1
|
Chen C, Xie Z, Yang S, Wu H, Bi Z, Zhang Q, Xiao Y. Machine Learning Approach to Investigating Macrophage Polarization on Various Titanium Surface Characteristics. BME FRONTIERS 2025; 6:0100. [PMID: 40012846 PMCID: PMC11862448 DOI: 10.34133/bmef.0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
Objective: Current laboratory studies on the effect of biomaterial properties on immune reactions are incomplete and based on a single or a few combination features of the biomaterial design. This study utilizes intelligent prediction models to explore the key features of titanium implant materials in macrophage polarization. Impact Statement: This pilot study provided some insights into the great potential of machine learning in exploring bone immunomodulatory biomaterials. Introduction: Titanium materials are commonly utilized as bone replacement materials to treat missing teeth and bone defects. The immune response caused by implant materials after implantation in the body has a double-edged sword effect on osseointegration. Macrophage polarization has been extensively explored to understand early material-mediated immunomodulation. However, understanding of implant material surface properties and immunoregulations remains limited due to current experimental settings, which are based on trial-by-trial approaches. Artificial intelligence, with its capacity to analyze large datasets, can help explore complex material-cell interactions. Methods: In this study, the effect of titanium surface properties on macrophage polarization was analyzed using intelligent prediction models, including random forest, extreme gradient boosting, and multilayer perceptron. Additionally, data extracted from the newly published literature were further input into the trained models to validate their performance. Results: The analysis identified "cell seeding density", "contact angle", and "roughness" as the most important features regulating interleukin 10 and tumor necrosis factor α secretion. Additionally, the predicted interleukin 10 levels closely matched the experimental results from newly published literature, while the tumor necrosis factor α predictions exhibited consistent trends. Conclusion: The polarization response of macrophages seeded on titanium materials is influenced by multiple factors, and artificial intelligence can assist in extracting the key features of implant materials for immunoregulation.
Collapse
Affiliation(s)
- Changzhong Chen
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine,
Guangzhou Medical University, Guangzhou 510182, China
| | - Zhenhuan Xie
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine,
Guangzhou Medical University, Guangzhou 510182, China
| | - Songyu Yang
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine,
Guangzhou Medical University, Guangzhou 510182, China
| | - Haitong Wu
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine,
Guangzhou Medical University, Guangzhou 510182, China
| | - Zhisheng Bi
- School of Basic Medical Sciences,
Guangzhou Medical University, Guangzhou 511436, China
| | - Qing Zhang
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine,
Guangzhou Medical University, Guangzhou 510182, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences,
Vrije Universiteit Amsterdam, 1081 BT Amsterdam, The Netherlands
| | - Yin Xiao
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine,
Guangzhou Medical University, Guangzhou 510182, China
- School of Medicine and Dentistry & Institute for Biomedicine and Glycomics,
Griffith University, Gold Coast, QLD 4222, Australia
| |
Collapse
|
2
|
Wang Q, Gao Y, Chen Y, Wang X, Pei Q, Zhang T, Wang C, Pan J. Synergistic Enhancement of Antibacterial and Osteo-Immunomodulatory Activities of Titanium Implants via Dual-Responsive Multifunctional Surfaces. Adv Healthc Mater 2025; 14:e2404260. [PMID: 39690750 DOI: 10.1002/adhm.202404260] [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: 10/31/2024] [Revised: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Bone implant-associated infections and inflammations, primarily caused by bacteria colonization, frequently result in unsuccessful procedures and pose significant health risks to patients. To mitigate these challenges, the development of engineered implants with spatiotemporal regulation capabilities, designed to inhibit bacterial survival and modulate immune responses in the early stage, while promoting bone defect healing in the late stage is proposed. The implants are functionalized with ε-poly-l-lysine-phenylboronic acid (PP) via dynamic boronic ester bonds, which facilitate its release through a reactive oxygen species (ROS) and pH-responsive strategy, thereby establishing an antibacterial microenvironment on and around the implants. Additionally, the dynamic metal coordination interaction facilitates the loading and sustained release of Sr2+ under an acidic environment, providing immunomodulatory and osteogenic effects. The ROS/pH-responsive feature, coupled with the implant-bone tissue integration process, affords precise spatiotemporal regulation of the Ti-TA-Sr-PP implants. This strategy represents a promising approach for the preparation of advanced bone implants.
Collapse
Affiliation(s)
- Qing Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ya Gao
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yanzheng Chen
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Xuan Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qingguo Pei
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Taiyu Zhang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Changping Wang
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jinsong Pan
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
3
|
de Almeida GS, Suter LC, Pinto TS, Carra MGJ, da Silva Feltran G, de Moraes JF, Corrêa DRN, Saeki MJ, Lisboa-Filho PN, Zambuzzi WF. The Biological Properties of Co-Doped Monetite Are Influenced by Thermal Treatment. J Biomed Mater Res B Appl Biomater 2025; 113:e35531. [PMID: 39853958 DOI: 10.1002/jbm.b.35531] [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: 01/09/2024] [Revised: 05/07/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025]
Abstract
Calcium phosphates, notably monetite, are valued biomaterials for bone applications owing to their osteogenic properties and rapid uptake by bone cells. This study investigates the enhancement of these properties through Cobalt doping, which is known to induce hypoxia and promote bone cell differentiation. Heat treatments at 700°C, 900°C, and 1050°C are applied to both monetite and Cobalt-doped monetite, facilitating the development of purer, more crystalline phases with varied particle sizes and optimized cellular responses. Comprehensive physicochemical characterization through XRD, FTIR, Raman, SEM/EDS, and ASAP analyses shows significant phase transformations into pyrophosphate, influencing the materials' structural and functional attributes. When utilized to condition a culture medium for MC3T3-E1 cells, these materials demonstrate non-cytotoxic behavior and provoke specific gene responses associated with the osteoblastic phenotype, angiogenesis, adhesion, and extracellular matrix remodeling. Significantly, non-heat-treated Cobalt-doped Monetite retains properties advantageous for clinical applications such as dental and orthopedic implants, where lower processing temperatures are crucial. This attribute, combined with the material's straightforward production, highlights its practicality and potential cost-effectiveness. Further research is essential to assess the long-term safety and efficacy of these materials in clinical settings. Our findings underscore the promising role of Cobalt-doped Monetite in advancing bone repair and regeneration, setting the stage for future innovations in treating bone lesions, enhancing implant integration, and developing advanced prosthetic coatings within the field of tissue engineering.
Collapse
Affiliation(s)
- Gerson Santos de Almeida
- Bioassays and Cellular Dynamics Lab, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP: São Paulo State University, São Paulo, Brazil
| | - Luisa Camilo Suter
- Bioassays and Cellular Dynamics Lab, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP: São Paulo State University, São Paulo, Brazil
| | - Thais Silva Pinto
- Bioassays and Cellular Dynamics Lab, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP: São Paulo State University, São Paulo, Brazil
| | - Maria Gabriela Jacheto Carra
- Materials and Electrochemistry Lab, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP: São Paulo State University, São Paulo, Brazil
| | - Géorgia da Silva Feltran
- Bioassays and Cellular Dynamics Lab, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP: São Paulo State University, São Paulo, Brazil
| | - Julia Ferreira de Moraes
- Bioassays and Cellular Dynamics Lab, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP: São Paulo State University, São Paulo, Brazil
| | - Diego Rafael Nespeque Corrêa
- Anelasticity and Biomaterials Lab, Department of Physics and Meteorology, School of Sciences, São Paulo State University - UNESP, São Paulo, Brazil
| | - Margarida Juri Saeki
- Materials and Electrochemistry Lab, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP: São Paulo State University, São Paulo, Brazil
| | | | - Willian Fernando Zambuzzi
- Bioassays and Cellular Dynamics Lab, Department of Chemical and Biological Sciences, Institute of Biosciences, UNESP: São Paulo State University, São Paulo, Brazil
| |
Collapse
|
4
|
Dos Santos Sanches N, Panahipour L, Wang L, Imani A, Marchiolli CL, Cervantes LCC, Stein MCRV, Berton SA, Souza FÁ, Okamoto R, Júnior IRG, Gruber R. Cytokine expression of soft tissue cells cultured with titanium discs and their respective supernatants in vitro. Clin Oral Investig 2025; 29:62. [PMID: 39809969 PMCID: PMC11732886 DOI: 10.1007/s00784-024-06123-1] [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: 11/11/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025]
Abstract
OBJECTIVE Titanium surface modifications improve osseointegration in dental and orthopedic implants. However, soft tissue cells can also reach the implant surface in immediate loading protocols. While previous research focused on osteogenic cells, the early response of soft tissue cells still needs to be better understood. MATERIAL AND METHODS We have established a bioassay to this aim where human gingival fibroblasts, HSC2 oral squamous carcinoma cells, and murine bone marrow cells were cultured onto titanium discs or exposed to the respective supernatants for overnight. Modifications were double acid-etching (SLA), and coating with simulated body fluid (SBF) with or without odanacatib (ODN), a selective cathepsin K inhibitor reducing bone resorption. RESULTS Our findings indicate that direct contact with titanium discs, with all surface modifications, slightly reduces cell viability. Growing gingival fibroblasts on discs consistently showed a trend toward increased IL8 expression. In HSC2 cells, this setting significantly increased IL1 and IL8 expression, confirmed by the immunoassay. Murine bone marrow macrophages also showed an increase in IL1 and IL6 expressions. Supernatants of the respective discs failed to cause these changes. Although ODN coating inhibited cathepsin K, osteoclastogenesis remained unchanged. CONCLUSIONS These findings suggest that titanium discs do not provide a favorable in vitro surface for oral soft tissue cells as they lose viability and respond with a moderately increased expression of inflammatory cytokines. CLINICAL RELEVANCE The soft tissue surrounding a dental implant can impact rehabilitation success. Understanding how soft tissue cells respond to titanium surface is potentially relevant to understand clinical outcomes.
Collapse
Affiliation(s)
- Natália Dos Santos Sanches
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090, Vienna, Austria
- Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araçatuba, 16015-050Sao Paulo, , Brazil
| | - Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090, Vienna, Austria
| | - Lei Wang
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090, Vienna, Austria
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Atefe Imani
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090, Vienna, Austria
| | - Caroline Liberato Marchiolli
- Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araçatuba, 16015-050Sao Paulo, , Brazil
| | - Lara Cristina Cunha Cervantes
- Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araçatuba, 16015-050Sao Paulo, , Brazil
- University of Brazil, 15600-000, São Paulo, Fernandópolis, Brazil
| | - Maria Cristina Ruiz Voms Stein
- Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araçatuba, 16015-050Sao Paulo, , Brazil
| | - Sara Alves Berton
- Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araçatuba, 16015-050Sao Paulo, , Brazil
| | - Francisley Ávila Souza
- Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araçatuba, 16015-050Sao Paulo, , Brazil
| | - Roberta Okamoto
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, 16015-050Sao Paulo, , Brazil
| | - Idelmo Rangel Garcia Júnior
- Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araçatuba, 16015-050Sao Paulo, , Brazil
| | - 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
|
5
|
Zheng Z, Gan S, Yang S, Hou C, Zhu Z, Wang H, Yu D, Qian Z, Xu HHK, Chen W. Enhanced surface hydrophilicity improves osseointegration of titanium implants via integrin-mediated osteoimmunomodulation. J Mater Chem B 2025; 13:496-510. [PMID: 39688175 DOI: 10.1039/d4tb02360a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Titanium (Ti) implants have become widespread especially in dentistry and orthopedics, where macrophage-driven osteoimmunomodulation is crucial to their success. Hydrophilic modification of Ti represents a promising strategy to enhance its immune and osteogenic responses. Herein, the osteoimmunomodulatory performance and integrin-mediated mechanism of novel non-thermal atmospheric plasma (NTAP) treatment to induce a hydrophilic Ti were investigated for the first time. Compared to a hydrophobic surface, NTAP-modified Ti possessed a 3-fold increase of pro-healing M2 macrophage makers, and the doubled osteogenic differentiation of mesenchymal stem cells was demonstrated in this immune microenvironment, thus improving early osseointegration. Mechanistically, the ameliorative osteoimmunomodulatory properties of NTAP were attributed to its positive and negative modulation in macrophages' integrin β1 or β2, and the subsequent FAK-PI3K/Akt or NF-κB signaling pathway. Collectively, this study highlighted the role of integrins and related signaling pathways in hydrophilic implant-caused macrophage polarization, therefore inventively unveiling the underlying mechanism of NTAP-enhanced osteoimmunomodulation. Furthermore, it established a robust theoretical foundation for the clinical application of this cost-effective, versatile, and transformation-valuable surface engineering strategy for the development of next-generation Ti implants.
