally, the adhesion rate of cells on scaffolds with TP coating was 1.5 times higher than that on scaffolds without coating, which significantly improved the cell proliferation rate and the morphology of cells with extension on the scaffolds. Furthermore, rabbit-derived cultured meat had similar appearance and textural characteristics to fresh meat. These conclusions indicate the high potential of the scaffolds with TP coating as a platform for the production of cultured meat products.
Collapse
Affiliation(s)
- Xiaohong Chen
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Linzi Li
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Lin Chen
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China.
| | - Wei Shao
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Yan Chen
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Xiaojing Fan
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Yaping Liu
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Changbo Tang
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Shijie Ding
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Xinglian Xu
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Guanghong Zhou
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Xianchao Feng
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China.
| |
Collapse
|
13
|
Iyigundogdu Z, Petek BS, Capkin Yurtsever M, Ceylan S. Melissa officinalisessential oil loaded polycaprolactone membranes: evaluation of antimicrobial activities and cytocompatibility for tissue engineering applications. Biomed Mater 2023; 18:065012. [PMID: 37741274 DOI: 10.1088/1748-605x/acfc9d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/22/2023] [Indexed: 09/25/2023]
Abstract
Antimicrobial biomaterials play important role in tissue engineering applications to protect damaged tissue from infections. The aim of this study is producing antimicrobial polycaprolactone (PCL) membranes by using a plant based antimicrobial agent. Therefore,Melissa officinalisessential oil (MEO) was investigated against ten types of microorganisms and remarkable antimicrobial activity was demonstrated. PCL:MEO membranes were prepared by solvent casting method by mixing MEO into PCL in various ratios (PCL:0M, PCL:0.25M, PCL:0.5M, and PCL:1M w/w). Water contact angle measurements showed that hydrophilicity of the membranes increased with increasing concentrations of MEO from 103.44° to 83.36° for PCL:0M and PCL:1M, respectively. It was determined that there was an inverse relationship between the MEO concentration and the mechanical properties. Notable antioxidant activity of PCL/MEO membranes was exhibited by the inhibition percent of 2,2-diphenyl-1-picrylhydrazyl (DPPH) which was increased from 24.74% to 44.79% for PCL:0M and PCL:1M, respectively. The antimicrobial activity of MEO was also highly maintained in PCL membranes. For PCL/MEO membranes, at least 99.9% of microorganisms were inhibited. Cytocompatibility of the membranes were investigated by resazurin assay, scanning electron microscopy analysis and 4',6-diamidino-2-phenylindole (DAPI) staining. PCL:0.25M and PCL:0.5M membranes supported the viability of L929 cells more than 87% when compared to PCL:0M membranes on day 6. However, the viability of L929 cells on PCL:1M membranes was about 43% indicating significant decrease on cellular activity. In conclusion, PCL:0.25M and PCL:0.5M membranes with their high antimicrobial activity, acceptable mechanical properties and cytocompatible properties, they can be considered as an alternative biomaterial for tissue engineering applications.
Collapse
Affiliation(s)
- Zeynep Iyigundogdu
- Department of Bioengineering, Adana Alparslan Turkes Science and Technology University, Adana, Türkiye
| | - Betül Sena Petek
- Department of Bioengineering, Adana Alparslan Turkes Science and Technology University, Adana, Türkiye
| | - Merve Capkin Yurtsever
- Department of Bioengineering, Adana Alparslan Turkes Science and Technology University, Adana, Türkiye
| | - Seda Ceylan
- Department of Bioengineering, Adana Alparslan Turkes Science and Technology University, Adana, Türkiye
| |
Collapse
|
14
|
Roato I, Genova T, Duraccio D, Ruffinatti FA, Zanin Venturini D, Di Maro M, Mosca Balma A, Pedraza R, Petrillo S, Chinigò G, Munaron L, Malucelli G, Faga MG, Mussano F. Mechanical and Biological Characterization of PMMA/Al 2O 3 Composites for Dental Implant Abutments. Polymers (Basel) 2023; 15:3186. [PMID: 37571080 PMCID: PMC10421041 DOI: 10.3390/polym15153186] [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: 07/05/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
The mechanical and biological behaviors of PMMA/Al2O3 composites incorporating 30 wt.%, 40 wt.%, and 50 wt.% of Al2O3 were thoroughly characterized as regards to their possible application in implant-supported prostheses. The Al2O3 particles accounted for an increase in the flexural modulus of PMMA. The highest value was recorded for the composite containing 40 wt.% Al2O3 (4.50 GPa), which was about 18% higher than that of its unfilled counterpart (3.86 GPa). The Al2O3 particles caused a decrease in the flexural strength of the composites, due to the presence of filler aggregates and voids, though it was still satisfactory for the intended application. The roughness (Ra) and water contact angle had the same trend, ranging from 1.94 µm and 77.2° for unfilled PMMA to 2.45 µm and 105.8° for the composite containing the highest alumina loading, respectively, hence influencing both the protein adsorption and cell adhesion. No cytotoxic effects were found, confirming that all the specimens are biocompatible and capable of sustaining cell growth and proliferation, without remarkable differences at 24 and 48 h. Finally, Al2O3 was able to cause strong cell responses (cell orientation), thus guiding the tissue formation in contact with the composite itself and not enhancing its osteoconductive properties, supporting the PMMA composite's usage in the envisaged application.
Collapse
Affiliation(s)
- Ilaria Roato
- CIR Dental School, Department of Surgical Sciences, University of Turin, Via Nizza 230, 10126 Torino, Italy; (I.R.); (A.M.B.); (R.P.); (F.M.)
| | - Tullio Genova
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Torino, Italy; (T.G.); (F.A.R.); (D.Z.V.); (G.C.); (L.M.)
| | - Donatella Duraccio
- Institute of Sciences and Technologies for Sustainable Energy and Mobility, National Council of Research, Strada delle Cacce 73, 10135 Torino, Italy; (M.D.M.); (M.G.F.)
| | - Federico Alessandro Ruffinatti
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Torino, Italy; (T.G.); (F.A.R.); (D.Z.V.); (G.C.); (L.M.)
| | - Diletta Zanin Venturini
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Torino, Italy; (T.G.); (F.A.R.); (D.Z.V.); (G.C.); (L.M.)
| | - Mattia Di Maro
- Institute of Sciences and Technologies for Sustainable Energy and Mobility, National Council of Research, Strada delle Cacce 73, 10135 Torino, Italy; (M.D.M.); (M.G.F.)
| | - Alessandro Mosca Balma
- CIR Dental School, Department of Surgical Sciences, University of Turin, Via Nizza 230, 10126 Torino, Italy; (I.R.); (A.M.B.); (R.P.); (F.M.)
| | - Riccardo Pedraza
- CIR Dental School, Department of Surgical Sciences, University of Turin, Via Nizza 230, 10126 Torino, Italy; (I.R.); (A.M.B.); (R.P.); (F.M.)
| | - Sara Petrillo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC), University of Turin, Via Nizza 52, 10126 Torino, Italy;
| | - Giorgia Chinigò
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Torino, Italy; (T.G.); (F.A.R.); (D.Z.V.); (G.C.); (L.M.)
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Torino, Italy; (T.G.); (F.A.R.); (D.Z.V.); (G.C.); (L.M.)
| | - Giulio Malucelli
- Politecnico di Torino, Department of Applied Science and Technology, C.so Duca Degli Abruzzi 24, 10129 Torino, Italy;
| | - Maria Giulia Faga
- Institute of Sciences and Technologies for Sustainable Energy and Mobility, National Council of Research, Strada delle Cacce 73, 10135 Torino, Italy; (M.D.M.); (M.G.F.)
| | - Federico Mussano
- CIR Dental School, Department of Surgical Sciences, University of Turin, Via Nizza 230, 10126 Torino, Italy; (I.R.); (A.M.B.); (R.P.); (F.M.)
| |
Collapse
|
15
|
Raptopoulos M, Fischer NG, Aparicio C. Implant surface physicochemistry affects keratinocyte hemidesmosome formation. J Biomed Mater Res A 2023; 111:1021-1030. [PMID: 36621832 DOI: 10.1002/jbm.a.37486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/04/2022] [Accepted: 12/05/2022] [Indexed: 01/10/2023]
Abstract
Previous studies have shown hydrophilic/hydrophobic implant surfaces stimulate/hinder osseointegration. An analogous concept was applied here using common biological functional groups on a model surface to promote oral keratinocytes (OKs) proliferation and hemidesmosomes (HD) to extend implant lifespans through increased soft tissue attachment. However, it is unclear what physicochemistry stimulates HDs. Thus, common biological functional groups (NH2 , OH, and CH3 ) were functionalized on glass using silanization. Non-functionalized plasma-cleaned glass and H silanization were controls. Surface modifications were confirmed with X-ray photoelectron spectroscopy and water contact angle. The amount of bovine serum albumin (BSA) and fibrinogen, and BSA thickness, were assessed to understand how adsorbed protein properties were influenced by physicochemistry and may influence HDs. OKs proliferation was measured, and HDs were quantified with immunofluorescence for collagen XVII and integrin β4. Plasma-cleaned surfaces were the most hydrophilic group overall, while CH3 was the most hydrophobic and OH was the most hydrophilic among functionalized groups. Modification with the OH chemical group showed the highest OKs proliferation and HD expression. The OKs response on OH surfaces appeared to not correlate to the amount or thickness of adsorbed model proteins. These results reveal relevant surface physicochemical features to favor HDs and improve implant soft tissue attachment.
Collapse
Affiliation(s)
- Michail Raptopoulos
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Periodontology, Department of Developmental and Surgical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nicholas G Fischer
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Conrado Aparicio
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, Minnesota, USA
- Basic and Translational Research Division, Department of Odontology, UIC Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- IBEC - Institute for BIoengineering of Catalonia, BIST-Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|
16
|
Lehká K, Starigazdová J, Mrázek J, Nešporová K, Šimek M, Pavlík V, Chmelař J, Čepa M, Barrios-Llerena ME, Kocurková A, Kriváková E, Koukalová L, Kubala L, Velebný V. An in vitro model that mimics the foreign body response in the peritoneum: Study of the bioadhesive properties of HA-based materials. Carbohydr Polym 2023; 310:120701. [PMID: 36925239 DOI: 10.1016/j.carbpol.2023.120701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/03/2023] [Accepted: 02/12/2023] [Indexed: 02/19/2023]
Abstract
A cascade of reactions known as the foreign body response (FBR) follows the implantation of biomaterials leading to the formation of a fibrotic capsule around the implant and subsequent health complications. The severity of the FBR is driven mostly by the physicochemical characteristics of implanted material, the method and place of implantation, and the degree of immune system activation. Here we present an in vitro model for assessing new materials with respect to their potential to induce a FBR in the peritoneum. The model is based on evaluating protein sorption and cell adhesion on the implanted material. We tested our model on the free-standing films prepared from hyaluronan derivatives with different hydrophobicity, swelling ratio, and rate of solubilization. The proteomic analysis of films incubated in the mouse peritoneum showed that the presence of fibrinogen was driving the cell adhesion. Neither the film surface hydrophobicity/hydrophilicity nor the quantity of adsorbed proteins were decisive for the induction of the long-term cell adhesion leading to the FBR, while the dissolution rate of the material proved to be a crucial factor. Our model thus helps determine the probability of a FBR to materials implanted in the peritoneum while limiting the need for in vivo animal testing.
Collapse
Affiliation(s)
- Kateřina Lehká
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic
| | - Jana Starigazdová
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic
| | - Jiří Mrázek
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic; Department of Medical Biophysics and Medical Informatics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Matěj Šimek
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic
| | - Vojtěch Pavlík
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic
| | - Josef Chmelař
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic
| | - Martin Čepa
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic
| | | | - Anna Kocurková
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37 Brno, Czech Republic; Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65 Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Pekařská 53, 656 91 Brno, Czech Republic
| | - Eva Kriváková
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65 Brno, Czech Republic
| | - Ludmila Koukalová
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic
| | - Lukáš Kubala
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37 Brno, Czech Republic; Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65 Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Pekařská 53, 656 91 Brno, Czech Republic
| | - Vladimír Velebný
- Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic
| |
Collapse
|
17
|
Robles D, Brizuela A, Fernández-Domínguez M, Gil J. Osteoblastic and Bacterial Response of Hybrid Dental Implants. J Funct Biomater 2023; 14:321. [PMID: 37367285 DOI: 10.3390/jfb14060321] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/10/2023] [Accepted: 06/11/2023] [Indexed: 06/28/2023] Open
Abstract
Bacterial infections in dental implants generate peri-implantitis disease that causes bone loss and the mobility of the dental implant. It is well known that specific ranges of roughness favor the proliferation of bacteria, and it is for this reason that new dental implants called hybrids have appeared. These implants have a smooth area in the coronal part and a rough surface in the apical part. The objective of this research is the physico-chemical characterization of the surface and the osteoblastic and microbiological behavior. One-hundred and eighty discs of titanium grade 3 with three different surfaces (smooth, smooth-rough, and completely rough) were studied. The roughness was determined by white light interferometry, and the wettability and surface energy by the sessile drop technique and the application of Owens and Wendt equations. Human osteoblast SaOS-2 was cultured to determine cell adhesion, proliferation, and differentiation. Microbiological studies were performed with two common bacterial strains in oral infection, E. faecalis and S. gordonii, at different times of culture. The roughness obtained for the smooth surface was Sa = 0.23 and for the rough surface it was 1.98 μm. The contact angles were more hydrophilic for the smooth surface (61.2°) than for the rough surface (76.1°). However, the surface energy was lower for the rough surface (22.70 mJ/m2) in both its dispersive and polar components than the smooth surface (41.77 mJ/m2). Cellular activity in adhesion, proliferation, and differentiation was much higher on rough surfaces than on smooth surfaces. After 6 h of incubation, the osteoblast number in rough surfaces was more than 32% higher in relation to the smooth surface. The cell area in smooth surfaces was higher than rough surfaces. The proliferation increased and the alkaline phosphatase presented a maximum after 14 days, with the mineral content of the cells being higher in rough surfaces. In addition, the rough surfaces showed greater bacterial proliferation at the times studied and in the two strains used. Hybrid implants sacrifice the good osteoblast behavior of the coronal part of the implant in order to obstruct bacterial adhesion. The following fact should be considered by clinicians: there is a possible loss of bone fixation when preventing peri-implantitis.
Collapse
Affiliation(s)
- Daniel Robles
- Department of Translational Medicine, CEU San Pablo University, Urbanización Montepríncipe, 28925 Madrid, Spain
- Facultad de Odontología, Universidad Europea Miguel de Cervantes, C/del Padre Julio Chevalier 2, 47012 Valladolid, Spain
| | - Aritza Brizuela
- Facultad de Odontología, Universidad Europea Miguel de Cervantes, C/del Padre Julio Chevalier 2, 47012 Valladolid, Spain
| | - Manuel Fernández-Domínguez
- Department of Oral and Maxillofacial Surgery, University Hospital Monteprincipe, University CEU San Pablo, Av. de Montepríncipe, s/n, 28668 Madrid, Spain
| | - Javier Gil
- Bioengineering Institute of Technology, Facultad de Medicina y Ciencias de la Salud, Universidad Internacional de Catalunya, Josep Trueta s/n. Sant Cugat del Vallés, 08195 Barcelona, Spain
| |
Collapse
|
18
|
Seddiqi H, Abbasi-Ravasjani S, Saatchi A, Amoabediny G, Zandieh-Doulabi B, Jin J, Klein-Nulend J. Osteogenic Activity on NaOH-Etched Three-Dimensional-Printed Poly-ɛ-Caprolactone Scaffolds in Perfusion or Spinner Flask Bioreactor. Tissue Eng Part C Methods 2023; 29:230-241. [PMID: 37253166 DOI: 10.1089/ten.tec.2023.0062] [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: 06/01/2023] Open
Abstract
Bioreactor systems, for example, spinner flask and perfusion bioreactors, and cell-seeded three-dimensional (3D)-printed scaffolds are used in bone tissue engineering strategies to stimulate cells and produce bone tissue suitable for implantation into the patient. The construction of functional and clinically relevant bone graft using cell-seeded 3D-printed scaffolds within bioreactor systems is still a challenge. Bioreactor parameters, for example, fluid shear stress and nutrient transport, will crucially affect cell function on 3D-printed scaffolds. Therefore, fluid shear stress induced by spinner flask and perfusion bioreactors might differentially affect osteogenic responsiveness of pre-osteoblasts inside 3D-printed scaffolds. We designed and fabricated surface-modified 3D-printed poly-ɛ-caprolactone (PCL) scaffolds, as well as static, spinner flask, and perfusion bioreactors to determine fluid shear stress and osteogenic responsiveness of MC3T3-E1 pre-osteoblasts seeded on the scaffolds in the bioreactors using finite element (FE)-modeling and experiments. FE-modeling was used to quantify wall shear stress (WSS) distribution and magnitude inside 3D-printed PCL scaffolds within spinner flask and perfusion bioreactors. MC3T3-E1 pre-osteoblasts were seeded on NaOH surface-modified 3D-printed PCL scaffolds, and cultured in customized static, spinner flask, and perfusion bioreactors up to 7 days. The scaffolds' physicochemical properties and pre-osteoblast function were assessed experimentally. FE-modeling showed that spinner flask and perfusion bioreactors locally affected WSS distribution and magnitude inside the scaffolds. The WSS distribution was more homogeneous inside scaffolds in perfusion than in spinner flask bioreactors. The average WSS on scaffold-strand surfaces ranged from 0 to 6.5 mPa for spinner flask bioreactors, and from 0 to 4.1 mPa for perfusion bioreactors. Surface modification of scaffolds by NaOH resulted in a surface with a honeycomb-like pattern and increased surface roughness (1.6-fold), but decreased water contact angle (0.3-fold). Both spinner flask and perfusion bioreactors increased cell spreading, proliferation, and distribution throughout the scaffolds. Perfusion, but not spinner flask bioreactors more strongly enhanced collagen (2.2-fold) and calcium deposition (2.1-fold) throughout the scaffolds after 7 days compared with static bioreactors, likely due to uniform WSS-induced mechanical stimulation of the cells revealed by FE-modeling. In conclusion, our findings indicate the importance of using accurate FE models to estimate WSS and determine experimental conditions for designing cell-seeded 3D-printed scaffolds in bioreactor systems. Impact Statement The success of cell-seeded three-dimensional (3D)-printed scaffolds depends on cell stimulation by biomechanical/biochemical factors to produce bone tissue suitable for implantation into the patient. We designed and fabricated surface-modified 3D-printed poly-ɛ-caprolactone (PCL) scaffolds, as well as static, spinner flask, and perfusion bioreactors to determine wall shear stress (WSS) and osteogenic responsiveness of pre-osteoblasts seeded on the scaffolds using finite element (FE)-modeling and experiments. We found that cell-seeded 3D-printed PCL scaffolds within perfusion bioreactors more strongly enhanced osteogenic activity than within spinner flask bioreactors. Our results indicate the importance of using accurate FE-models to estimate WSS and determine experimental conditions for designing cell-seeded 3D-printed scaffolds in bioreactor systems.