Collapse
Affiliation(s)
- Zheng Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Shuaiqi Gan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Shuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Chuping Hou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhimin Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Hang Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Deping Yu
- School of Mechanical Engineering, Sichuan University, Chengdu 610041, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, Sichuan, China
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Wenchuan Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
6
|
Soe ZC, Wahyudi R, Mattheos N, Lertpimonchai A, Everts V, Tompkins KA, Osathanon T, Limjeerajarus CN, Limjeerajarus N. Application of nanoparticles as surface modifiers of dental implants for revascularization/regeneration of bone. BMC Oral Health 2024; 24:1175. [PMID: 39367468 PMCID: PMC11451240 DOI: 10.1186/s12903-024-04966-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Osseointegrated dental implants are widely established as a first-choice treatment for the replacement of missing teeth. Clinical outcomes are however often compromised by short or longer-term biological complications and pathologies. Nanoparticle-coated materials represent a very active research area with the potential to enhance clinical outcomes and reduce complications of implant therapy. This scoping review aimed to summarize current research on various types of nanoparticles (NPs) used as surface modifiers of dental implants and their potential to promote biological and clinical outcomes. METHODS A systematic electronic search was conducted in SCOPUS, PubMed and Google Scholar aiming to identify in vivo, in situ, or in vitro studies published between 2014 and 2024. Inclusion and exclusion criteria were determined and were described in the methods section. RESULTS A total of 169 articles (44 original papers from Scopus and PubMed, and 125 articles from Google Scholar) were identified by the electronic search. Finally, 30 studies fit the inclusion criteria and were further used in this review. The findings from the selected papers suggest that nanoparticle-coated dental implants show promising results in enhancing bone regeneration and promoting angiogenesis around the implant site. These effects are due to the unique physicochemical properties of nanoparticle-coated implants and the controlled release of bioactive molecules from nanoparticle-modified surfaces. CONCLUSION Nanoscale modifications displayed unique properties which could significantly enhance the properties of dental implants and further accelerate revascularization, and osseointegration while facilitating early implant loading. Yet, since many of these findings were based on in-vitro/in-situ systems, further research is required before such technology reaches clinical application.
Collapse
Affiliation(s)
- Zar Chi Soe
- Faculty of Dentistry, Graduate Program in Oral Biology, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Rahman Wahyudi
- Faculty of Dentistry, Graduate Program in Oral Biology, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nikos Mattheos
- Department of Dental Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Attawood Lertpimonchai
- Department of Periodontology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Vincent Everts
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
- Office of Research Affairs, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kevin A Tompkins
- Office of Research Affairs, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chalida Nakalekha Limjeerajarus
- Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence in Genomics and Precision Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | | |
Collapse
|
7
|
Gil-Cantero S, Iorio F, Unalan I, Kurtuldu F, Künig S, Wenhardt C, Pinnaro V, Aigner-Radakovics K, Steinberger P, Boccaccini AR, Stöckl J. Impact of morphological features and chemical composition of tendon biomimetic scaffolds on immune recognition via Toll-like receptors. Biomater Sci 2024; 12:4695-4712. [PMID: 39082440 DOI: 10.1039/d4bm00147h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Tendinopathies are a major worldwide clinical problem. The development of tendon biomimetic scaffolds is considered a promising, therapeutic approach. However, to be clinically effective, scaffolds should avoid immunological recognition. It has been well described that scaffolds composed of aligned fibers lead to a better tenocyte differentiation, vitality, proliferation and motility. However, little has been studied regarding the impact of fiber spatial distribution on the recognition by immune cells. Additionally, it has been suggested that higher hydrophilicity would reduce their immune recognition. Herein, polycaprolactone (PCL)-hyaluronic acid (HA)-based electrospun scaffolds were generated with different fiber diameters (in the nano- and micro-scales) and orientations as well as different grades of wettability and the impact of these properties on immunological recognition has been assessed, by means of Toll-like receptor (TLR) reporter cells. Our results showed that TLR 2/1 and TLR 2/6 were not triggered by the scaffolds. In addition, the TLR 4 signalling pathway seems to be triggered to a greater extent by higher PCL and HA concentrations, but the alignment of the fibers prevents the triggering of this receptor. Taken together, TLR reporter cells were shown to be a useful and effective tool to study the potential of scaffolds to induce immune responses and the results obtained can be used to inform the design of fibrous scaffolds for tendon repair.
Collapse
Affiliation(s)
- Sara Gil-Cantero
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Germany.
| | - Francesco Iorio
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Germany.
| | - Irem Unalan
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Germany.
| | - Fatih Kurtuldu
- FunGlass, Alexander Dubček University of Trenčín, Trenčín, Slovakia
| | - Sarojinidevi Künig
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
| | - Claus Wenhardt
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
| | - Veronica Pinnaro
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
| | - Katharina Aigner-Radakovics
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
| | - Peter Steinberger
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Germany.
| | - Johannes Stöckl
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
8
|
ten Brink T, Damanik F, Rotmans JI, Moroni L. Unraveling and Harnessing the Immune Response at the Cell-Biomaterial Interface for Tissue Engineering Purposes. Adv Healthc Mater 2024; 13:e2301939. [PMID: 38217464 PMCID: PMC11468937 DOI: 10.1002/adhm.202301939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/14/2023] [Indexed: 01/15/2024]
Abstract
Biomaterials are defined as "engineered materials" and include a range of natural and synthetic products, designed for their introduction into and interaction with living tissues. Biomaterials are considered prominent tools in regenerative medicine that support the restoration of tissue defects and retain physiologic functionality. Although commonly used in the medical field, these constructs are inherently foreign toward the host and induce an immune response at the material-tissue interface, defined as the foreign body response (FBR). A strong connection between the foreign body response and tissue regeneration is suggested, in which an appropriate amount of immune response and macrophage polarization is necessary to trigger autologous tissue formation. Recent developments in this field have led to the characterization of immunomodulatory traits that optimizes bioactivity, the integration of biomaterials and determines the fate of tissue regeneration. This review addresses a variety of aspects that are involved in steering the inflammatory response, including immune cell interactions, physical characteristics, biochemical cues, and metabolomics. Harnessing the advancing knowledge of the FBR allows for the optimization of biomaterial-based implants, aiming to prevent damage of the implant, improve natural regeneration, and provide the tools for an efficient and successful in vivo implantation.
Collapse
Affiliation(s)
- Tim ten Brink
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Febriyani Damanik
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Joris I. Rotmans
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333ZAThe Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| |
Collapse
|
9
|
Chato-Astrain J, Toledano-Osorio M, Alaminos M, Toledano M, Sanz M, Osorio R. Effect of functionalized titanium particles with dexamethasone-loaded nanospheres on macrophage polarization and activity. Dent Mater 2024; 40:66-79. [PMID: 37914549 DOI: 10.1016/j.dental.2023.10.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/09/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE The aim of this study was to determine the effect of titanium micro particles (TiP) previously functionalized with nanoparticles doped with dexamethasone (Dex) and doxycycline (Dox), on macrophage polarization and activity. METHODS Macrophages RAW264.7 were cultured in the presence TiP loaded with dexamethasone -NPs (Dex)- and doxycycline -NPs (Dox)-, and as control, TiP with or without doped NPs. Cells were tested with and without previous bacterial lipopolysaccharide endotoxin (LPS) stimulation. Their morphology, proliferation, cytotoxicity, phenotypic change, and cytokines release were assessed by LIVE/DEAD, DNA release, metabolic activity, brightfield and scanning electron microscopy. The test Kruskall-Wallis was used for comparisons, while the cytokine expression profiles were examined by hierarchical clustering (p < 0.05). RESULTS Upon exposure with TiP macrophages were activated and polarized to M1, but without depicting cytotoxic effects. The particles were phagocytised, and vacuolized. When exposed to functionalised TiP with NPs(Dex) and NPs(Dox), the ratio M1/M2 was up to forty times lower compared to TiP alone. When exposed to LPS, TiP reduced cell viability in half. Functionalised TiP with NPs(Dex) inhibited the cytokine release exerted by TiP on macrophages. When macrophages were exposed to functionalised TiPs with NPs(Dex) with and without LPS, the effect of TiP on cytokine secretion was inhibited. SIGNIFICANCE Functionalised TiPs with NPs(Dex) and NPs(Dox) may potentially have beneficial effects on modulating titanium and LPS-related inflammatory reactions.
Collapse
Affiliation(s)
- Jesús Chato-Astrain
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria IBS. Granada, Granada, Spain
| | - Manuel Toledano-Osorio
- Dental School, Faculty of Dentistry, University of Granada, Colegio Máximo de Cartuja s/n, Granada 18071, Spain..
| | - Miguel Alaminos
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria IBS. Granada, Granada, Spain
| | - Manuel Toledano
- Instituto de Investigación Biosanitaria IBS. Granada, Granada, Spain; Dental School, Faculty of Dentistry, University of Granada, Colegio Máximo de Cartuja s/n, Granada 18071, Spain
| | - Mariano Sanz
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group. University Complutense of Madrid, Madrid, Spain
| | - Raquel Osorio
- Instituto de Investigación Biosanitaria IBS. Granada, Granada, Spain; Dental School, Faculty of Dentistry, University of Granada, Colegio Máximo de Cartuja s/n, Granada 18071, Spain
| |
Collapse
|
10
|
Zhang S, Yang H, Wang M, Mantovani D, Yang K, Witte F, Tan L, Yue B, Qu X. Immunomodulatory biomaterials against bacterial infections: Progress, challenges, and future perspectives. Innovation (N Y) 2023; 4:100503. [PMID: 37732016 PMCID: PMC10507240 DOI: 10.1016/j.xinn.2023.100503] [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: 05/12/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023] Open
Abstract
Bacterial infectious diseases are one of the leading causes of death worldwide. Even with the use of multiple antibiotic treatment strategies, 4.95 million people died from drug-resistant bacterial infections in 2019. By 2050, the number of deaths will reach 10 million annually. The increasing mortality may be partly due to bacterial heterogeneity in the infection microenvironment, such as drug-resistant bacteria, biofilms, persister cells, intracellular bacteria, and small colony variants. In addition, the complexity of the immune microenvironment at different stages of infection makes biomaterials with direct antimicrobial activity unsatisfactory for the long-term treatment of chronic bacterial infections. The increasing mortality may be partly attributed to the biomaterials failing to modulate the active antimicrobial action of immune cells. Therefore, there is an urgent need for effective alternatives to treat bacterial infections. Accordingly, the development of immunomodulatory antimicrobial biomaterials has recently received considerable interest; however, a comprehensive review of their research progress is lacking. In this review, we focus mainly on the research progress and future perspectives of immunomodulatory antimicrobial biomaterials used at different stages of infection. First, we describe the characteristics of the immune microenvironment in the acute and chronic phases of bacterial infections. Then, we highlight the immunomodulatory strategies for antimicrobial biomaterials at different stages of infection and their corresponding advantages and disadvantages. Moreover, we discuss biomaterial-mediated bacterial vaccines' potential applications and challenges for activating innate and adaptive immune memory. This review will serve as a reference for future studies to develop next-generation immunomodulatory biomaterials and accelerate their translation into clinical practice.