Collapse
Affiliation(s)
- Hadi Seddiqi
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Sonia Abbasi-Ravasjani
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Alireza Saatchi
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
- Department of Biomedical Engineering, Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
| | - Ghassem Amoabediny
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
- Department of Biomedical Engineering, Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
| | - Behrouz Zandieh-Doulabi
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Jianfeng Jin
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Ding Q, Ding C, Liu X, Zheng Y, Zhao Y, Zhang S, Sun S, Peng Z, Liu W. Preparation of nanocomposite membranes loaded with taxifolin liposome and its mechanism of wound healing in diabetic mice. Int J Biol Macromol 2023; 241:124537. [PMID: 37086765 DOI: 10.1016/j.ijbiomac.2023.124537] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/24/2023]
Abstract
In this study, a new wound dressing was developed to speed up the healing process of diabetic wounds. First of all, taxifolin liposome (TL) was manufactured in this study. Then, taxifolin (TAX) and TL were mixed with polyvinyl alcohol (PVA) and chitosan (CS) by electrostatic spinning to prepare nanocomposite membranes. Finally, the mechanism of nanocomposite membranes to accelerate diabetic wound healing was investigated. The diameter of TL-loaded polyvinyl alcohol/chitosan nanocomposite membranes (PVA/CS/TL) was 429.43 ± 78.07 nm. The results of in vitro experiments demonstrated that the PVA/CS/TL had better water absorption, water vapor transmission rate (WVTR), porosity, hydrophilicity, mechanical properties, slow-release, antioxidant capacity, and antibacterial properties. The results of in vivo experiments demonstrated that the wound healing rate of mice treated with PVA/CS/TL for eighteen days was 98.39 ± 0.34 %. Histopathological staining, immunohistochemical staining, and western blot experiments also demonstrated that PVA/CS/TL could promote wound healing in diabetic mice by inhibiting the activation of inhibitor kappa B alpha (IκBα)/nuclear factor-kappa B (NF-κB) signaling pathway and related pro-inflammatory factors to increase the expression of CD31 and VEGF in skin tissues. These results suggested that PVA/CS/TL could be a potential candidate for wound dressing to promote chronic skin wound healing.
Collapse
Affiliation(s)
- Qiteng Ding
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Chuanbo Ding
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Xinglong Liu
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Yinan Zheng
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Yingchun Zhao
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Shuai Zhang
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Shuwen Sun
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Zanwen Peng
- School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543002, China.
| | - Wencong Liu
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543002, China.
| |
Collapse
|
20
|
Liu Z, Tang Q, Liu RT, Yu MZ, Peng H, Zhang CQ, Zhu ZZ, Wei XJ. Laponite intercalated biomimetic multilayer coating prevents glucocorticoids induced orthopedic implant failure. Bioact Mater 2023; 22:60-73. [PMID: 36203962 PMCID: PMC9519439 DOI: 10.1016/j.bioactmat.2022.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/18/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022] Open
|
21
|
Babić Radić MM, Filipović VV, Vuković JS, Vukomanović M, Ilic-Tomic T, Nikodinovic-Runic J, Tomić SL. 2-Hydroxyethyl Methacrylate/Gelatin/Alginate Scaffolds Reinforced with Nano TiO2 as a Promising Curcumin Release Platform. Polymers (Basel) 2023; 15:polym15071643. [PMID: 37050256 PMCID: PMC10097359 DOI: 10.3390/polym15071643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/16/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
The idea of this study was to create a new scaffolding system based on 2-hydroxyethyl methacrylate, gelatin, and alginate that contains titanium(IV) oxide nanoparticles as a platform for the controlled release of the bioactive agent curcumin. The innovative strategy to develop hybrid scaffolds was the modified porogenation method. The effect of the scaffold composition on the chemical, morphology, porosity, mechanical, hydrophilicity, swelling, degradation, biocompatibility, loading, and release features of hybrid scaffolds was evaluated. A porous structure with interconnected pores in the range of 52.33–65.76%, favorable swelling capacity, fully hydrophilic surfaces, degradability to 45% for 6 months, curcumin loading efficiency above 96%, and favorable controlled release profiles were obtained. By applying four kinetic models of release, valuable parameters were obtained for the curcumin/PHEMA/gelatin/alginate/TiO2 release platform. Cytotoxicity test results depend on the composition of the scaffolds and showed satisfactory cell growth with visible cell accumulation on the hybrid surfaces. The constructed hybrid scaffolds have suitable high-performance properties, suggesting potential for further in vivo and clinical studies.
Collapse
|
22
|
Liao TY, King PC, Zhu D, Crawford RJ, Ivanova EP, Thissen H, Kingshott P. Surface Characteristics and Bone Biocompatibility of Cold-Sprayed Porous Titanium on Polydimethylsiloxane Substrates. ACS Biomater Sci Eng 2023; 9:1402-1421. [PMID: 36813258 DOI: 10.1021/acsbiomaterials.2c01506] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
A variant of the cold spray (CS) technique was applied for the functionalization of polymer-based materials such as polydimethylsiloxane (PDMS) to improve the extent of mammalian cell interactions with these substrates. This was demonstrated by the embedment of porous titanium (pTi) into PDMS substrates using a single-step CS technique. CS processing parameters such as gas pressure and temperature were optimized to achieve the mechanical interlocking of pTi in the compressed PDMS to fabricate a unique hierarchical morphology possessing micro-roughness. As evidenced by the preserved porous structure, the pTi particles did not undergo any significant plastic deformation upon impact with the polymer substrate. The thickness of the particle embedment layer was determined, by cross-sectional analysis, ranging from 120 μm to over 200 μm. The behavior of osteoblast-like cells MG63 coming into contact with the pTi-embedded PDMS was examined. The results showed that the pTi-embedded PDMS samples promoted 80-96% of cell adhesion and proliferation during the early stages of incubation. The low cytotoxicity of the pTi-embedded PDMS was confirmed, with cell viability of the MG63 cells being above 90%. Furthermore, the pTi-embedded PDMS facilitated the production of alkaline phosphatase and calcium deposition in the MG63 cells, as demonstrated by the higher amount of alkaline phosphatase (2.6 times) and calcium (10.6 times) on the pTi-embedded PDMS sample fabricated at 250 °C, 3 MPa. Overall, the work demonstrated that the CS process provided flexibility in the parameters used for the production of the modified PDMS substrates and is highly efficient for the fabrication of coated polymer products. The results obtained in this study suggest that a tailorable porous and rough architecture could be achieved that promoted osteoblast function, indicating that the method has promise in the design of titanium-polymer composite materials applied to biomaterials used in musculoskeletal applications.
Collapse
Affiliation(s)
- Tzu-Ying Liao
- School of Science, Computing & Engineering Technologies, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- Australian Research Council (ARC) Training Centre in Surface Engineering for Advanced Materials (SEAM), Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | - Peter C King
- Australian Research Council (ARC) Training Centre in Surface Engineering for Advanced Materials (SEAM), Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | - Deming Zhu
- School of Science, Computing & Engineering Technologies, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Russell J Crawford
- Australian Research Council (ARC) Training Centre in Surface Engineering for Advanced Materials (SEAM), Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- College of STEM, School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Elena P Ivanova
- Australian Research Council (ARC) Training Centre in Surface Engineering for Advanced Materials (SEAM), Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- College of STEM, School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Helmut Thissen
- Australian Research Council (ARC) Training Centre in Surface Engineering for Advanced Materials (SEAM), Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | - Peter Kingshott
- School of Science, Computing & Engineering Technologies, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- Australian Research Council (ARC) Training Centre in Surface Engineering for Advanced Materials (SEAM), Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| |
Collapse
|
23
|
Zhang B, Leng J, Ouyang Z, Yang Z, Zhang Q, Li Q, Li D, Zhao H. Superhydrophilic and topography-regulatable surface grafting on PEEK to improve cellular affinity. BIOMATERIALS ADVANCES 2023; 146:213310. [PMID: 36716597 DOI: 10.1016/j.bioadv.2023.213310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/29/2022] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Abstract
Polyetheretherketone (PEEK) has been widely used in the preparation of orthopedic implants due to its biological inertness and similar mechanical modulus to natural bone. However, the affinity between biological tissue (bone and soft tissue) and PEEK surface is weak, leading to low osseointegration and an increased risk of inflammation. The situation could be improved by modifying PEEK surface. Surfaces with good hydrophilicity and proper microtopography would promote cellular adhesion and proliferation. This work presented a two-step surface modification method to achieve the effect. Polyacrylic acid (PAA) chains were grafted on PEEK surface by UV irradiation. Then, ethylenediamine (EDA) was added to introduce amino groups and promote the cross-linking of PAA chains. Furthermore, a mathematical model was built to describe and regulate the surface topography growth process semi-quantitatively. The model fits experimental data quite well (adjusted R2 = 0.779). Results showed that the modified PEEK surface obtained superhydrophilicity. It significantly improved the adhesion and proliferation of BMSCs and MFBs by activating the FAK pathway and Rho family GTPase. The cellular affinity performed better when the surface topography was in network structure with holes in about 25 μm depth and 20-50 μm diameter. Good hydrophilicity seems necessary for the FAK pathway activation, but simply improving surface hydrophilicity might not be enough for cellular affinity improvement. Surface topography at micron scale should be a more important cue. This simple surface modification method could be contributed to further study of cell-microtopography interaction and have potential applications in clinical PEEK orthopedic implants.