Collapse
Affiliation(s)
- Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Hongtao Yang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Minqi Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Frank Witte
- Department of Prosthodontics, Geriatric Dentistry and Craniomandibular Disorders, Charite Medical University, Assmannshauser Strasse 4–6, 14197 Berlin, Germany
| | - Lili Tan
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| |
Collapse
|
11
|
Nevins M, Chen CY, Parma-Benfenati S, Kim DM. Gas Plasma Treatment Improves Titanium Dental Implant Osseointegration-A Preclinical In Vivo Experimental Study. Bioengineering (Basel) 2023; 10:1181. [PMID: 37892911 PMCID: PMC10604242 DOI: 10.3390/bioengineering10101181] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 09/30/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Recent technological advancements led to the development of various plasma-based technologies for post-packaging modifications. The purpose of the present preclinical in vivo study was to assess the safety and efficacy of a novel chairside nonthermal gas plasma treatment for enhancing osseointegration of titanium implants. Six male mixed foxhounds underwent extraction of mandibular premolars and first molars, and the sockets healed for 42 days. Canine mandibles were randomized to receive either plasma-treated (test) or non-plasma-treated (control) dental implants. A total of 36 implants were placed in six animals, and they were sacrificed at 2 weeks (two animals), 4 weeks (two animals), and 6 weeks (two animals) after the implant surgery. When the radiographic analysis was performed, the changes in bone level were not statistically significant between the two groups at 2 weeks and 4 weeks. The difference became significant at 6 weeks (p = 0.016), indicating more bone loss from baseline to 6 weeks for the control group. The bone-to-implant contact (BIC) appeared to be higher for the test groups at all time points, and the BIC was significantly higher for the test group at 4 weeks (p = 0.046). In conclusion, this study underscored the potential of nonthermal plasma treatment in enhancing implant osseointegration.
Collapse
Affiliation(s)
- Myron Nevins
- Department of Oral Medicine, Infection and Immunity, Division of Periodontology, Harvard School of Dental Medicine, Boston, MA 02115, USA; (M.N.); (C.-Y.C.)
| | - Chia-Yu Chen
- Department of Oral Medicine, Infection and Immunity, Division of Periodontology, Harvard School of Dental Medicine, Boston, MA 02115, USA; (M.N.); (C.-Y.C.)
| | | | - David M. Kim
- Department of Oral Medicine, Infection and Immunity, Division of Periodontology, Harvard School of Dental Medicine, Boston, MA 02115, USA; (M.N.); (C.-Y.C.)
| |
Collapse
|
12
|
Parisi L, Bianchi MG, Ghezzi B, Maurizi E, Macaluso GM, Bussolati O, Lumetti S. Preparation of human primary macrophages to study the polarization from monocyte-derived macrophages to pro- or anti-inflammatory macrophages at biomaterial interface in vitro. J Dent Sci 2023; 18:1630-1637. [PMID: 37799917 PMCID: PMC10547954 DOI: 10.1016/j.jds.2023.01.020] [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: 01/13/2023] [Revised: 01/20/2023] [Indexed: 02/16/2023] Open
Abstract
Background/purpose Testing of dental materials when in contact with innate immune cells has been so far hindered by the lack of proper in vitro models. Human primary monocyte-derived macrophages (MDMs) would be an excellent option to this aim. However, the inability to detach them from the tissue culture plates contrast the possibility to culture them on biomaterials. The goal of the present work is to present and validate an innovative protocol to obtain MDMs from peripheral blood monocytes, and to reseed them in contact with biomaterials without altering their viability and phenotype. Materials and methods We differentiated MDMs on ultra-low attachment tissue culture plastics and recovered them with specific detachment solution in order to be reseeded on a secondary substrate. Therefore, using biological assays (RT-PCR, Western blot, and immunofluorescence) we compared their phenotype to MDMs differentiated on standard culture plates. Results Transferred MDMs keep their differentiated M0 resting state, as well as the ability to be polarized into M1 (pro-inflammatory) or M2 (anti-inflammatory) macrophages. Conclusion These data provide the dental material research community the unprecedented possibility to investigate the immunomodulatory properties of biomaterials for dental application.
Collapse
Affiliation(s)
- Ludovica Parisi
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - Massimiliano Giovanni Bianchi
- Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
- Microbiome Research Hub, Università di Parma, Parma, Italy
| | - Benedetta Ghezzi
- Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
- Centro Universitario di Odontoiatria, Università di Parma, Parma, Italy
- IMEM-CNR, Parma, Italy
| | - Eleonora Maurizi
- Centro Universitario di Odontoiatria, Università di Parma, Parma, Italy
- Centre for Regenerative Medicine “S.Ferrari”, University of Modena and Reggio Emilia, Modena, Italy
| | - Guido Maria Macaluso
- Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
- Centro Universitario di Odontoiatria, Università di Parma, Parma, Italy
- IMEM-CNR, Parma, Italy
| | - Ovidio Bussolati
- Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
- Microbiome Research Hub, Università di Parma, Parma, Italy
| | - Simone Lumetti
- Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
- Centro Universitario di Odontoiatria, Università di Parma, Parma, Italy
- IMEM-CNR, Parma, Italy
| |
Collapse
|
13
|
Soni SS, D'Elia AM, Rodell CB. Control of the post-infarct immune microenvironment through biotherapeutic and biomaterial-based approaches. Drug Deliv Transl Res 2023; 13:1983-2014. [PMID: 36763330 PMCID: PMC9913034 DOI: 10.1007/s13346-023-01290-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2023] [Indexed: 02/11/2023]
Abstract
Ischemic heart failure (IHF) is a leading cause of morbidity and mortality worldwide, for which heart transplantation remains the only definitive treatment. IHF manifests from myocardial infarction (MI) that initiates tissue remodeling processes, mediated by mechanical changes in the tissue (loss of contractility, softening of the myocardium) that are interdependent with cellular mechanisms (cardiomyocyte death, inflammatory response). The early remodeling phase is characterized by robust inflammation that is necessary for tissue debridement and the initiation of repair processes. While later transition toward an immunoregenerative function is desirable, functional reorientation from an inflammatory to reparatory environment is often lacking, trapping the heart in a chronically inflamed state that perpetuates cardiomyocyte death, ventricular dilatation, excess fibrosis, and progressive IHF. Therapies can redirect the immune microenvironment, including biotherapeutic and biomaterial-based approaches. In this review, we outline these existing approaches, with a particular focus on the immunomodulatory effects of therapeutics (small molecule drugs, biomolecules, and cell or cell-derived products). Cardioprotective strategies, often focusing on immunosuppression, have shown promise in pre-clinical and clinical trials. However, immunoregenerative therapies are emerging that often benefit from exacerbating early inflammation. Biomaterials can be used to enhance these therapies as a result of their intrinsic immunomodulatory properties, parallel mechanisms of action (e.g., mechanical restraint), or by enabling cell or tissue-targeted delivery. We further discuss translatability and the continued progress of technologies and procedures that contribute to the bench-to-bedside development of these critically needed treatments.
Collapse
Affiliation(s)
- Shreya S Soni
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Arielle M D'Elia
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA.
| |
Collapse
|
14
|
Gulati K, Ding C, Guo T, Guo H, Yu H, Liu Y. Craniofacial therapy: advanced local therapies from nano-engineered titanium implants to treat craniofacial conditions. Int J Oral Sci 2023; 15:15. [PMID: 36977679 PMCID: PMC10050545 DOI: 10.1038/s41368-023-00220-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/05/2023] [Accepted: 02/28/2023] [Indexed: 03/30/2023] Open
Abstract
Nano-engineering-based tissue regeneration and local therapeutic delivery strategies show significant potential to reduce the health and economic burden associated with craniofacial defects, including traumas and tumours. Critical to the success of such nano-engineered non-resorbable craniofacial implants include load-bearing functioning and survival in complex local trauma conditions. Further, race to invade between multiple cells and pathogens is an important criterion that dictates the fate of the implant. In this pioneering review, we compare the therapeutic efficacy of nano-engineered titanium-based craniofacial implants towards maximised local therapy addressing bone formation/resorption, soft-tissue integration, bacterial infection and cancers/tumours. We present the various strategies to engineer titanium-based craniofacial implants in the macro-, micro- and nano-scales, using topographical, chemical, electrochemical, biological and therapeutic modifications. A particular focus is electrochemically anodised titanium implants with controlled nanotopographies that enable tailored and enhanced bioactivity and local therapeutic release. Next, we review the clinical translation challenges associated with such implants. This review will inform the readers of the latest developments and challenges related to therapeutic nano-engineered craniofacial implants.
Collapse
Affiliation(s)
- Karan Gulati
- The University of Queensland, School of Dentistry, Herston, QLD, Australia
| | - Chengye Ding
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Tianqi Guo
- The University of Queensland, School of Dentistry, Herston, QLD, Australia
| | - Houzuo Guo
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
- Department of Oral Implantology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Huajie Yu
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China.
- Fourth Clinical Division, Peking University School and Hospital of Stomatology, Beijing, China.
| | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China.
| |
Collapse
|
15
|
Wang J, Yuan B, Yin R, Zhang H. Inflammation Responses to Bone Scaffolds under Mechanical Stimuli in Bone Regeneration. J Funct Biomater 2023; 14:jfb14030169. [PMID: 36976093 PMCID: PMC10059255 DOI: 10.3390/jfb14030169] [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: 02/05/2023] [Revised: 03/05/2023] [Accepted: 03/18/2023] [Indexed: 03/29/2023] Open
Abstract
Physical stimuli play an important role in one tissue engineering. Mechanical stimuli, such as ultrasound with cyclic loading, are widely used to promote bone osteogenesis; however, the inflammatory response under physical stimuli has not been well studied. In this paper, the signaling pathways related to inflammatory responses in bone tissue engineering are evaluated, and the application of physical stimulation to promote osteogenesis and its related mechanisms are reviewed in detail; in particular, how physical stimulation alleviates inflammatory responses during transplantation when employing a bone scaffolding strategy is discussed. It is concluded that physical stimulation (e.g., ultrasound and cyclic stress) helps to promote osteogenesis while reducing the inflammatory response. In addition, apart from 2D cell culture, more consideration should be given to the mechanical stimuli applied to 3D scaffolds and the effects of different force moduli while evaluating inflammatory responses. This will facilitate the application of physiotherapy in bone tissue engineering.
Collapse
Affiliation(s)
- Junjie Wang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Bo Yuan
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Ruixue Yin
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Hongbo Zhang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
16
|
Gao Y, Pang Y, Wei S, Han Q, Miao S, Li M, Tian J, Fu C, Wang Z, Zhang X, Yang P, Liu Y. Amyloid-Mediated Nanoarchitectonics with Biomimetic Mineralization of Polyetheretherketone for Enhanced Osseointegration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10426-10440. [PMID: 36791143 DOI: 10.1021/acsami.2c20711] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Polyetheretherketone (PEEK), a widely used implant material, has attracted the attention of scientific researchers because of its bone-matched elastic modulus, radiolucency, and chemical resistance. However, the bioinert chemical properties of PEEK do not promote bone apposition once implanted. In this study, using a phase-transitioned lysozyme (PTL) nanofilm as a sandwiched layer, a robust hydroxyapatite (HAp) coating on PEEK (HAp@PTL@PEEK) is constructed. The PTL nanofilm shows strong adhesion to the PEEK surface and induces biomimetic mineralization to form a compact HAp coating on PEEK in simulated body fluids. This HAp coating not only shares a higher adhesion strength and better stability but can also be applied to implants with complex 3D structures. HAp@PTL@PEEK showed significantly enhanced osteogenic capacity when cultured with rat bone marrow mesenchymal stem cells by promoting initial cell adhesion, proliferation, and osteogenic differentiation in vitro. In vivo evaluations utilizing models of femoral condyle defects and skull defects confirm that the HAp coating substantially augments bone remodeling and osseointegration ability. Compared with the traditional method, our modified method is simpler, more environmentally friendly, and uses less hazardous components. Furthermore, the obtained HAp coating shares a higher adhesion strength to PEEK and a better osteogenic capacity. The study offers a novel method to improve the osseointegration of PEEK-based implants in biointerfaces and tissue engineering.
Collapse
Affiliation(s)
- Yingtao Gao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Yanyun Pang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Shuo Wei
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Qian Han
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Shuting Miao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Min Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Juanhua Tian
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, West Five Road, No. 157, Xi'an 710004, China
| | - Chengyu Fu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Zhengge Wang
- College of Chemical Engineering, Hebei Normal University of Science and Technology, Qinhuangdao 066600, China
| | - Xu Zhang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
- Institute of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Yongchun Liu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
17
|
Modulation of Macrophage Function by Bioactive Wound Dressings with an Emphasis on Extracellular Matrix-Based Scaffolds and Nanofibrous Composites. Pharmaceutics 2023; 15:pharmaceutics15030794. [PMID: 36986655 PMCID: PMC10053223 DOI: 10.3390/pharmaceutics15030794] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Bioactive wound dressings that are capable of regulating the local wound microenvironment have attracted a very large interest in the field of regenerative medicine. Macrophages have many critical roles in normal wound healing, and the dysfunction of macrophages significantly contributes to impaired or non-healing skin wounds. Regulation of macrophage polarization towards an M2 phenotype provides a feasible strategy to enhance chronic wound healing, mainly by promoting the transition of chronic inflammation to the proliferation phase of wound healing, upregulating the level of anti-inflammatory cytokines around the wound area, and stimulating wound angiogenesis and re-epithelialization. Based on this, modulation of macrophage functions by the rational design of bioactive scaffolds has emerged as a promising way to accelerate delayed wound healing. This review outlines current strategies to regulate the response of macrophages using bioactive materials, with an emphasis on extracellular matrix-based scaffolds and nanofibrous composites.
Collapse
|
18
|
Liu Z, Zhu J, Li Z, Liu H, Fu C. Biomaterial scaffolds regulate macrophage activity to accelerate bone regeneration. Front Bioeng Biotechnol 2023; 11:1140393. [PMID: 36815893 PMCID: PMC9932600 DOI: 10.3389/fbioe.2023.1140393] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Bones are important for maintaining motor function and providing support for internal organs. Bone diseases can impose a heavy burden on individuals and society. Although bone has a certain ability to repair itself, it is often difficult to repair itself alone when faced with critical-sized defects, such as severe trauma, surgery, or tumors. There is still a heavy reliance on metal implants and autologous or allogeneic bone grafts for bone defects that are difficult to self-heal. However, these grafts still have problems that are difficult to circumvent, such as metal implants that may require secondary surgical removal, lack of bone graft donors, and immune rejection. The rapid advance in tissue engineering and a better comprehension of the physiological mechanisms of bone regeneration have led to a new focus on promoting endogenous bone self-regeneration through the use of biomaterials as the medium. Although bone regeneration involves a variety of cells and signaling factors, and these complex signaling pathways and mechanisms of interaction have not been fully understood, macrophages undoubtedly play an essential role in bone regeneration. This review summarizes the design strategies that need to be considered for biomaterials to regulate macrophage function in bone regeneration. Subsequently, this review provides an overview of therapeutic strategies for biomaterials to intervene in all stages of bone regeneration by regulating macrophages.
Collapse
Affiliation(s)
- Zongtai Liu
- Department of Spine Surgery, First Hospital of Jilin University, Changchun, China
- Department of Orthopedics, Affiliated Hospital of Beihua University, Jilin, China
| | - Jiabo Zhu
- Department of Orthopedics, Affiliated Hospital of Beihua University, Jilin, China
| | - Zhuohan Li
- Department of Gynecology, Affiliated Hospital of Beihua University, Jilin, China
| | - Hanyan Liu
- Department of Orthopedics, Baicheng Central Hospital, Baicheng, China
| | - Changfeng Fu
- Department of Spine Surgery, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Pitchai M, Ipe D, Tadakamadla S, Hamlet S. Titanium Implant Surface Effects on Adherent Macrophage Phenotype: A Systematic Review. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7314. [PMID: 36295379 PMCID: PMC9609829 DOI: 10.3390/ma15207314] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/06/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Immunomodulatory biomaterials have the potential to stimulate an immune response able to promote constructive and functional tissue remodeling responses as opposed to persistent inflammation and scar tissue formation. As such, the controlled activation of macrophages and modulation of their phenotype through implant surface modification has emerged as a key therapeutic strategy. METHODS Online databases were searched for in vitro studies between January 1991 and June 2020 which examined the effect of titanium implant surface topography on the adherent macrophage phenotype at either the gene or protein level. RESULTS Thirty-nine studies were subsequently included for review. Although there was significant heterogeneity between studies, treatment of titanium surfaces increased the surface roughness or hydrophilicity, and hence increased macrophage attachment but decreased cell spreading. Physical coating of the titanium surface also tended to promote the formation of cell clusters. Titanium and titanium-zirconium alloy with a micro- or nano-scale rough topography combined with a hydrophilic surface chemistry were the most effective surfaces for inducing an anti-inflammatory phenotype in adherent macrophages, as indicated by significant changes in cytokine gene expression and or cytokine secretion profiles. CONCLUSIONS The published data support the hypothesis that incorporation of specific topographical and physiochemical surface modifications to titanium can modulate the phenotypic response of adherent macrophages.
Collapse
Affiliation(s)
| | | | | | - Stephen Hamlet
- School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| |
Collapse
|
20
|
Avery D, Morandini L, Sheakley LS, Shah AH, Bui L, Abaricia JO, Olivares-Navarrete R. Canonical Wnt signaling enhances pro-inflammatory response to titanium by macrophages. Biomaterials 2022; 289:121797. [PMID: 36156410 PMCID: PMC10262842 DOI: 10.1016/j.biomaterials.2022.121797] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
Biomaterial characteristics like surface roughness and wettability can determine the phenotype of macrophages following implantation. We have demonstrated that inhibiting Wnt ligand secretion abolishes macrophage polarization in vitro and in vivo; however, the role of canonical Wnt signaling in macrophage activation in response to physical and chemical biomaterial cues is unknown. The aim of this study was to understand whether canonical Wnt signaling affects the response of macrophages to titanium (Ti) surface roughness or wettability in vitro and in vivo. Activating canonical Wnt signaling increased expression of toll-like receptors and interleukin receptors and secreted pro-inflammatory cytokines and reduced anti-inflammatory cytokines on Ti, regardless of surface properties. Inhibiting canonical Wnt signaling reduced pro-inflammatory cytokines on all Ti surfaces and increased anti-inflammatory cytokines on rough or rough-hydrophilic Ti. In vivo, activating canonical Wnt signaling increased total macrophages, pro-inflammatory macrophages, and T cells and decreased anti-inflammatory macrophages on both smooth and rough-hydrophilic implants. Functionally, canonical Wnt activation increases pro-inflammatory macrophage response to cell and cell-extracellular matrix lysates. These results demonstrate that activating canonical Wnt signaling primes macrophages to a pro-inflammatory phenotype that affects their response to Ti implants in vitro and in vivo.
Collapse
Affiliation(s)
- Derek Avery
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Lais Morandini
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Luke S Sheakley
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Arth H Shah
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Loc Bui
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Jefferson O Abaricia
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
21
|
Ivanovski S, Bartold PM, Huang Y. The role of foreign body response in peri-implantitis: What is the evidence? Periodontol 2000 2022; 90:176-185. [PMID: 35916872 PMCID: PMC9804527 DOI: 10.1111/prd.12456] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Historically, there has been broad consensus that osseointegration represents a homeostasis between a titanium dental implant and the surrounding bone, and that the crestal bone loss characteristic of peri-implantitis is a plaque-induced inflammatory process. However, this notion has been challenged over the past decade by proponents of a theory that considers osseointegration an inflammatory process characterized by a foreign body reaction and peri-implant bone loss as an exacerbation of this inflammatory response. A key difference in these two schools of thought is the perception of the relative importance of dental plaque in the pathogenesis of crestal bone loss around implants, with obvious implications for treatment. This review investigates the evidence for a persistent foreign body reaction at osseointegrated dental implants and its possible role in crestal bone loss characteristic of peri-implantitis. Further, the role of implant-related material release within the surrounding tissue, particularly titanium particles and corrosion by-products, in the establishment and progression in peri-implantitis is explored. While it is acknowledged that these issues require further investigation, the available evidence suggests that osseointegration is a state of homeostasis between the titanium implant and surrounding tissues, with little evidence that a persistent foreign body reaction is responsible for peri-implant bone loss after osseointegration is established. Further, there is a lack of evidence for a unidirectional causative role of corrosion by-products and titanium particles as possible non-plaque related factors in the etiology of peri-implantitis.
Collapse
Affiliation(s)
- Sašo Ivanovski
- School of DentistryThe University of QueenslandHerstonQueenslandAustralia
| | - Peter Mark Bartold
- School of DentistryUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Yu‐Sheng Huang
- School of DentistryThe University of QueenslandHerstonQueenslandAustralia
| |
Collapse
|
22
|
Cruz MB, Silva N, Marques JF, Mata A, Silva FS, Caramês J. Biomimetic Implant Surfaces and Their Role in Biological Integration-A Concise Review. Biomimetics (Basel) 2022; 7:74. [PMID: 35735590 PMCID: PMC9220941 DOI: 10.3390/biomimetics7020074] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The increased use of dental implants in oral rehabilitation has been followed by the development of new biomaterials as well as improvements in the performance of biomaterials already in use. This triggers the need for appropriate analytical approaches to assess the biological and, ultimately, clinical benefits of these approaches. AIMS To address the role of physical, chemical, mechanical, and biological characteristics in order to determine the critical parameters to improve biological responses and the long-term effectiveness of dental implant surfaces. DATA SOURCES AND METHODS Web of Science, MEDLINE and Lilacs databases were searched for the last 30 years in English, Spanish and Portuguese idioms. RESULTS Chemical composition, wettability, roughness, and topography of dental implant surfaces have all been linked to biological regulation in cell interactions, osseointegration, bone tissue and peri-implant mucosa preservation. CONCLUSION Techniques involving subtractive and additive methods, especially those involving laser treatment or embedding of bioactive nanoparticles, have demonstrated promising results. However, the literature is heterogeneous regarding study design and methodology, which limits comparisons between studies and the definition of the critical determinants of optimal cell response.
Collapse
Affiliation(s)
- Mariana Brito Cruz
- Universidade de Lisboa, Faculdade de Medicina Dentária, Unidade de Investigação em Ciências Orais e Biomédicas (UICOB), Rua Professora Teresa Ambrósio, 1600-277 Lisboa, Portugal; (J.F.M.); (A.M.)
| | - Neusa Silva
- Universidade de Lisboa, Faculdade de Medicina Dentária, Unidade de Investigação em Ciências Orais e Biomédicas (UICOB), LIBPhys-FTC UID/FIS/04559/2013, Rua Professora Teresa Ambrósio, 1600-277 Lisboa, Portugal;
| | - Joana Faria Marques
- Universidade de Lisboa, Faculdade de Medicina Dentária, Unidade de Investigação em Ciências Orais e Biomédicas (UICOB), Rua Professora Teresa Ambrósio, 1600-277 Lisboa, Portugal; (J.F.M.); (A.M.)
| | - António Mata
- Universidade de Lisboa, Faculdade de Medicina Dentária, Unidade de Investigação em Ciências Orais e Biomédicas (UICOB), Rua Professora Teresa Ambrósio, 1600-277 Lisboa, Portugal; (J.F.M.); (A.M.)
- Cochrane Portugal, Instituto de Saúde Baseada na Evidência (ISBE), Faculdade de Medicina Dentária, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Felipe Samuel Silva
- Center for Microelectromechanical Systems (CMEMS), Department of Mechanical Engineering, University of Minho, 4800-058 Guimarães, Portugal;
| | - João Caramês
- Bone Physiology Research Group, Faculdade de Medicina Dentária, Universidade de Lisboa, Rua Professora Teresa Ambrósio, 1600-277 Lisboa, Portugal;
| |
Collapse
|
23
|
Kim YH, Oreffo ROC, Dawson JI. From hurdle to springboard: The macrophage as target in biomaterial-based bone regeneration strategies. Bone 2022; 159:116389. [PMID: 35301163 DOI: 10.1016/j.bone.2022.116389] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 12/16/2022]
Abstract
The past decade has seen a growing appreciation for the role of the innate immune response in mediating repair and biomaterial directed tissue regeneration. The long-held view of the host immune/inflammatory response as an obstacle limiting stem cell regenerative activity, has given way to a fresh appreciation of the pivotal role the macrophage plays in orchestrating the resolution of inflammation and launching the process of remodelling and repair. In the context of bone, work over the past decade has established an essential coordinating role for macrophages in supporting bone repair and sustaining biomaterial driven osteogenesis. In this review evidence for the role of the macrophage in bone regeneration and repair is surveyed before discussing recent biomaterial and drug-delivery based approaches that target macrophage modulation with the goal of accelerating and enhancing bone tissue regeneration.