Collapse
Affiliation(s)
- Bowen Zhang
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China; National Medical Products Administration (NMPA), Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China
| | - Junqing Leng
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China; National Medical Products Administration (NMPA), Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China
| | - Zhicong Ouyang
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Department of Spine Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, 510665 Guangzhou, China
| | - Zijian Yang
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Department of Spine Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, 510665 Guangzhou, China
| | - Qing Zhang
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China; Center for Medical Device Evaluation, National Medical Products Administration (NMPA), 100081 Beijing, China
| | - Qingchu Li
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Department of Spine Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, 510665 Guangzhou, China
| | - Dichen Li
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China; National Medical Products Administration (NMPA), Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China.
| | - Huiyu Zhao
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Department of Spine Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, 510665 Guangzhou, China.
| |
Collapse
|
24
|
Sahu MR, Sampath Kumar TS, Chakkingal U, Dewangan VK, Doble M. Influence of fine‐grained structure produced by groove pressing on the properties of pure Mg and commercial
ZE41
alloy. J Biomed Mater Res A 2023. [DOI: 10.1002/jbm.a.37502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Affiliation(s)
- Manas Ranjan Sahu
- Department of Metallurgical and Materials Engineering Indian Institute of Technology Madras Chennai India
| | - T. S. Sampath Kumar
- Department of Metallurgical and Materials Engineering Indian Institute of Technology Madras Chennai India
| | - Uday Chakkingal
- Department of Metallurgical and Materials Engineering Indian Institute of Technology Madras Chennai India
| | - Vimal Kumar Dewangan
- Department of Metallurgical and Materials Engineering Indian Institute of Technology Madras Chennai India
- Department of Biotechnology Indian Institute of Technology Madras Chennai India
| | - Mukesh Doble
- Department of Biotechnology Indian Institute of Technology Madras Chennai India
| |
Collapse
|
25
|
Yamanaka JS, Oliveira AC, Bastos AR, Fernandes EM, Reis RL, Correlo VM, Shimano AC. Collagen membrane from bovine pericardium for treatment of long bone defect. J Biomed Mater Res B Appl Biomater 2023; 111:261-270. [PMID: 36507698 DOI: 10.1002/jbm.b.35148] [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: 07/11/2020] [Revised: 06/20/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022]
Abstract
The treatment of bone regeneration failures has been constantly improved with the study of new biomaterials. Techgraft® is a collagen membrane derived from bovine pericardium, which has been shown to have biocompatibility and effectiveness in tissue repair. However, its use in orthopedics has not yet been evaluated. Therefore, the purpose of this study was to characterize a bovine pericardium collagen membrane and evaluate the effects of its use in the regeneration of a bone defect in rat tibia. Scanning electron microscopy, atomic force microscopy, weight lost and water uptake tests, and mechanical test were performed. Afterwards, the membrane was tested in an experimental study, using 12 male Sprague Dawley rats. A bone defect was surgically made in tibiae of animals, which were assigned to two groups (n = 6): bone defect treated with collagen membrane (TG) and bone defect without treatment (CONT). Then, tibiae were submitted to micro-CT. The membranes preserved their natural collagen characteristics, presenting great strength, high water absorption, hydrophilicity, and almost complete dissolution in 30 days. In the experimental study, the membrane enhanced the growth of bone tissue in contact with its surface. A higher bone volume and trabeculae number and less trabecular space was observed in bone defects of the membrane group compared to the control group at 21 days. In conclusion, the Techgraft membrane seems to have favorable characteristics for treatment of long bone repair.
Collapse
Affiliation(s)
- Jéssica S Yamanaka
- Departamento de Ortopedia e Anestesiologia. Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Clara Oliveira
- Departamento de Ortopedia e Anestesiologia. Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Raquel Bastos
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark - Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco - Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Emanuel M Fernandes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark - Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco - Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark - Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco - Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Vitor M Correlo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark - Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco - Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Antônio Carlos Shimano
- Departamento de Ortopedia e Anestesiologia. Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Wang H, Xia H, Xu Z, Natsuki T, Ni QQ. Effect of surface structure on the antithrombogenicity performance of poly(-caprolactone)-cellulose acetate small-diameter tubular scaffolds. Int J Biol Macromol 2023; 226:132-142. [PMID: 36470437 DOI: 10.1016/j.ijbiomac.2022.11.315] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Small-diameter artificial blood vessels have always faced the problem of thrombosis. In this research, three types of poly(-caprolactone)-cellulose acetate (PCL-CA) composite nanofiber membranes were prepared by various collectors to make into a tubular scaffold with a 4.5-mm diameter. The collector consisted of two sizes of stainless steel wire mesh large-mesh (LM) and small-mesh (SM), respectively. There is also a random flat (RF) that acts as the third type collector. The nanofiber membrane's surface structure mimicked the collectors' surface morphology, they named LM, SM and RF scaffolds. The water contact angles of RF and LM scaffolds are 126.5° and 105.5°, and the distinct square-groove construction greatly improves the contact angle of LM. The tubular scaffolds' radial mechanical property test demonstrated that the large-mesh (LM) tubular scaffold enhanced the strain and tensile strength; the tensile strength and strain are 30 % and 148 % higher than that of the random-flat (RF) tubular scaffold, respectively. The suture retention strength value of the LM tubular scaffold was 103 % higher than that of the RF tubular scaffold. The cytotoxicity and antithrombogenicity performance were also evaluated, the LM tubular scaffold has 88 % cell viability, and the 5-min blood coagulation index (BCI) value was 89 %, which is much higher than other tubular scaffolds. The findings indicate that changing the tubular scaffold's surface morphology cannot only enhance the mechanical and hydrophilic properties but also increase cell survival and antithrombogenicity performance. Thus, the development of a small-diameter artificial blood vessel will be a big step toward solving the problem on thrombosis. Furthermore, artificial blood vessel is expected to be a candidate material for biomedical applications.
Collapse
Affiliation(s)
- Hao Wang
- Interdisciplinary Graduate School of Science and Technology, Shinshu University, Ueda 386-8567, Japan
| | - Hong Xia
- Department of Mechanical Engineering and Robotics, Shinshu University, Ueda 386-8567, Japan
| | - Zhenzhen Xu
- College of Textiles and Garments, Anhui Polytechnic University, Wuhu 241000, Anhui, China.
| | - Toshiaki Natsuki
- Institute for Fiber Engineering (IFES), Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, 3-15-1 Tokida, Ueda, Nagano 386-8567, Japan; Faculty of Textile Science and Technology, Shinshu University, 3-15-1 Tokida, Ueda 386-8567, Japan
| | - Qing-Qing Ni
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China; Department of Mechanical Engineering and Robotics, Shinshu University, Ueda 386-8567, Japan; Key Laboratory of Advanced Textile Materials and Manufacturing Technology Ministry of Education Zhejiang Sci-Tech University, 310018 Hangzhou, China.
| |
Collapse
|
27
|
Gonçalves AM, Leal F, Moreira A, Schellhorn T, Blahnová VH, Zeiringer S, Vocetková K, Tetyczka C, Simaite A, Buzgo M, Roblegg E, Costa PF, Ertl P, Filová E, Kohl Y. Potential of Electrospun Fibrous Scaffolds for Intestinal, Skin, and Lung Epithelial Tissue Modeling. ADVANCED NANOBIOMED RESEARCH 2023. [DOI: 10.1002/anbr.202200104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
| | - Filipa Leal
- BIOFABICS Rua Alfredo Allen 455 4200-135 Porto Portugal
| | | | - Tobias Schellhorn
- Institute of Chemical Technologies and Analytics Vienna University of Technology Getreidemarkt 9/164 1060 Vienna Austria
| | - Veronika Hefka Blahnová
- Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 14220 Prague Czechia
| | - Scarlett Zeiringer
- Institute of Pharmaceutical Sciences University of Graz Universitaetsplatz 1 8010 Graz Austria
| | - Karolina Vocetková
- Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 14220 Prague Czechia
| | - Carolin Tetyczka
- Institute of Pharmaceutical Sciences University of Graz Universitaetsplatz 1 8010 Graz Austria
| | - Aiva Simaite
- InoCure s.r.o. Politických vězňů 935/13 11000 Praha 1 Prague Czech Republic
| | - Matej Buzgo
- BIOFABICS Rua Alfredo Allen 455 4200-135 Porto Portugal
| | - Eva Roblegg
- Institute of Pharmaceutical Sciences University of Graz Universitaetsplatz 1 8010 Graz Austria
| | | | - Peter Ertl
- Institute of Chemical Technologies and Analytics Vienna University of Technology Getreidemarkt 9/164 1060 Vienna Austria
| | - Eva Filová
- Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 14220 Prague Czechia
| | - Yvonne Kohl
- Fraunhofer Institute for Biomedical Engineering IBMT Joseph-von-Fraunhofer-Weg 1 66280 Sulzbach/Saar Germany
| |
Collapse
|
28
|
Hlinka J, Dostalova K, Cabanova K, Madeja R, Frydrysek K, Koutecky J, Rybkova Z, Malachova K, Umezawa O. Electrochemical, Biological, and Technological Properties of Anodized Titanium for Color Coded Implants. MATERIALS (BASEL, SWITZERLAND) 2023; 16:632. [PMID: 36676374 PMCID: PMC9866561 DOI: 10.3390/ma16020632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
Anodization coloring of titanium tools or implants is one of the common methods for the differentiation of each application by its size or type. Commercial purity titanium grade 4 plates (50 × 20 × 0.1 mm) were tested to obtain their electrochemical and other technological properties. The coloring process was done using the potential of 15, 30, 45, 60, and 75 Volts for 5 s in 1 wt. % citric acid in demineralized water solution. Organic acids solutions generally produce better surface quality compared to inorganic acids. The contact angle of colored surfaces was measured by the sessile drop method. Electrochemical impedance spectroscopy and potentiodynamic polarization were used for the determination of selected electrochemical and corrosion parameters of the tested surfaces. It was found that the anodization process decreases corrosion potential significantly. It was also confirmed that a higher potential used for anodization results in higher polarization resistance but also a decrease in corrosion potential. The anodization process at 75 V produces surfaces with the lowest corrosion rate under 1 nm/year and the noblest corrosion potential. It was confirmed that the anodization process in citric acid does not affect titanium cytotoxicity.