Collapse
Affiliation(s)
- Yang-Hee Kim
- Bone and Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Jonathan I Dawson
- Bone and Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
24
|
Elangovan G, Mello-Neto JM, Tadakamadla SK, Reher P, Figueredo CMS. A systematic review on neutrophils interactions with titanium and zirconia surfaces: Evidence from in vitro studies. Clin Exp Dent Res 2022; 8:950-958. [PMID: 35535662 PMCID: PMC9382042 DOI: 10.1002/cre2.582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/05/2022] [Accepted: 04/19/2022] [Indexed: 01/04/2023] Open
Abstract
Objectives This systematic review aimed to assess in vitro studies that evaluated neutrophil interactions with different roughness levels in titanium and zirconia implant surfaces. Material and Methods An electronic search for literature was conducted on PubMed, Embase, Scopus, and Web of Science and a total of 14 studies were included. Neutrophil responses were assessed based on adhesion, cell number, surface coverage, cell structure, cytokine secretion, reactive oxygen species (ROS) production, neutrophil activation, receptor expression, and neutrophil extracellular traps (NETs) release. The method of assessing the risk of bias was done using the toxicological data reliability assessment tool (TOXRTOOL). Results Ten studies have identified a significant increase in neutrophil functions, such as surface coverage, cell adhesion, ROS production, and NETs released when interacting with rough titanium surfaces. Moreover, neutrophil interaction with rough–hydrophilic surfaces seems to produce less proinflammatory cytokines and ROS when compared to naive smooth and rough titanium surfaces. Regarding membrane receptor expression, two studies have reported that the FcγIII receptor (CD16) is responsible for initial neutrophil adhesion to hydrophilic titanium surfaces. Only one study compared neutrophil interaction with titanium alloy and zirconia toughened alumina surfaces and reported no significant differences in neutrophil cell count, activation, receptor expression, and death. Conclusions There are not enough studies to conclude neutrophil interactions with titanium and zirconia surfaces. However, different topographic modifications such as roughness and hydrophilicity might influence neutrophil interactions with titanium implant surfaces.
Collapse
Affiliation(s)
- Gayathiri Elangovan
- School of Medicine and Dentistry, Griffith University, Southport, Queensland, Australia
| | - Joao M Mello-Neto
- School of Medicine and Dentistry, Griffith University, Southport, Queensland, Australia
| | - Santosh K Tadakamadla
- School of Medicine and Dentistry, Griffith University, Southport, Queensland, Australia
| | - Peter Reher
- School of Medicine and Dentistry, Griffith University, Southport, Queensland, Australia
| | - Carlos Marcelo S Figueredo
- School of Medicine and Dentistry, Griffith University, Southport, Queensland, Australia.,Affiliated to research, Department of Dental Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
25
|
Sun H, Yang Y, Yu L, Liu K, Fei Y, Guo C, Zhou Y, Hu J, Shi L, Ji H. Inhibition of Inflammatory Response and Promotion of Osteogenic Activity of Zinc-Doped Micro-Arc Titanium Oxide Coatings. ACS OMEGA 2022; 7:14920-14932. [PMID: 35557686 PMCID: PMC9089342 DOI: 10.1021/acsomega.2c00579] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/22/2022] [Indexed: 05/31/2023]
Abstract
An early and sustained immune response can lead to chronic inflammation after the implant is placed in the body. The implantable materials with immunomodulatory effects can reduce the body's immune response and promote the formation of ideal osseointegration between the implants and bone tissue. In this study, zinc-coated titanium micro-arc oxide coating was prepared on titanium surface by micro-arc oxidation. The physical properties, anti-inflammation, and osteogenesis of the material were evaluated. We have physically characterized the surface structure of the coatings by scanning electron microscopy (SEM), energy-dispersive X-ray spectroscopy (EDS), and atomic force microscopy (AFM) and detected the release of Zn2+ from the coating surface by inductively coupled optical plasma emission spectrometry (ICP-OES). The BMSCs were inoculated on the surface of the coating, and the biocompatibility of the coating was evaluated by CCK-8 analysis and living and dead cell staining. The osteogenic effect of the layer on BMSCs was evaluated by alkaline phosphatase (ALP) assays, osteocalcin (OCN) immunofluorescence, and quantitative polymerase chain reaction (q-PCR). The survival status of RAW264.7 on the coating surface and the mRNA expression of the associated proinflammatory markers, tumor necrosis factor-α (TNF-α), cluster of differentiation 86 (CD86), and inducible nitric oxide (INOS) were detected by CCK-8 analysis and q-PCR. In parallel, the cell counting kit-8 (CCK-8) analysis and q-PCR screened and evaluated the effective concentration of Zn2+ anti-inflammatory in vitro. The results show that the coating has good physical characterization, and Zn is uniformly bound to the surface of titanium and shows stable release and good biocompatibility to BMSCs, downregulating the expression of inflammation-related genes promoting the bone formation of BMSCs. We have successfully prepared zinc-coated micro-arc titanium oxide coating on the titanium surface, which has good osteogenesis and great anti-inflammatory potential and provides a new way for osseointegration in the implant.
Collapse
Affiliation(s)
- Haishui Sun
- School
of Stomatology, Weifang Medical University, Weifang 261053, Shandong, China
| | - Yiming Yang
- College
of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, Shanghai 200011, China
- National
Clinical Research Center for Oral Diseases, Shanghai 200011, China
- Shanghai
Key Laboratory of Stomatology, Shanghai 200072, China
- Department
of Orthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Yu
- School
of Stomatology, Weifang Medical University, Weifang 261053, Shandong, China
| | - Ke Liu
- School
of Stomatology, Weifang Medical University, Weifang 261053, Shandong, China
| | - Yifan Fei
- Department
of Oral & Maxillofacial-Head & Neck Oncology, Ninth People’s
Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai 200011, China
| | - Chaoyang Guo
- School
of Stomatology, Weifang Medical University, Weifang 261053, Shandong, China
| | - Yuqi Zhou
- School
of Stomatology, Weifang Medical University, Weifang 261053, Shandong, China
| | - Jingzhou Hu
- Department
of Oral & Maxillofacial-Head & Neck Oncology, Ninth People’s
Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai 200011, China
| | - Lei Shi
- Department
of Oral and Maxillofacial Surgery, Gansu
Provincial Hospital, Lanzhou 730000, China
| | - Honghai Ji
- School
of Stomatology, Weifang Medical University, Weifang 261053, Shandong, China
| |
Collapse
|
26
|
Titanium Surface Characteristics Induce the Specific Reprogramming of Toll-like Receptor Signaling in Macrophages. Int J Mol Sci 2022; 23:ijms23084285. [PMID: 35457102 PMCID: PMC9030374 DOI: 10.3390/ijms23084285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
Most of the research on titanium-based dental implants (Ti-discs) is focused on how they are able to stimulate the formation of new tissue and/or cytotoxic studies, with very scarce data on their effects on functional responses by immunocompetent cells. In particular, the link between the rewiring of innate immune responses and surface biomaterials properties is poorly understood. To address this, we characterize the functional response of macrophage cultures to four different dental titanium surfaces (MA: mechanical abrasion; SB + AE: sandblasting plus etching; SB: sandblasting; AE: acid etching). We use different Toll-like receptor (TLR) ligands towards cell surface receptors (bacterial lipopolysaccharide LPS for TLR4; imiquimod for TLR7; synthetic bacterial triacylated lipoprotein for TLR2/TLR1) and endosomal membrane receptor (poly I:C for TLR3) to simulate bacterial (cell wall bacterial components) or viral infections (dsRNA and ssRNA). The extracellular and total LDH levels indicate that exposure to the different Ti-surfaces is not cytotoxic for macrophages under resting or TLR-stimulated conditions, although there is a tendency towards an impairment in macrophage proliferation, viability or adhesion under TLR4, TLR3 and TLR2/1 stimulations in SB discs cultures. The secreted IL-6 and IL-10 levels are not modified upon resting macrophage exposure to the Ti-surfaces studied as well as steady state levels of iNos or ArgI mRNA. However, macrophage exposure to MA Ti-surface do display an enhanced immune response to TLR4, TLR7 or TLR2/1 compared to other Ti-surfaces in terms of soluble immune mediators secreted and M1/M2 gene expression profiling. This change of characteristics in cellular phenotype might be related to changes in cellular morphology. Remarkably, the gene expression of Tlr3 is the only TLR that is differentially affected by distinct Ti-surface exposure. These results highlight the relevance of patterned substrates in dental implants to achieve a smart manipulation of the immune responses in the context of personalized medicine, cell-based therapies, preferential lineage commitment of precursor cells or control of tissue architecture in oral biology.
Collapse
|
27
|
Necula MG, Mazare A, Negrescu AM, Mitran V, Ozkan S, Trusca R, Park J, Schmuki P, Cimpean A. Macrophage-like Cells Are Responsive to Titania Nanotube Intertube Spacing-An In Vitro Study. Int J Mol Sci 2022; 23:3558. [PMID: 35408918 PMCID: PMC8998567 DOI: 10.3390/ijms23073558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/23/2022] Open
Abstract
With the introduction of a new interdisciplinary field, osteoimmunology, today, it is well acknowledged that biomaterial-induced inflammation is modulated by immune cells, primarily macrophages, and can be controlled by nanotopographical cues. Recent studies have investigated the effect of surface properties in modulating the immune reaction, and literature data indicate that various surface cues can dictate both the immune response and bone tissue repair. In this context, the purpose of the present study was to investigate the effects of titanium dioxide nanotube (TNT) interspacing on the response of the macrophage-like cell line RAW 264.7. The cells were maintained in contact with the surfaces of flat titanium (Ti) and anodic TNTs with an intertube spacing of 20 nm (TNT20) and 80 nm (TNT80), under standard or pro-inflammatory conditions. The results revealed that nanotube interspacing can influence macrophage response in terms of cell survival and proliferation, cellular morphology and polarization, cytokine/chemokine expression, and foreign body reaction. While the nanostructured topography did not tune the macrophages' differentiation into osteoclasts, this behavior was significantly reduced as compared to flat Ti surface. Overall, this study provides a new insight into how nanotubes' morphological features, particularly intertube spacing, could affect macrophage behavior.
Collapse
Affiliation(s)
- Madalina Georgiana Necula
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| | - Anca Mazare
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany; (A.M.); (S.O.); (P.S.)
- Advanced Institute for Materials Research (AIMR), National University Corporation Tohoku University (TU), Sendai 980-8577, Japan
| | - Andreea Mariana Negrescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| | - Valentina Mitran
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| | - Selda Ozkan
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany; (A.M.); (S.O.); (P.S.)
| | - Roxana Trusca
- Faculty of Engineering in Foreign Languages, University Politehnica of Bucharest, 313 Splaiul Indendentei, 060042 Bucharest, Romania;
| | - Jung Park
- Department of Pediatrics, Division of Molecular Pediatrics, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Patrik Schmuki
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany; (A.M.); (S.O.); (P.S.)
- Regional Centre of Advanced Technologies and Materials, 78371 Olomouc, Czech Republic
- Department of Chemistry, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21569, Saudi Arabia
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| |
Collapse
|
28
|
Tuning the surface potential to reprogram immune microenvironment for bone regeneration. Biomaterials 2022; 282:121408. [DOI: 10.1016/j.biomaterials.2022.121408] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/16/2022] [Accepted: 02/11/2022] [Indexed: 12/21/2022]
|
29
|
Antmen E, Vrana NE, Hasirci V. The role of biomaterials and scaffolds in immune responses in regenerative medicine: macrophage phenotype modulation by biomaterial properties and scaffold architectures. Biomater Sci 2021; 9:8090-8110. [PMID: 34762077 DOI: 10.1039/d1bm00840d] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Scaffolds are an integral part of the regenerative medicine field. The contact of biomaterials with tissue, as was clearly observed over the years, induces immune reactions in a material and patient specific manner, where both surface and bulk properties of scaffolds, together with their 3D architecture, have a significant influence on the outcome. This review presents an overview of the reactions to the biomaterials with a specific focus on clinical complications with the implants in the context of immune reactions and an overview of the studies involving biomaterial properties and interactions with innate immune system cells. We emphasize the impact of these studies on scaffold selection and upscaling of microenvironments created by biomaterials from 2D to 3D using immune cell encapsulation, seeding in a 3D scaffold and co-culture with relevant tissue cells. 3D microenvironments are covered with a specific focus on innate cells since a large proportion of these studies used innate immune cells. Finally, the recent studies on the incorporation of adaptive immune cells in immunomodulatory systems are covered in this review. Biomaterial-immune cell interactions are a critical part of regenerative medicine applications. Current efforts in establishing the ground rules for such interactions following implantation can control immune response during all phases of inflammation. Thus, in the near future for complete functional recovery, tissue engineering and control over biomaterials must be considered at the first step of immune modulation and this review covers these interactions, which have remained elusive up to now.
Collapse
Affiliation(s)
- Ezgi Antmen
- BIOMATEN, Middle East Technical University, Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey.
| | - Nihal Engin Vrana
- SPARTHA Medical, 14B Rue de la Canardiere, Strasbourg Cedex 67100, France. .,INSERM Unité 1121 Biomaterials and Bioengineering, CRBS, 1 Rue Eugène Boeckel, Strasbourg Cedex 67000, France
| | - Vasif Hasirci
- BIOMATEN, Middle East Technical University, Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey. .,Biomaterials A&R Center, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,Department of Medical Engineering, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
30
|
Abaricia JO, Farzad N, Heath TJ, Simmons J, Morandini L, Olivares-Navarrete R. Control of innate immune response by biomaterial surface topography, energy, and stiffness. Acta Biomater 2021; 133:58-73. [PMID: 33882355 DOI: 10.1016/j.actbio.2021.04.021] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/27/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022]
Abstract
As the focus of implantable biomaterials has shifted from bioinert implants to bioactive designs, recent research has highlighted the complex interactions between cell physiologic systems and material properties, particularly physical cues. From the cells known to interact with implanted biomaterials, the response of the immune system has been a critical target of study recently. Here, we review studies characterizing the response of innate immune cells to various material cues, particularly of those at the surface of implanted materials.The innate immune system consists of cell types with various roles in inflammation. Neutrophils and macrophages serve both phagocytic and signaling roles, especially early in the inflammatory phase of biomaterial implantation. These cell types ultimately dictate the outcome of implants as chronic inflammation, fibrosis, or integration. Other cell types like dendritic cells, mast cells, natural killer cells, and innate lymphoid cells may also serve an immunomodulatory role in the biomaterial context. This review highlights recent advances in our understanding of the role of innate immunity in the response to implantable biomaterials as well as key mechanobiological findings in innate immune cells underpinning these advances. STATEMENT OF SIGNIFICANCE: This review highlights recent advances in the understanding of the role of innate immunity in the response to implantable biomaterials, especially in neutrophils and macrophages, as well as key mechanobiological findings in innate immune cells underpinning these advances. Here we discuss how physicochemical properties of biomaterials control innate immune cell behavior.
Collapse
|
31
|
Zhou L, Han Y, Ding J, Chen X, Huang S, Xing X, Wu D, Chen J. Regulation of an Antimicrobial Peptide GL13K-Modified Titanium Surface on Osteogenesis, Osteoclastogenesis, and Angiogenesis Base on Osteoimmunology. ACS Biomater Sci Eng 2021; 7:4569-4580. [PMID: 34432981 DOI: 10.1021/acsbiomaterials.1c00639] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Creating a pro-regenerative immune microenvironment around implant biomaterial surfaces is significant to osseointegration. Immune cells, especially macrophages that participate in the osseointegration, including osteogenesis, osteoclastogenesis, and angiogenesis, should be considered when testing biomaterials. In this study, we immobilized an antimicrobial peptide GL13K with immunomodulatory properties onto a titanium surface via silanization. The modified surfaces show good biocompatibility with bone mesenchymal stromal cells (BMSCs), human umbilical vein endothelial cells (HUVECs), and RAW264.7. By co-culturing BMSCs with RAW264.7, we found that the GL13K-coated titanium surfaces could promote late-stage osteogenesis as demonstrated by the upregulated expression of recombinant collagen type I alpha 1 (COL-1α1) and more extracellular matrix mineralization, while the early phase remained unchanged. The surfaces inhibited the osteoclastogenic differentiation of RAW264.7 cells by restraining nuclear factor-activated T cells, cytoplasmic 1 (NFATc1), the main factor of the receptor activator of nuclear factor-κ B, and the receptor activator of the nuclear factor-κ B ligand signaling pathway, from entering the nucleus and further reduced the expression of the activating osteoclastogenic tartrate-resistant acid phosphatase gene. Moreover, the GL13K-coated titanium surface demonstrated significant promotion of angiogenesis differentiation of HUVECs as indicated by the upregulated expression of essential angiogenesis function genes, including hypoxia-inducible factor-1α, endothelial nitric oxide synthase, kinase insert domain receptor, and vascular endothelial growth factor A (HIF-1α, eNOS, KDR, and VEGF-A). Taken together, these results demonstrated that the GL13K coating had properties of osteogenesis, angiogenesis, and anti-osteoclastogenesis via its immunomodulatory potential.
Collapse
Affiliation(s)
- Lin Zhou
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Yu Han
- Stomatological Key Lab of Fujian College and University, Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Jiamin Ding
- Department of Oral Mucosa Affiliated Stomatological Hospital of Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Xuxi Chen
- Institute of Stomatology, Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Shiying Huang
- Fujian Provincial Engineering Research Center of Oral Biomaterial, Fuzhou 350001, People's Republic of China
| | - Xiaojie Xing
- Research Center of Dental Esthetics and Biomechanics, Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Dong Wu
- Research Center of Dental and Craniofacial Implants, Fujian Medical University, Fuzhou 350001, People's Republic of China
| | - Jiang Chen
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350001, People's Republic of China
| |
Collapse
|
32
|
Review on material parameters to enhance bone cell function in vitro and in vivo. Biochem Soc Trans 2021; 48:2039-2050. [PMID: 32940685 DOI: 10.1042/bst20200210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
Bone plays critical roles in support, protection, movement, and metabolism. Although bone has an innate capacity for regeneration, this capacity is limited, and many bone injuries and diseases require intervention. Biomaterials are a critical component of many treatments to restore bone function and include non-resorbable implants to augment bone and resorbable materials to guide regeneration. Biomaterials can vary considerably in their biocompatibility and bioactivity, which are functions of specific material parameters. The success of biomaterials in bone augmentation and regeneration is based on their effects on the function of bone cells. Such functions include adhesion, migration, inflammation, proliferation, communication, differentiation, resorption, and vascularization. This review will focus on how different material parameters can enhance bone cell function both in vitro and in vivo.
Collapse
|
33
|
Wheelis SE, Biguetti CC, Natarajan S, Arteaga A, Allami JE, Chandrashekar BL, Garlet G, Rodrigues DC. Cellular and Molecular Dynamics during Early Oral Osseointegration: A Comprehensive Characterization in the Lewis Rat. ACS Biomater Sci Eng 2021; 7:2392-2407. [PMID: 33625829 PMCID: PMC8796703 DOI: 10.1021/acsbiomaterials.0c01420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE There is a need to improve the predictability of osseointegration in implant dentistry. Current literature uses a variety of in vivo titanium (Ti) implantation models to investigate failure modes and test new materials and surfaces. However, these models produce a variety of results, making comparison across studies difficult. The purpose of this study is to validate an oral osseointegration in the Lewis rat to provide a reproducible baseline to track the inflammatory response and healing of Ti implants. METHODS Ti screws (0.76 mm Ø × 2 mm length) were implanted into the maxillary diastema of 52 adult male Lewis rats. Peri-implant tissues were evaluated 2, 7, 14, and 30 days after implantation (n = 13). Seven of the 13 samples underwent microtomographic analysis, histology, histomorphometry, and immunohistochemistry to track healing parameters. The remaining six samples underwent quantitative polymerase chain reaction (qPCR) to evaluate gene expression of inflammation and bone remodeling markers over time. RESULTS This model achieved a 78.5% success rate. Successful implants had a bone to implant contact (BIC)% of 68.86 ± 3.15 at 30 days on average. Histologically, healing was similar to other rodent models: hematoma and acute inflammation at 2 days, initial bone formation at 7, advanced bone formation and remodeling at 14, and bone maturation at 30. qPCR indicated the highest expression of bone remodeling and inflammatory markers 2-7 days, before slowly declining to nonsurgery control levels at 14-30 days. CONCLUSION This model combines cost-effectiveness and simplicity of a rodent model, while maximizing BIC, making it an excellent candidate for evaluation of new surfaces.
Collapse
Affiliation(s)
| | | | - Shruti Natarajan
- Department of Biological Sciences, University of Texas at Dallas
- Texas A&M College of Dentistry
| | | | | | | | - Gustavo Garlet
- Bauru School of Dentistry, Department of Biological Sciences, University of São Paulo São Paulo, Brazil
| | | |
Collapse
|
34
|
Jinno Y, Stocchero M, Galli S, Toia M, Becktor JP. Impact of a Hydrophilic Dental Implant Surface on Osseointegration: Biomechanical Results in Rabbit. J ORAL IMPLANTOL 2021; 47:163-168. [PMID: 32663272 DOI: 10.1563/aaid-joi-d-19-00217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study aimed to evaluate the effect of surface hydrophilicity on the biomechanical aspects of osseointegration of dental implants in the tibia and femur of rabbits. Forty-eight mature female New Zealand White rabbits were included, and 96 commercially pure, Grade 4, titanium dental implants (control group), and 96 implants of same macro geometry with the hydrophilic surface (test group) were used in this study. One osteotomy was performed in each tibia and femur on both sides of the rabbit, and four implants were placed in each rabbit. Control and test groups were randomly allocated on the left and right sides. During surgery, insertion torque (ITQ) value of the complete implant placement was recorded. After healing periods of 0, 2, 4, and 8 weeks after surgery, implant stability quotient (ISQ) value, and removal torque (RTQ) values were measured. No statistical difference was observed for ITQ, for ISQ and for RTQ between the control group and test group in tibia/femur for all time periods. The effect of hydrophilic properties on moderately roughened surfaces has no impact in terms of biomechanical outcomes (ISQ values and RTQ values) after a healing period of 2 to 8 weeks in rabbit tibias /femurs.
Collapse
Affiliation(s)
- Yohei Jinno
- Department of Oral and Maxillofacial Surgery and Oral Medicine, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Michele Stocchero
- Department of Oral and Maxillofacial Surgery and Oral Medicine, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Silvia Galli
- Department of Prosthodontics, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Marco Toia
- Department of Oral and Maxillofacial Surgery and Oral Medicine, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Jonas P Becktor
- Department of Oral and Maxillofacial Surgery and Oral Medicine, Faculty of Odontology, Malmö University, Malmö, Sweden
| |
Collapse
|
35
|
Orchestrating soft tissue integration at the transmucosal region of titanium implants. Acta Biomater 2021; 124:33-49. [PMID: 33444803 DOI: 10.1016/j.actbio.2021.01.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 12/20/2022]
Abstract
Osseointegration at the bone-implant interface and soft tissue integration (STI) at the trans-mucosal region are crucial for the long-term success of dental implants, especially in compromised patient conditions. The STI quality of conventional smooth and bio-inert titanium-based implants is inferior to that of natural tissue (i.e. teeth), and hence various surface modifications have been suggested. This review article compares and contrasts the various modification strategies (physical, chemical and biological) utilized to enhance STI of Ti implants. It also details the STI challenges associated with conventional Ti-based implants, current surface modification strategies and cutting-edge nano-engineering solutions. The topographical, biological and therapeutic advances achievable via electrochemically anodized Ti implants with TiO2 nanotubes/nanopores are highlighted. Finally, the status and future directions of such nano-engineered implants is discussed, with emphasis on bridging the gap between research and clinical translation.