Collapse
Affiliation(s)
- Josef Hlinka
- Faculty of Materials and Technology, Department of Materials Engineering, VSB-Technical University of Ostrava, 17. Listopadu 2172/15, 708 00 Ostrava, Czech Republic
- Centre for Advanced Innovation Technologies, VSB-Technical University of Ostrava, 17. Listopadu 2172/15, 708 00 Ostrava, Czech Republic
| | - Kamila Dostalova
- Centre for Advanced Innovation Technologies, VSB-Technical University of Ostrava, 17. Listopadu 2172/15, 708 00 Ostrava, Czech Republic
| | - Kristina Cabanova
- Centre for Advanced Innovation Technologies, VSB-Technical University of Ostrava, 17. Listopadu 2172/15, 708 00 Ostrava, Czech Republic
| | - Roman Madeja
- Trauma Center, University Hospital Ostrava, 17. Listopadu 1790, 708 52 Ostrava, Czech Republic
| | - Karel Frydrysek
- Institute of Emergency Medicine, University of Ostrava, Syllabova 19, 703 00 Ostrava, Czech Republic
| | - Jan Koutecky
- Medin a.s., Vlachovicka 619, 592 31 Nove Mesto na Morave, Czech Republic
| | - Zuzana Rybkova
- Institute of Emergency Medicine, University of Ostrava, Syllabova 19, 703 00 Ostrava, Czech Republic
| | - Katerina Malachova
- Institute of Emergency Medicine, University of Ostrava, Syllabova 19, 703 00 Ostrava, Czech Republic
| | - Osamu Umezawa
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogoaya, Yokohama 240-8501, Japan
| |
Collapse
|
29
|
Characteristic Evaluation of Recombinant MiSp/Poly(lactic- co-glycolic) Acid (PLGA) Nanofiber Scaffolds as Potential Scaffolds for Bone Tissue Engineering. Int J Mol Sci 2023; 24:ijms24021219. [PMID: 36674734 PMCID: PMC9861889 DOI: 10.3390/ijms24021219] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/31/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Biomaterial-based nanofibrous scaffolds are the most effective alternative to bone transplantation therapy. Here, two recombinant minor ampullate spidroins (spider silk proteins), R1SR2 and NR1SR2C, were blended with Poly(lactic-co-glycolic) Acid (PLGA), respectively, to generate nanofiber scaffolds by electrospinning. The N-terminal (N), C-terminal (C), repeating (R1 and R2) and spacer (S) modules were all derived from the minor ampullate spidroins (MiSp). The physical properties and structures of the blended scaffolds were measured by scanning electron microscopy (SEM), water contact angle measurement, Fourier transform infrared spectroscopy (FTIR), Differential scanning calorimetry (DSC), and Tensile mechanical testing. The results showed that blending of MiSp (R1SR2 and NR1SR2C) reduced the diameter of nanofibers, increased the porosity and glass transition temperatures of nanofibrous scaffolds, and effectively improved the hydrophilicity and ultimate strain of scaffolds. It is worth noting that the above changes were more significant in the presence of the N- and C-termini of MiSp. In cell culture assays, human bone mesenchymal stem cells (HBMSCs) grown on NR1SR2C/PLGA (20/80) scaffolds displayed markedly enhanced proliferative and adhesive abilities compared with counterparts grown on pure PLGA scaffolds. Jointly, these findings indicated recombinant MiSp/PLGA, particularly NR1SR2C/PLGA (20/80) blend nanofibrous scaffolds, is promising for bone tissue engineering.
Collapse
|
30
|
Furko M, Horváth ZE, Czömpöly O, Balázsi K, Balázsi C. Biominerals Added Bioresorbable Calcium Phosphate Loaded Biopolymer Composites. Int J Mol Sci 2022; 23:ijms232415737. [PMID: 36555378 PMCID: PMC9779388 DOI: 10.3390/ijms232415737] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Nanocrystalline calcium phosphate (CP) bioceramic coatings and their combination with biopolymers are innovative types of resorbable coatings for load-bearing implants that can promote the integration of metallic implants into human bodies. The nanocrystalline, amorphous CP particles are an advantageous form of the various calcium phosphate phases since they have a faster dissolution rate than that of crystalline hydroxyapatite. Owing to the biomineral additions (Mg, Zn, Sr) in optimized concentrations, the base CP particles became more similar to the mineral phase in human bones (dCP). The effect of biomineral addition into the CaP phases was thoroughly studied. The results showed that the shape, morphology, and amorphous characteristic slightly changed in the case of biomineral addition in low concentrations. The optimized dCP particles were then incorporated into a chosen polycaprolactone (PCL) biopolymer matrix. Very thin, non-continuous, rough layers were formed on the surface of implant substrates via the spin coating method. The SEM elemental mapping proved the perfect incorporation and distribution of dCP particles into the polymer matrix. The bioresorption rate of thin films was followed by corrosion measurements over a long period of time. The corrosion results indicated a faster dissolution rate for the dCP-PCL composite compared to the dCP and CP powder layers.
Collapse
|
31
|
Kairalla EC, Bressiani JC, de Almeida Bressiani AH, de Carvalho Pinto Ribela MT, Higa OZ, de Queiroz AAA. Physicochemical and biological properties of nanohydroxyapatite grafted with star-shaped poly(ε-caprolactone). JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2353-2384. [PMID: 35876732 DOI: 10.1080/09205063.2022.2104599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
To overcome the disadvantages generated by the lack of interfacial bonding between hydroxyapatite nanocrystals (HAPN) and agglomeration of particles in the development of biodegradable nanocomposites a chemical grafting method was applied to modify the surface of HAPN through grafting of the three-arms star-shaped poly(ε-caprolactone) (SPCL) onto the nanoparticles. The chemical grafting of SPCL onto HAPN (SPCL-g-HAPN) has been investigated using Fourier transform infrared spectroscopy, thermogravimetric analysis, transmission electron microscopy (TEM), photoelectron spectroscopy, X-ray diffraction, zeta potential (ZP) and contact angle (CA). TEM micrographs of the SPCL-g-HAPN revealed the existence of hybrid organic/inorganic (O/I) nanoscale domains. The results of albumin (HSA) and fibrinogen (HFb) adsorption indicate resistance to HFb adsorption by SPCL-g-HAPN relatively to unmodified HAPN. The ZP and CA measurement suggest a heterogeneous topology for SPCL-g-HAPN likely due to the existence of hydrophobic-hydrophilic regions on the nanocomposite surface. The enzyme degradation by cholesterol esterase and lipase indicates that the rates of hydrolysis for SPCL-g-HAPN were very slow relative to the SPCL/HAPN blends. The in vitro biological studies showed that the human osteoblast-like cells (MG-63) cells had normal morphology and they were able to attach and spread out on SPCL-g-HAPN surfaces. A higher overall cellular proliferation was observed on SPCL-g-HAPN scaffolds compared to pure HAPN or SPCL materials.
Collapse
Affiliation(s)
- Eleni Cristina Kairalla
- Centro de Biotecnologia - CEBIO, Instituto de Pesquisas Energéticas e Nucleares (IPEN), São Paulo, SP, Brazil
| | - José Carlos Bressiani
- Centro de Biotecnologia - CEBIO, Instituto de Pesquisas Energéticas e Nucleares (IPEN), São Paulo, SP, Brazil
| | | | | | - Olga Zazuco Higa
- Centro de Biotecnologia - CEBIO, Instituto de Pesquisas Energéticas e Nucleares (IPEN), São Paulo, SP, Brazil
| | | |
Collapse
|
32
|
Sarkar S, Kundu S. Effect of different valent ions (Na+, Ca2+ & Y3+) on structural and morphological features of protein (BSA) thin films adsorbed on hydrophobic silicon (H-Si) surface. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
33
|
Sarıipek FB, Özaytekin İ, Erci F. Effect of ultrasound treatment on bacteriostatic activity of piezoelectric
PHB‐TiO
2
hybrid biodegradable scaffolds prepared by electrospinning technique. J Appl Polym Sci 2022. [DOI: 10.1002/app.53437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
| | - İlkay Özaytekin
- Department of Chemical Engineering Konya Technical University Konya Turkey
| | - Fatih Erci
- Department of Biotechnology Necmettin Erbakan University Konya Turkey
| |
Collapse
|
34
|
Fountain JN, Hawker MJ, Hartle L, Wu J, Montanari V, Sahoo JK, Davis LM, Kaplan DL, Kumar K. Towards Non-stick Silk: Tuning the Hydrophobicity of Silk Fibroin Protein. Chembiochem 2022; 23:e202200429. [PMID: 35998090 PMCID: PMC9830957 DOI: 10.1002/cbic.202200429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/22/2022] [Indexed: 02/03/2023]
Abstract
Silk fibroin protein is a biomaterial with excellent biocompatibility and low immunogenicity. These properties have catapulted the material as a leader for extensive use in stents, catheters, and wound dressings. Modulation of hydrophobicity of silk fibroin protein to further expand the scope and utility however has been elusive. We report that installing perfluorocarbon chains on the surface of silk fibroin transforms this water-soluble protein into a remarkably hydrophobic polymer that can be solvent-cast. A clear relationship emerged between fluorine content of the modified silk and film hydrophobicity. Water contact angles of the most decorated silk fibroin protein exceeded that of Teflon®. We further show that water uptake in prefabricated silk bars is dramatically reduced, extending their lifetimes, and maintaining mechanical integrity. These results highlight the power of chemistry under moderate conditions to install unnatural groups onto the silk fibroin surface and will enable further exploration into applications of this versatile biomaterial.
Collapse
Affiliation(s)
| | - Morgan J. Hawker
- Department of Chemistry and Biochemistry, California State University, Fresno, Fresno, CA 93740
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155
| | - Lauren Hartle
- Department of Chemistry, Tufts University, Medford, MA 02155
- Present address: Prime Impact Fund, Cambridge, MA 02139
| | - Junqi Wu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155
| | | | | | - Luke M. Davis
- Department of Chemistry, Tufts University, Medford, MA 02155
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155
| | - Krishna Kumar
- Department of Chemistry, Tufts University, Medford, MA 02155
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155
| |
Collapse
|
35
|
Preparation and Characterization of Nanofibrous Membranes Electro-Spun from Blended Poly(l-lactide-co-ε-caprolactone) and Recombinant Spider Silk Protein as Potential Skin Regeneration Scaffold. Int J Mol Sci 2022; 23:ijms232214055. [PMID: 36430534 PMCID: PMC9698895 DOI: 10.3390/ijms232214055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Biomaterial scaffolding serves as an important strategy in skin tissue engineering. In this research, recombinant spider silk protein (RSSP) and poly(L-lactide-co-ε-caprolactone) (PLCL) were blended in different ratios to fabricate nanofibrous membranes as potential skin regeneration scaffolds with an electro-spinning process. Scanning electron microscopy (SEM), water contact angles measurement, Fourier transform infrared (FTIR) spectroscopy, wide angle X-ray diffraction (WAXD), tensile mechanical tests and thermo-gravimetric analysis (TGA) were carried out to characterize the nanofibrous membranes. The results showed that the blending of RSSP greatly decreased the nanofibers' average diameter, enhanced the hydrophilicity, changed the microstructure and thermal properties, and could enable tailored mechanical properties of the nanofibrous membranes. Among the blended membranes, the PLCL/RSSP (75/25) membrane was chosen for further investigation on biocompatibility. The results of hemolysis assays and for proliferation of human foreskin fibroblast cells (hFFCs) confirmed the membranes potential use as skin-regeneration scaffolds. Subsequent culture of mouse embryonic fibroblast cells (NIH-3T3) demonstrated the feasibility of the blended membranes as a human epidermal growth factor (hEGF) delivery matrix. The PLCL/RSSP (75/25) membrane possessed good properties comparable to those of human skin with high biocompatibility and the ability of hEGF delivery. Further studies can be carried out on such membranes with chemical or genetic modifications to make better scaffolds for skin regeneration.