Collapse
|
36
|
Stavropoulos A, Sandgren R, Bellon B, Sculean A, Pippenger BE. Greater Osseointegration Potential with Nanostructured Surfaces on TiZr: Accelerated vs. Real-Time Ageing. MATERIALS 2021; 14:ma14071678. [PMID: 33805477 PMCID: PMC8036800 DOI: 10.3390/ma14071678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 11/16/2022]
Abstract
Surface chemistry and nanotopography of dental implants can have a substantial impact on osseointegration. The aim of this investigation was to evaluate the effects of surface chemistry and nanotopography on the osseointegration of titanium-zirconium (TiZr; Roxolid®) discs, using a biomechanical pull-out model in rabbits. Two discs each were placed in both the right and left tibiae of 16 rabbits. Five groups of sandblasted acid etched (SLA) discs were tested: (1) hydrophobic without nanostructures (dry/micro) (n = 13); (2) hydrophobic with nanostructures, accelerated aged (dry/nano/AA) (n = 12); (3) hydrophilic without nanostructures (wet/micro) (n = 13); (4) hydrophilic with nanostructures, accelerated aged (wet/nano/AA; SLActive®) (n = 13); (5) hydrophilic with nanostructures, real-time aged (wet/nano/RTA). The animals were sacrificed after four weeks and the biomechanical pull-out force required to remove the discs was evaluated. Adjusted mean pull-out force was greatest for group wet/nano/RTA (64.5 ± 17.7 N) and lowest for group dry/micro (33.8 ± 10.7 N). Multivariate mixed model analysis showed that the pull-out force was significantly greater for all other disc types compared to the dry/micro group. Surface chemistry and topography both had a significant effect on pull-out force (p < 0.0001 for both), but the effect of the interaction between chemistry and topography was not significant (p = 0.1056). The introduction of nanostructures on the TiZr surface significantly increases osseointegration. The introduction of hydrophilicity to the TiZr implant surface significantly increases the capacity for osseointegration, irrespective of the presence or absence of nanotopography.
Collapse
Affiliation(s)
- Andreas Stavropoulos
- Division of Regenerative Dental Medicine and Periodontology, University of Geneva, CH-1211 Genève 4, Switzerland
- Department of Periodontology, Faculty of Odontology, Malmö University, SE-205 06 Malmö, Sweden
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, AT-1090 Vienna, Austria
- Correspondence: or (A.S.); (B.E.P.)
| | - Rebecca Sandgren
- Department of Biomedicine, Medical Faculty, Lunds University, SE-223 62 Lund, Sweden;
| | - Benjamin Bellon
- Department of Preclinical & Translational Research, Institut Straumann, CH-4002 Basel, Switzerland;
- Department of Periodontology, Faculty of Dentistry, University of Zurich, CH-8032 Zurich, Switzerland
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, CH-3210 Bern, Switzerland;
| | - Benjamin E. Pippenger
- Department of Preclinical & Translational Research, Institut Straumann, CH-4002 Basel, Switzerland;
- Department of Periodontology, School of Dental Medicine, University of Bern, CH-3210 Bern, Switzerland;
- Correspondence: or (A.S.); (B.E.P.)
| |
Collapse
|
37
|
Abaricia JO, Shah AH, Ruzga MN, Olivares-Navarrete R. Surface characteristics on commercial dental implants differentially activate macrophages in vitro and in vivo. Clin Oral Implants Res 2021; 32:487-497. [PMID: 33502059 DOI: 10.1111/clr.13717] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/16/2020] [Accepted: 01/20/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Biomaterial implantation provokes an inflammatory response that controls integrative fate. M2 macrophages regulate the response to implants by resolving the inflammatory phase and recruiting progenitor cells to aid healing. We have previously shown that modified titanium (Ti) disks directly induce M2 macrophage polarization. The aim of this study was to examine macrophage response to commercially available Ti or Ti alloy implants with comparable roughness and varying hydrophilicity. MATERIAL AND METHODS Eleven commercially available Ti (A-F) or Ti alloy (G-K) dental implants were examined in this study. Surface topography, chemistry, and hydrophilicity were characterized for each implant. To compare the immune response in vitro, human monocyte-derived macrophages were seeded on implants and secreted pro- and anti-inflammatory proteins measured. To evaluate the inflammatory response in vivo, mice were subcutaneously instrumented with clinical implants, and implant adherent macrophage populations were characterized by flow cytometry. RESULTS Macrophages on hydrophobic Implant C produced the highest level of pro-inflammatory proteins in vitro. In contrast, hydrophilic Implant E produced the second-highest pro-inflammatory response. Implants F and K, both hydrophilics, produced the highest anti-inflammatory protein secretions. Likewise, pro-inflammatory CD80hi macrophages predominated in vivo on implants C and E, and M2 CD206 + macrophages predominated on implants F and K. CONCLUSIONS These findings show that hydrophilicity alone is insufficient to predict the anti-inflammatory effect on macrophage polarization and that other properties-surface composition or topography-determine immune modulation. This in vivo model may be a useful screening method to compare the immunomodulatory response to clinical implants of disparate geometry or size.
Collapse
Affiliation(s)
- Jefferson O Abaricia
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Arth H Shah
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Marissa N Ruzga
- Department of Biomedical Engineering, College of Engineering, Ohio State University, Columbus, OH, USA
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
38
|
He M, Gao X, Fan Y, Xie L, Yang M, Tian W. Tannic acid/Mg 2+-based versatile coating to manipulate the osteoimmunomodulation of implants. J Mater Chem B 2021; 9:1096-1106. [PMID: 33427278 DOI: 10.1039/d0tb01577f] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Instead of directly stimulating osteogenesis, endowing an implant surface with a favourable osteoimmunomodulatory (OIM) function has emerged as a new effective strategy to enhance osteointegration. Though metal-phenolic coatings have demonstrated to possess an immunomodulatory function, their potential application in manipulating an osteoimmune response has not been well explored. Herein, in order to develop a simple, rapid and universal coating method to impart excellent OIM to hard tissue implants, tannic acid (TA) and Mg2+ were selected to form a coating on Ti plate based on metal-phenolic chemistry. Besides its virtues of simplicity, ultrafastness, low-cost, and versatility, another merit for the coating method is that it can easily combine the unique functions of metal ions and phenolic ligands. The chelated Mg2+ can not only activate macrophage polarization towards the anti-inflammatory phenotype but also directly stimulate the osteogenic differentiation of bone marrow-derived stem cells (BMSCs). TA motifs rendered the coating with an excellent reactive oxygen species (ROS) scavenging capacity. TA and Mg2+ showed synergistic effects on regulating macrophage biological behaviour, suppressing its polarization towards the M1 phenotype, and promoting its polarization towards the M2 phenotype. In vivo histological analysis also demonstrated that the TA/Mg2+ coating could effectively inhibit the host response. Finally, the formed osteoimmune environment obviously enhanced the osteogenic differentiation of BMSCs. The above results demonstrated that the designed TA/Mg2+ coating not only possessed the function of directly stimulating osteogenesis but also the function of manipulating OIM to a desired one. Hence, it has great potential to be applied on advanced hard tissue implants to enhance osteointegration.
Collapse
Affiliation(s)
- Min He
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | | | | | | | | | | |
Collapse
|
39
|
Batool F, Özçelik H, Stutz C, Gegout PY, Benkirane-Jessel N, Petit C, Huck O. Modulation of immune-inflammatory responses through surface modifications of biomaterials to promote bone healing and regeneration. J Tissue Eng 2021; 12:20417314211041428. [PMID: 34721831 PMCID: PMC8554547 DOI: 10.1177/20417314211041428] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/06/2021] [Indexed: 12/25/2022] Open
Abstract
Control of inflammation is indispensable for optimal oral wound healing and tissue regeneration. Several biomaterials have been used to enhance the regenerative outcomes; however, the biomaterial implantation can ensure an immune-inflammatory response. The interface between the cells and the biomaterial surface plays a critical role in determining the success of soft and hard tissue regeneration. The initial inflammatory response upon biomaterial implantation helps in tissue repair and regeneration, however, persistant inflammation impairs the wound healing response. The cells interact with the biomaterials through extracellular matrix proteins leading to protein adsorption followed by recruitment, attachment, migration, and proliferation of several immune-inflammatory cells. Physical nanotopography of biomaterials, such as surface proteins, roughness, and porosity, is crucial for driving cellular attachment and migration. Similarly, modification of scaffold surface chemistry by adapting hydrophilicity, surface charge, surface coatings, can down-regulate the initiation of pro-inflammatory cascades. Besides, functionalization of scaffold surfaces with active biological molecules can down-regulate pro-inflammatory and pro-resorptive mediators' release as well as actively up-regulate anti-inflammatory markers. This review encompasses various strategies for the optimization of physical, chemical, and biological properties of biomaterial and the underlying mechanisms to modulate the immune-inflammatory response, thereby, promoting the tissue integration and subsequent soft and hard tissue regeneration potential of the administered biomaterial.
Collapse
Affiliation(s)
- Fareeha Batool
- Faculté de Chirurgie-dentaire, Université de Strasbourg, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Hayriye Özçelik
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Céline Stutz
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Pierre-Yves Gegout
- Faculté de Chirurgie-dentaire, Université de Strasbourg, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Pôle de médecine et chirurgie bucco-dentaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Catherine Petit
- Faculté de Chirurgie-dentaire, Université de Strasbourg, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Pôle de médecine et chirurgie bucco-dentaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Olivier Huck
- Faculté de Chirurgie-dentaire, Université de Strasbourg, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Pôle de médecine et chirurgie bucco-dentaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
40
|
Dos Santos Trento G, Hassumi JS, Buzo Frigério P, Farnezi Bassi AP, Okamoto R, Gabrielli MAC, Pereira-Filho VA. Gene expression, immunohistochemical and microarchitectural evaluation of bone formation around two implant surfaces placed in bone defects filled or not with bone substitute material. Int J Implant Dent 2020; 6:80. [PMID: 33258065 PMCID: PMC7704835 DOI: 10.1186/s40729-020-00279-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/06/2020] [Indexed: 11/19/2022] Open
Abstract
Objective The aim of this study is to evaluate through gene expression, immunohistochemical and microtomographic (micro-CT) analysis the response of peri-implant bone tissue around titanium implants with different surface treatments, placed in bone defects filled or not with bone substitute materials. In addition, to investigate the hypothesis that porous-hydrophilic surface induces a faster bone formation. Materials and methods Twenty-six animals were divided into two groups according to implant surface treatment. In each tibia, a bone defect was created followed by the placement of one implant. On the left tibia, the defect was filled with blood clot (BC), and on the right tibia, the defect was filled with biphasic hydroxyapatite/β-tricalcium-phosphate (HA/TCP) generating four subgroups: BC-N: bone defect filled with blood clot and porous surface titanium implant installed; BC-A: bone defect filled with blood clot and porous-hydrophilic surface titanium implant installed; HA/TCP-N: bone defect filled with bone substitute material and porous surface titanium implant installed; and HA/TCP-A: bone defect filled with bone substitute material and porous-hydrophilic surface titanium implant installed. The animals were submitted to euthanasia at 15, 30, and 60 days after implant installation. The expression of two genes was evaluated: RUNX2 and BSP. Immunohistochemical analyses were performed for detection of RUNX2, OPN, OCN, OPG, and RANKL antibodies and bone matrix proteins. Finally, four parameters were chosen for micro-CT analysis: trabecular number, separation and thickness, and connectivity density. Results Descriptive analysis showed similar findings among the experimental groups. Moreover, porous-hydrophilic surfaces presented a higher expression of RUNX2, which is probably an indicative of better osteogenesis; although the data from this study may be considered an insufficient support for a concrete statement. Conclusion Porous hydrophilic surface can improve and accelerate protein expression and bone formation.