Collapse
|
36
|
García-Cerna S, Sánchez-Pacheco U, Meneses-Acosta A, Rojas-García J, Campillo-Illanes B, Segura-González D, Peña-Malacara C. Evaluation of Poly-3-Hydroxybutyrate (P3HB) Scaffolds Used for Epidermal Cells Growth as Potential Biomatrix. Polymers (Basel) 2022; 14:polym14194021. [PMID: 36235969 PMCID: PMC9572615 DOI: 10.3390/polym14194021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Advances in tissue engineering have made possible the construction of organs and tissues with the use of biomaterials and cells. Three important elements are considered: a specific cell culture, an adequate environment, and a scaffold. The present study aimed to develop P3HB scaffolds by 3D printing and evaluate their biocompatibility with HaCaT epidermal cells, as a potential model that allows the formation of functional tissue. By using a method of extraction and purification with ethanol and acetone, a biopolymer having suitable properties for use as a tissue support was obtained. This polymer exhibited a higher molecular weight (1500 kDa) and lower contact angle (less than 90°) compared to the material obtained using the conventional method. The biocompatibility analysis reveals that the scaffold obtained using the ethanol–acetone method and produced by 3D printing without pores was not cytotoxic, did not self-degrade, and allowed high homogenous cell proliferation of HaCaT cells. In summary, it is possible to conclude that the P3HB scaffold obtained by 3D printing and a simplified extraction method is a suitable support for the homogeneous development of HaCaT keratinocyte cell lineage, which would allow the evaluation of this material to be used as a biomatrix for tissue engineering.
Collapse
Affiliation(s)
- Sandra García-Cerna
- Laboratorio 7 de la Facultad de Farmacia, Universidad Autonoma del Estado de Morelos, Avenida Universidad No. 1001, Chamilpa, Cuernavaca C.P. 62209, Morelos, Mexico
| | - Uriel Sánchez-Pacheco
- Laboratorio 7 de la Facultad de Farmacia, Universidad Autonoma del Estado de Morelos, Avenida Universidad No. 1001, Chamilpa, Cuernavaca C.P. 62209, Morelos, Mexico
| | - Angélica Meneses-Acosta
- Laboratorio 7 de la Facultad de Farmacia, Universidad Autonoma del Estado de Morelos, Avenida Universidad No. 1001, Chamilpa, Cuernavaca C.P. 62209, Morelos, Mexico
| | - José Rojas-García
- CIATEQ A. C. Plasticos y Materiales Avanzados. Av. Del Retablo 150, Queretaro C.P. 76150, Queretaro, Mexico
| | - Bernardo Campillo-Illanes
- Instituto de Ciencias Fisicas, Universidad Nacional Autonoma de Mexico, Avenida Universidad S/N, Chamilpa, Cuernavaca C.P. 62210, Morelos, Mexico
| | - Daniel Segura-González
- Departamento de Ingenieria Celular y Biocatalisis, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, Apdo. Post. 510-3, Cuernavaca C.P. 62250, Morelos, Mexico
| | - Carlos Peña-Malacara
- Departamento de Ingenieria Celular y Biocatalisis, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, Apdo. Post. 510-3, Cuernavaca C.P. 62250, Morelos, Mexico
- Correspondence: ; Tel.: +52-(777)-329-1617
| |
Collapse
|
37
|
Development and Characterization of Zein/Ag-Sr Doped Mesoporous Bioactive Glass Nanoparticles Coatings for Biomedical Applications. Bioengineering (Basel) 2022; 9:bioengineering9080367. [PMID: 36004892 PMCID: PMC9404864 DOI: 10.3390/bioengineering9080367] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 12/04/2022] Open
Abstract
Implants are used to replace damaged biological structures in human body. Although stainless steel (SS) is a well-known implant material, corrosion of SS implants leads to the release of toxic metallic ions, which produce harmful effects in human body. To prevent material degradation and its harmful repercussions, these implanted materials are subjected to biocompatible coatings. Polymeric coatings play a vital role in enhancing the mechanical and biological integrity of the implanted devices. Zein is a natural protein extracted from corn and is known to have good biocompatibility and biodegradability. In this study, zein/Ag-Sr doped mesoporous bioactive glass nanoparticles (Ag-Sr MBGNs) were deposited on SS substrates via electrophoretic deposition (EPD) at different parameters. Ag and Sr ions were added to impart antibacterial and osteogenic properties to the coatings, respectively. In order to examine the surface morphology of coatings, optical microscopy and scanning electron microscopy (SEM) were performed. To analyze mechanical strength, a pencil scratch test, bend test, and corrosion and wear tests were conducted on zein/Ag-Sr doped MBGN coatings. The results show good adhesion strength, wettability, corrosion, and wear resistance for zein/Ag-Sr doped MBGN coatings as compared to bare SS substrate. Thus, good mechanical and biological properties were observed for zein/Ag-Sr doped MBGN coatings. Results suggested these zein/Ag-Sr MBGNs coatings have great potential in bone regeneration applications.
Collapse
|
38
|
Engineered polymer matrix novel biocompatible materials decorated with eucalyptus oil and zinc nitrate with superior mechanical and bone forming abilities. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
39
|
A Simple Replica Method as the Way to Obtain a Morphologically and Mechanically Bone-like Iron-Based Biodegradable Material. MATERIALS 2022; 15:ma15134552. [PMID: 35806677 PMCID: PMC9267498 DOI: 10.3390/ma15134552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/04/2022]
Abstract
Porous iron-based scaffolds were prepared by the simple replica method using polyurethane foam as a template and applying the sintering process in a tube furnace. Their surface morphology was characterized using scanning electron microscopy (SEM) and phase homogeneity was confirmed using X-ray diffraction (XRD). Corrosion behavior was determined using immersion and potentiodynamic polarization methods in phosphate buffered saline (PBS). The surface energy was calculated by studying the changes of enthalpy of calorimetric immersion. A preliminary biological test was also carried out and was done using the albumin adsorption procedure. Results of our work showed that in using the simple replica method it is possible to obtain iron biomaterial with morphology and mechanical properties almost identical to bones, and possessing adequate wettability, which gives the potential to use this material as biomaterial for scaffolds in orthopedics.
Collapse
|
40
|
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: 1.5] [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
|
41
|
Lackington WA, Fleyshman L, Schweizer P, Elbs-Glatz Y, Guimond S, Rottmar M. The response of soft tissue cells to Ti implants is modulated by blood-implant interactions. Mater Today Bio 2022; 15:100303. [PMID: 35655805 PMCID: PMC9151735 DOI: 10.1016/j.mtbio.2022.100303] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/10/2022] [Accepted: 05/18/2022] [Indexed: 11/18/2022] Open
Abstract
Titanium-based dental implants have been highly optimized to enhance osseointegration, but little attention has been given to the soft tissue-implant interface, despite being a major contributor to long term implant stability. This is strongly linked to a lack of model systems that enable the reliable evaluation of soft tissue-implant interactions. Current in vitro platforms to assess these interactions are very simplistic, thus suffering from limited biological relevance and sensitivity to varying implant surface properties. The aim of this study was to investigate how blood-implant interactions affect downstream responses of different soft tissue cells to implants in vitro, thus taking into account not only the early events of blood coagulation upon implantation, but also the multicellular nature of soft tissue. For this, three surfaces (smooth and hydrophobic; rough and hydrophobic; rough and hydrophilic with nanostructures), which reflect a wide range of implant surface properties, were used to study blood-material interactions as well as cell-material interactions in the presence and absence of blood. Rough surfaces stimulated denser fibrin network formation compared to smooth surfaces and hydrophilicity accelerated the rate of blood coagulation compared to hydrophobic surfaces. In the absence of blood, smooth surfaces supported enhanced attachment of human gingival fibroblasts and keratinocytes, but limited changes in gene expression and cytokine production were observed between surfaces. In the presence of blood, rough surfaces supported enhanced fibroblast attachment and stimulated a stronger anti-inflammatory response from macrophage-like cells than smooth surfaces, but only smooth surfaces were capable of supporting long-term keratinocyte attachment and formation of a layer of epithelial cells. These findings indicate that surface properties not only govern blood-implant interactions, but that this can in turn also significantly modulate subsequent soft tissue cell-implant interactions.
Collapse
Affiliation(s)
- William A. Lackington
- Biointerfaces Laboratory, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Lada Fleyshman
- Biointerfaces Laboratory, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Peter Schweizer
- Mechanics of Materials & Nanostructures Lab, Empa, Swiss Federal Laboratories for Materials Science and Technology, Thun, Switzerland
| | - Yvonne Elbs-Glatz
- Biointerfaces Laboratory, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Stefanie Guimond
- Biointerfaces Laboratory, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Markus Rottmar
- Biointerfaces Laboratory, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| |
Collapse
|
42
|
Hu Q, Wu J, Zhang H, Dong W, Gu Y, Liu S. Designing Double-Layer Multi-Material Composite Patch Scaffold with Adhesion Resistance for Hernia Repair. Macromol Biosci 2022; 22:e2100510. [PMID: 35471592 DOI: 10.1002/mabi.202100510] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/12/2022] [Indexed: 11/10/2022]
Abstract
Hernia repair mesh is associated with a number of complications, including adhesions and limited mobility, due to insufficient mechanical strength and non-resorbability. Among them, visceral adhesions are one of the most serious complications of patch repair. In this study, a degradable patch with an anti-adhesive layer was prepared for hernia repair by 3D printing and electrospinning techniques using polycaprolactone (PCL), polyvinyl alcohol (PVA), and soybean peptide (SP). The study into the physicochemical properties of the patch was found that it had adequate mechanical strength requirements (16 N cm-1 ) and large elongation at break, which were superior than commercial polypropylene (PP) patches. In vivo and in vitro experiments showed that human umbilical vein endothelial cells (HUVECs) proliferated well on composite patches, and showed excellent biocompatibility with the host and little adhesion through a rat abdominal wall defect model. In conclusion, the results of this study show that composite patch can effectively reduce the occurrence of adhesions, while the addition of SP in the patch further enhances its biocompatibility. We believe that a regenerative biological patch with great potential in hernia repair provides a new strategy for the development of new biomimetic biodegradable patches. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Qingxi Hu
- Rapid Manufacturing Engineering Center, School of Mechatronical Engineering and Automation, Shanghai University, Shanghai, 200444, China.,Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, Shanghai University, Shanghai, 200072, China.,National Demonstration Center for Experimental Engineering Training Education, Shanghai University, Shanghai, 200444, China
| | - Junjie Wu
- Rapid Manufacturing Engineering Center, School of Mechatronical Engineering and Automation, Shanghai University, Shanghai, 200444, China
| | - Haiguang Zhang
- Rapid Manufacturing Engineering Center, School of Mechatronical Engineering and Automation, Shanghai University, Shanghai, 200444, China.,Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, Shanghai University, Shanghai, 200072, China.,National Demonstration Center for Experimental Engineering Training Education, Shanghai University, Shanghai, 200444, China
| | - Wenpei Dong
- Department of General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Yan Gu
- Department of General Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Suihong Liu
- Rapid Manufacturing Engineering Center, School of Mechatronical Engineering and Automation, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
43
|
Lee M, Chun D, Park S, Choi G, Kim Y, Kang SJ, Im SG. Engineering of Surface Energy of Cell-Culture Platform to Enhance the Growth and Differentiation of Dendritic Cells via Vapor-Phase Synthesized Functional Polymer Films. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106648. [PMID: 35297560 DOI: 10.1002/smll.202106648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/23/2022] [Indexed: 06/14/2023]
Abstract
Although the dendritic cell (DC)-based modulation of immune responses has emerged as a promising therapeutic strategy for tumors, infections, and autoimmune diseases, basic research and therapeutic applications of DCs are hampered by expensive growth factors and sophisticated culture procedures. Furthermore, the platform to drive the differentiation of a certain DC subset without any additional biochemical manipulations has not yet been developed. Here, five types of polymer films with different hydrophobicity via an initiated chemical vapor deposition (iCVD) process to modulate the interactions related to cell-substrate adhesion are introduced. Especially, poly(cyclohexyl methacrylate) (pCHMA) substantially enhances the expansion and differentiation of conventional type 1 DCs (cDC1s), the prime DC subset for antigen cross-presentation, and CD8+ T cell activation, by 4.8-fold compared to the conventional protocol. The cDC1s generated from the pCHMA-coated plates retain the bona fide DC functions including the expression of co-stimulatory molecules, cytokine secretion, antigen uptake and processing, T cell activation, and induction of antitumor immune responses. To the authors' knowledge, this is the first report highlighting that the modulation of surface hydrophobicity of the culture plate can be an incisive approach to construct an advanced DC culture platform with high efficiency, which potentially facilitates basic research and the development of immunotherapy employing DCs.
Collapse
Affiliation(s)
- Minseok Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Dongmin Chun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Seonghyeon Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Goro Choi
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Yesol Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Suk-Jo Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- KAIST Institute For NanoCentury (KINC), Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| |
Collapse
|
44
|
Varshney N, Sahi AK, Poddar S, Vishwakarma NK, Kavimandan G, Prakash A, Mahto SK. Freeze-Thaw-Induced Physically Cross-linked Superabsorbent Polyvinyl Alcohol/Soy Protein Isolate Hydrogels for Skin Wound Dressing: In Vitro and In Vivo Characterization. ACS APPLIED MATERIALS & INTERFACES 2022; 14:14033-14048. [PMID: 35312269 DOI: 10.1021/acsami.1c23024] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In this work, polyvinyl alcohol (PVA)- and soy protein isolate (SPI)-based scaffolds were prepared by physical cross-linking using the freeze-thaw method. The PVA/SPI ratio was varied to examine the individual effects of the two constituents. The physicochemical properties of the fabricated scaffolds were analyzed through Fourier transform infrared spectroscopy, scanning electron microscopy, X-ray diffraction, thermogravimetric analysis, and differential scanning calorimetry. The SPI concentration significantly affected the properties of scaffolds, such as the extent of gelation (%), pore size, porosity, degradation, swelling, and surface wettability. The in vitro degradation of fabricated hydrogels was evaluated in phosphate-buffered saline and lysozyme solution for a duration of 14 days. The in vitro compatibility of prepared hydrogels was evaluated by the MTT assay with NIH-3T3 cells (fibroblast). The water vapor transmission rate (WVTR) assays showed that all hydrogels possessed WVTR values in the range of 2000-2500 g m-2 day-1, which is generally recommended for ideal wound dressing. Overall, the obtained results reveal that the fabricated scaffolds have excellent biocompatibility, mechanical strength, porosity, stability, and degradation rate and thus carry enormous potential for tissue engineering applications. Furthermore, a full-thickness wound healing study performed in rats supported them as a promising wound dressing material.
Collapse
Affiliation(s)
- Neelima Varshney
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Ajay Kumar Sahi
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Suruchi Poddar
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Niraj K Vishwakarma
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Gauri Kavimandan
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Archisha Prakash
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Sanjeev Kumar Mahto
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
- Centre for Advanced Biomaterials and Tissue Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
45
|
Hussain Z, Ding P, Zhang L, Zhang Y, Ullah S, Liu Y, Ullah I, Wang Z, Zheng P, Pei R. Multifaceted tannin crosslinked bioinspired dECM decorated nanofibers modulating cell-scaffold biointerface for tympanic membrane perforation bioengineering. Biomed Mater 2022; 17. [PMID: 35334475 DOI: 10.1088/1748-605x/ac6125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/25/2022] [Indexed: 11/12/2022]
Abstract
Tympanic membrane (TM) perforation leads to persistent otitis media, conductive deafness, and affects life quality. Ointment medication may not be sufficient to treat TM perforation due to the lack of an underlying tissue matrix and thus requiring a scaffold-based application. The engineering of scaffold biointerface close to the matrix via tissue-specific decellularized extracellular matrix (dECM) is crucial in instructing cell behaviour and regulating cell-material interaction in the bioengineering domain. Herein, polycaprolactone (PCL) and TM-dECM (from SD rats) were combined in a different ratio in nanofibrous form using an electrospinning process and crosslinked via tannic acid. The histological and biochemical assays demonstrated that chemical and enzymatic decellularization steps removed cellular/immunogenic contents while retaining collagen and glycosaminoglycan. The morphological, physicochemical, thermomechanical, contact angle, and surface chemical studies demonstrated that the tannin crosslinked PCL/dECM nanofibers fine-tune biophysical and biochemical properties. The multifaceted crosslinked nanofibers hold the tunable distribution of dECM moieties, assembled into a spool-shaped membrane, and could easily insert into perforated sites. The dECM decorated fibers provide a preferable biomimetic matrix for L929 fibroblast adhesion, proliferation, matrix adsorption, and f-actin saturation, which could be crucial for bioengineering. Overall, dECM patterning, surface hydrophilicity, interconnected microporosities, and multifaceted nanofibrous biosystem modulate cell-scaffold performance and could open opportunities to reconstruct TM perforation in a biomimetic fashion.
Collapse
Affiliation(s)
- Zahid Hussain
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Hefei, Anhui, 230026, CHINA
| | - Pi Ding
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Hefei, Anhui, 230026, CHINA
| | - Liwei Zhang
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, Suzhou, Jiangsu, 215123, CHINA
| | - Yajie Zhang
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, Suzhou, Jiangsu, 215123, CHINA
| | - Salim Ullah
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Hefei, Anhui, 230026, CHINA
| | - Yuanshan Liu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Hefei, Anhui, 230026, CHINA
| | - Ismat Ullah
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, Suzhou, Jiangsu, 215123, CHINA
| | - Zhili Wang
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, Suzhou, Jiangsu, 215123, CHINA
| | - Penghui Zheng
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, Suzhou, Jiangsu, 215123, CHINA
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, Suzhou, Jiangsu, 215123, CHINA
| |
Collapse
|
46
|
Gritsch L, Granel H, Charbonnel N, Jallot E, Wittrant Y, Forestier C, Lao J. Tailored therapeutic release from polycaprolactone-silica hybrids for the treatment of osteomyelitis: antibiotic rifampicin and osteogenic silicates. Biomater Sci 2022; 10:1936-1951. [PMID: 35258044 DOI: 10.1039/d1bm02015c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The treatment of osteomyelitis, a destructive inflammatory process caused by bacterial infections to bone tissue, is one of the most critical challenges of orthopedics and bone regenerative medicine. The standard treatment consists of intense antibiotic therapies combined with tissue surgical debridement and the application of a bone defect filler material. Unfortunately, commercially available candidates, such as gentamicin-impregnated polymethylmethacrylate cements, possess very poor pharmacokinetics (i.e., 24 hours burst release) and little to no regenerative potential. Fostered by the intrinsic limitations associated with conventional treatments, alternative osteostimulative biomaterials with local drug delivery have recently started to emerge. In this study, we propose the use of a polycaprolactone-silica sol-gel hybrid material as carrier for the delivery of rifampicin, an RNA-polymerase blocker often used to treat bone infections, and of osteostimulative silicate ions. The release of therapeutic agents from the material is dual, offering two separate and simultaneous effects, and decoupled, meaning that the kinetics of rifampicin and silicate releases are independent from each other. A series of hybrid formulations with increasing amounts of rifampicin was prepared. The antibiotic loading efficacy, as well as the release profiles of rifampicin and silicates were measured. The characterization of cell viability and differentiation of rat primary osteoblasts and antibacterial performance were also performed. Gram-positive Staphylococcus aureus and Gram-negative Pseudomonas aeruginosa and Escherichia coli were selected due to their high occurrence in bone infections. Results confirmed that rifampicin can be successfully loaded within the hybrids without significant degradation and that it is possible to tailor the antibiotic release according to need. Once in a physiological environment, the rapid release of silicates was associated with optimal cell proliferation and the overexpression of osteoblastic differentiation. Simultaneously, rifampicin is delivered over the course of several weeks with significant inhibition of all tested strains. In particular, the materials caused a growth reduction of 7-10 orders of magnitude in Staphylococcus aureus, the major strain responsible for osteomyelitis worldwide. Our data strongly suggest that PCL/silica hybrids are a very promising candidate to develop bone fillers with superior biological performance compared to currently available options. Thanks to their unique synthesis route and their dual tailored release they can promote bone regeneration while reducing the risk of infection for several weeks upon implantation.
Collapse
Affiliation(s)
- Lukas Gritsch
- Laboratoire de Physique de Clermont, UMR CNRS 6533, Université Clermont Auvergne, 4 avenue Blaise Pascal, 63178 Aubière, France.
| | - Henri Granel
- Unité de Nutrition Humaine UMR 1019 INRAE, Université Clermont Auvergne, 28 place Henri-Dunant, 63001 Clermont-Ferrand, France
| | - Nicolas Charbonnel
- Université Clermont Auvergne, CNRS, LMGE, 63000 Clermont-Ferrand, France
| | - Edouard Jallot
- Laboratoire de Physique de Clermont, UMR CNRS 6533, Université Clermont Auvergne, 4 avenue Blaise Pascal, 63178 Aubière, France.
| | - Yohann Wittrant
- Unité de Nutrition Humaine UMR 1019 INRAE, Université Clermont Auvergne, 28 place Henri-Dunant, 63001 Clermont-Ferrand, France
| | | | - Jonathan Lao
- Laboratoire de Physique de Clermont, UMR CNRS 6533, Université Clermont Auvergne, 4 avenue Blaise Pascal, 63178 Aubière, France.
| |
Collapse
|
47
|
Adel IM, ElMeligy MF, Elkasabgy NA. Conventional and Recent Trends of Scaffolds Fabrication: A Superior Mode for Tissue Engineering. Pharmaceutics 2022; 14:306. [DOI: https:/doi.org/10.3390/pharmaceutics14020306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
Abstract
Tissue regeneration is an auto-healing mechanism, initiating immediately following tissue damage to restore normal tissue structure and function. This falls in line with survival instinct being the most dominant instinct for any living organism. Nevertheless, the process is slow and not feasible in all tissues, which led to the emergence of tissue engineering (TE). TE aims at replacing damaged tissues with new ones. To do so, either new tissue is being cultured in vitro and then implanted, or stimulants are implanted into the target site to enhance endogenous tissue formation. Whichever approach is used, a matrix is used to support tissue growth, known as ‘scaffold’. In this review, an overall look at scaffolds fabrication is discussed, starting with design considerations and different biomaterials used. Following, highlights of conventional and advanced fabrication techniques are attentively presented. The future of scaffolds in TE is ever promising, with the likes of nanotechnology being investigated for scaffold integration. The constant evolvement of organoids and biofluidics with the eventual inclusion of organ-on-a-chip in TE has shown a promising prospect of what the technology might lead to. Perhaps the closest technology to market is 4D scaffolds following the successful implementation of 4D printing in other fields.
Collapse
|
48
|
Adel IM, ElMeligy MF, Elkasabgy NA. Conventional and Recent Trends of Scaffolds Fabrication: A Superior Mode for Tissue Engineering. Pharmaceutics 2022; 14:306. [PMID: 35214038 PMCID: PMC8877304 DOI: 10.3390/pharmaceutics14020306] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/16/2022] Open
Abstract
Tissue regeneration is an auto-healing mechanism, initiating immediately following tissue damage to restore normal tissue structure and function. This falls in line with survival instinct being the most dominant instinct for any living organism. Nevertheless, the process is slow and not feasible in all tissues, which led to the emergence of tissue engineering (TE). TE aims at replacing damaged tissues with new ones. To do so, either new tissue is being cultured in vitro and then implanted, or stimulants are implanted into the target site to enhance endogenous tissue formation. Whichever approach is used, a matrix is used to support tissue growth, known as 'scaffold'. In this review, an overall look at scaffolds fabrication is discussed, starting with design considerations and different biomaterials used. Following, highlights of conventional and advanced fabrication techniques are attentively presented. The future of scaffolds in TE is ever promising, with the likes of nanotechnology being investigated for scaffold integration. The constant evolvement of organoids and biofluidics with the eventual inclusion of organ-on-a-chip in TE has shown a promising prospect of what the technology might lead to. Perhaps the closest technology to market is 4D scaffolds following the successful implementation of 4D printing in other fields.
Collapse
Affiliation(s)
- Islam M. Adel
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt; (M.F.E.); (N.A.E.)
| | | | | |
Collapse
|
49
|
Kim HY, Kim BH, Kim MS. Amine Plasma-Polymerization of 3D Polycaprolactone/β-Tricalcium Phosphate Scaffold to Improving Osteogenic Differentiation In Vitro. MATERIALS 2022; 15:ma15010366. [PMID: 35009509 PMCID: PMC8745968 DOI: 10.3390/ma15010366] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/20/2021] [Accepted: 01/02/2022] [Indexed: 01/19/2023]
Abstract
This study aims to investigate the surface characterization and pre-osteoblast biological behaviors on the three-dimensional (3D) poly(ε-caprolactone)/β-tricalcium phosphate (β-TCP) scaffold modified by amine plasma-polymerization. The 3D PCL scaffolds were fabricated using fused deposition modeling (FDM) 3D printing. To improve the pre-osteoblast bioactivity, the 3D PCL scaffold was modified by adding β-TCP nanoparticles, and then scaffold surfaces were modified by amine plasma-polymerization using monomer allylamine (AA) and 1,2-diaminocyclohexane (DACH). After the plasma-polymerization of PCL/β-TCP, surface characterizations such as contact angle, AFM, XRD, and FTIR were evaluated. In addition, mechanical strength was measured by UTM. The pre-osteoblast bioactivities were evaluated by focal adhesion and cell proliferation. Osteogenic differentiation was investigated by ALP activity, Alizarin red staining, and Western blot. Plasma-polymerization induced the increase in hydrophilicity of the surface of the 3D PCL/β-TCP scaffold due to the deposition of amine polymeric thin film on the scaffold surface. Focal adhesion and proliferation of pre-osteoblast improved, and osteogenic differentiation was increased. These results indicated that 3D PCL/β-TCP scaffolds treated with DACH plasma-polymerization showed the highest bioactivity compared to the other samples. We suggest that 3D PCL/β-TCP scaffolds treated with DACH and AA plasma-polymerization can be used as a promising candidate for osteoblast differentiation of pre-osteoblast.
Collapse
Affiliation(s)
- Hee-Yeon Kim
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Korea;
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Korea
| | - Byung-Hoon Kim
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Korea
- Correspondence: (B.-H.K.); (M.-S.K.); Tel.: +82-62-230-6447 (B.-H.K.); +82-62-227-1640 (M.-S.K.)
| | - Myung-Sun Kim
- Department of Orthopaedic Surgery, College of Medicine, Chonnam National University, Gwangju 61469, Korea
- Correspondence: (B.-H.K.); (M.-S.K.); Tel.: +82-62-230-6447 (B.-H.K.); +82-62-227-1640 (M.-S.K.)
| |
Collapse
|
50
|
Nakashima Y, Akaike M, Kounoura M, Hayashi K, Morita K, Oki Y, Nakanishi Y. Evaluation of osteoblastic cell behavior upon culture on titanium substrates photo-functionalized by vacuum ultra-violet treatment. Exp Cell Res 2022; 410:112944. [PMID: 34822810 DOI: 10.1016/j.yexcr.2021.112944] [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: 06/10/2021] [Revised: 11/15/2021] [Accepted: 11/20/2021] [Indexed: 11/20/2022]
Abstract
Photo-functionalization of titanium orthopedic/prosthetic implants using ultraviolet illumination is known to improve osteogenesis. Therefore, in this study, we aimed to examine the influence of vacuum ultraviolet (VUV)-treated titanium surfaces on osteoblast cell adhesion, activity, and differentiation. Osteoblastic cells were cultured on titanium substrates treated with various VUV treatment conditions (0, 6.2, 18.7, and 37.4 J/cm2) and their behavior was evaluated. The results revealed that cell adhesion was increased whereas cell activity and differentiation ability were decreased upon cell culture on VUV-treated substrates. In particular, cell activity and differentiation ability were dramatically suppressed with 18.7 J/cm2 VUV irradiation. Within the limitations of this cell-based experiment, we clarified the VUV treatment conditions in which cell adhesion was improved but cell activity and differentiation ability were suppressed. These results indicate that VUV-treatment can be used to influence cell growth properties and can be used to accelerate or suppress cell differentiation on implant substrates.
Collapse
Affiliation(s)
- Yuta Nakashima
- Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami Chuo-ku, Kumamoto, 860-8555, Japan; International Research Organization for Advanced Science & Technology, Kumamoto University, 2-39-1 Kurokami Chuo-ku, Kumamoto, 860-8555, Japan; Institute of Industrial Nanomaterials, 2-39-1 Kurokami Chuo-ku, Kumamoto, 860-8555, Japan.
| | - Mami Akaike
- Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-8555, Japan
| | - Masaki Kounoura
- Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-8555, Japan
| | - Keita Hayashi
- Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-8555, Japan
| | - Kinichi Morita
- Department of I&E Visionaries, Kyusyu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Yuji Oki
- Department of I&E Visionaries, Kyusyu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Yoshitaka Nakanishi
- Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami Chuo-ku, Kumamoto, 860-8555, Japan; Institute of Industrial Nanomaterials, 2-39-1 Kurokami Chuo-ku, Kumamoto, 860-8555, Japan
| |
Collapse
|