Collapse
Affiliation(s)
- Guilherme Dos Santos Trento
- Department of Diagnosis and Surgery, School of Dentistry, Sao Paulo State University (Unesp), 1680th Humaitá Street, Araraquara, SP, 14801-903, Brazil.
| | - Jaqueline Suemi Hassumi
- Department of Oral and Maxillofacial Surgery, School of Dentistry, São Paulo State University (Unesp), Araçatuba, Brazil
| | - Paula Buzo Frigério
- Department of Oral and Maxillofacial Surgery, School of Dentistry, São Paulo State University (Unesp), Araçatuba, Brazil
| | - Ana Paula Farnezi Bassi
- Department of Oral and Maxillofacial Surgery, School of Dentistry, São Paulo State University (Unesp), Araçatuba, Brazil
| | - Roberta Okamoto
- Department of Oral and Maxillofacial Surgery, School of Dentistry, São Paulo State University (Unesp), Araçatuba, Brazil
| | - Marisa Aparecida Cabrini Gabrielli
- Department of Diagnosis and Surgery, School of Dentistry, Sao Paulo State University (Unesp), 1680th Humaitá Street, Araraquara, SP, 14801-903, Brazil
| | - Valfrido Antonio Pereira-Filho
- Department of Diagnosis and Surgery, School of Dentistry, Sao Paulo State University (Unesp), 1680th Humaitá Street, Araraquara, SP, 14801-903, Brazil
| |
Collapse
|
41
|
Lee RSB, Hamlet SM, Moon HJ, Ivanovski S. Re-establishment of macrophage homeostasis by titanium surface modification in type II diabetes promotes osseous healing. Biomaterials 2020; 267:120464. [PMID: 33130322 DOI: 10.1016/j.biomaterials.2020.120464] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/19/2020] [Accepted: 10/18/2020] [Indexed: 12/27/2022]
Abstract
Titanium surface mediated immunomodulation may address compromised post-implantation bone healing in diabetes mellitus. To assess in vitro phenotypic changes, M1 and M2 polarised Type 2 diabetic rat (Goto Kakizaki, GK) macrophages were cultured on micro-rough (SLA) or hydrophilic nanostructured SLA (modSLA) titanium. The in vivo effects of the SLA and modSLA surfaces on macrophage phenotype, wound-associated protein expression and bone formation were investigated using a critical-sized calvarial defect model. Compared to healthy macrophages, GK M2 macrophage function was compromised, secreting significantly lower levels of the anti-inflammatory cytokine IL-10. The modSLA surface attenuated the pro-inflammatory cellular environment, reducing pro-inflammatory cytokine production and promoting M2 macrophage phenotype differentiation. ModSLA also suppressed gene expression associated with macrophage multinucleation and giant cell formation and stimulated pro-osteogenic genes in co-cultured osteoblasts. In vivo, modSLA enhanced osteogenesis compared to SLA in GK rats. During early healing, proteomic analysis of both surface adherent and wound exudate material showed that modSLA promoted an immunomodulatory pro-reparative environment. The modSLA surface therefore successfully compensated for the compromised M2 macrophage function in Type 2 diabetes by attenuating the pro-inflammatory response and promoting M2 macrophage activity, thus restoring macrophage homeostasis and resulting in a cellular environment favourable for enhanced osseous healing.
Collapse
Affiliation(s)
- Ryan S B Lee
- The University of Queensland, School of Dentistry, Herston, Australia; School of Dentistry and Oral Health, Griffith University, Gold Coast, Australia
| | - Stephen M Hamlet
- School of Dentistry and Oral Health, Griffith University, Gold Coast, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Ho-Jin Moon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Saso Ivanovski
- The University of Queensland, School of Dentistry, Herston, Australia.
| |
Collapse
|
42
|
Matos AO, de Almeida AB, Beline T, Tonon CC, Casarin RCV, Windsor LJ, Duarte S, Nociti FH, Rangel EC, Gregory RL, Barão VAR. Synthesis of multifunctional chlorhexidine-doped thin films for titanium-based implant materials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 117:111289. [PMID: 32919650 DOI: 10.1016/j.msec.2020.111289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 12/28/2022]
Abstract
Our goal was to create bio-functional chlorhexidine (CHX)-doped thin films on commercially pure titanium (cpTi) discs using the glow discharge plasma approach. Different plasma deposition times (50, 35 and 20 min) were used to create bio-functional surfaces based on silicon films with CHX that were compared to the control groups [no CHX and bulk cpTi surface (machined)]. Physico-chemical and biological characterizations included: 1. Morphology, roughness, elemental chemical composition, film thickness, contact angle and surface free energy; 2. CHX-release rate; 3. Antibacterial effect on Streptococcus sanguinis biofilms at 24, 48 and 72 h; 4. Cytotoxicity and metabolic activity using fibroblasts cell culture (NIH-F3T3 cells) at 1, 2, 3 and 4 days; 5. Protein expression by NIH-F3T3 cells at 1, 2, 3 and 4 days; and 6. Co-culture assay of fibroblasts cells and S. sanguinis to assess live and dead cells on the confocal laser scanning microscopy, mitochondrial activity (XTT), membrane leakage (LDH release), and metabolic activity (WST-1 assay) at 1, 2 and 3 days of co-incubation. Data analysis showed that silicon films, with or without CHX coated cpTi discs, increased surface wettability and free energy (p < 0.05) without affecting surface roughness. CHX release was maintained over a 22-day period and resulted in a significant inhibition of biofilm growth (p < 0.05) at 48 and 72 h of biofilm formation for 50 min and 20 min of plasma deposition time groups, respectively. In general, CHX treatment did not significantly affect NIH-F3T3 cell viability (p > 0.05), whereas cell metabolism (MTT assay) was affected by CHX, with the 35 min of plasma deposition time group displaying the lowest values as compared to bulk cpTi (p < 0.05). Moreover, data analysis showed that films, with or without CHX, significantly affected the expression profile of inflammatory cytokines, including IL-4, IL-6, IL-17, IFN-y and TNF-α by NIH-F3T3 cells (p < 0.05). Co-culture demonstrated that CHX-doped film did not affect the metabolic activity, cytotoxicity and viability of fibroblasts cells (p > 0.05). Altogether, the findings of the current study support the conclusion that silicon films added with CHX can be successfully created on titanium discs and have the potential to affect bacterial growth and inflammatory markers without affecting cell viability/proliferation rates.
Collapse
Affiliation(s)
- Adaias Oliveira Matos
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Amanda Bandeira de Almeida
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Thamara Beline
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Caroline C Tonon
- Department of Cariology, Operative Dentistry and Dental Public Health, Indiana University, Purdue University Indianapolis, School of Dentistry, Indianapolis, IN, USA
| | - Renato Corrêa Viana Casarin
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Lester Jack Windsor
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Simone Duarte
- Department of Cariology, Operative Dentistry and Dental Public Health, Indiana University, Purdue University Indianapolis, School of Dentistry, Indianapolis, IN, USA
| | - Francisco Humberto Nociti
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Elidiane Cipriano Rangel
- Laboratory of Technological Plasmas (LaPTec), São Paulo State University (UNESP), Science and Technology Institute of Sorocaba (ICTS), Sorocaba, São Paulo, Brazil
| | - Richard L Gregory
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Valentim Adelino Ricardo Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil.
| |
Collapse
|
43
|
Zhu W, Nie X, Tao Q, Yao H, Wang DA. Interactions at engineered graft-tissue interfaces: A review. APL Bioeng 2020; 4:031502. [PMID: 32844138 PMCID: PMC7443169 DOI: 10.1063/5.0014519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
The interactions at the graft-tissue interfaces are critical for the results of engraftments post-implantation. To improve the success rate of the implantations, as well as the quality of the patients' life, understanding the possible reactions between artificial materials and the host tissues is helpful in designing new generations of material-based grafts aiming at inducing specific responses from surrounding tissues for their own reparation and regeneration. To help researchers understand the complicated interactions that occur after implantations and to promote the development of better-designed grafts with improved biocompatibility and patient responses, in this review, the topics will be discussed from the basic reactions that occur chronologically at the graft-tissue interfaces after implantations to the existing and potential applications of the mechanisms of such reactions in designing of grafts. It offers a chance to bring up-to-date advances in the field and new strategies of controlling the graft-tissue interfaces.
Collapse
Affiliation(s)
- Wenzhen Zhu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Xiaolei Nie
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Qi Tao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, People's Republic of China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, People's Republic of China
| | - Dong-An Wang
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
44
|
Zhou A, Yu H, Liu J, Zheng J, Jia Y, Wu B, Xiang L. Role of Hippo-YAP Signaling in Osseointegration by Regulating Osteogenesis, Angiogenesis, and Osteoimmunology. Front Cell Dev Biol 2020; 8:780. [PMID: 32974339 PMCID: PMC7466665 DOI: 10.3389/fcell.2020.00780] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/24/2020] [Indexed: 02/05/2023] Open
Abstract
The social demand for dental implantation is growing at a rapid rate, while dentists are faced with the dilemma of implantation failures associated with unfavorable osseointegration. Clinical-friendly osteogenesis, angiogenesis and osteoimmunology around dental implants play a pivotal role in a desirable osseointegration and it's increasingly appreciated that Hippo-YAP signaling pathway is implicated in those biological processes both in vitro and in vivo in a variety of study. In this article we review the multiple effects of Hippo-YAP signaling in osseointegration of dental implants by regulating osteogenesis, angiogenesis and osteoimmunology in peri-implant tissue, as well as highlight prospective future directions of relevant investigation.
Collapse
Affiliation(s)
- Anqi Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hui Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiayi Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianan Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yinan Jia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bingfeng Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
El Kholy K, Buser D, Wittneben JG, Bosshardt DD, Van Dyke TE, Kowolik MJ. Investigating the Response of Human Neutrophils to Hydrophilic and Hydrophobic Micro-Rough Titanium Surfaces. MATERIALS 2020; 13:ma13153421. [PMID: 32756413 PMCID: PMC7435731 DOI: 10.3390/ma13153421] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 01/19/2023]
Abstract
Various treatments have been used to change both the topography and chemistry of titanium surfaces, aiming to enhance tissue response and reduce healing times of endosseous implants. Most studies to date focused on bone healing around dental implants occurring later during the healing cascade. However, the impact of the initial inflammatory response in the surgical wound site on the success and healing time of dental implants is crucial for implant integration and success, yet it is still poorly understood. The purpose of this study was to investigate the effect of titanium surface hydrophilicity on the response of human neutrophils by monitoring oxygen radical production, which was measured as chemiluminescence activity. Materials and Methods: Neutrophils were isolated from human donors’ blood buffy coats using the double sucrose gradient method. Neutrophils were exposed to both hydrophilic and hydrophobic titanium surfaces with identical topographies in the presence and absence of human serum. This resulted in six experimental groups including two different implant surfaces, with and without exposure to human serum, and two control groups including an active control with cells alone and a passive control with no cells. Two samples from each group were fixed and analyzed by SEM. Comparisons between surface treatments for differences in chemiluminescence values were performed using analysis of variance ANOVA. Results and Conclusion: In the absence of exposure to serum, there was no significant difference noted between the reaction of neutrophils to hydrophilic and hydrophobic surfaces. However, there was a significant reduction in the mean and active chemiluminescence activity of neutrophils to serum-coated hydrophilic titanium surfaces than to serum-coated hydrophobic titanium surfaces. This suggests that surface hydrophilicity promotes enhanced adsorption of serum proteins, which leads to decreased provocation of initial immune cells and reduction of local oxygen radical production during wound healing. This can help explain the faster osseointegration demonstrated by hydrophilic titanium implants.
Collapse
Affiliation(s)
- Karim El Kholy
- Department of Oral Medicine, Infection and Immunity, Harvard University School of Dental Medicine, Boston, MA 02115, USA
- Center of Clinical and Translational Research, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Daniel Buser
- Department of Oral Surgery and Stomatology, University of Bern School of Dental Medicine, 3010 Bern, Switzerland
| | - Julia-Gabriella Wittneben
- Department of Reconstructive Dentistry and Gerodontology, University of Bern School of Dental Medicine, 3010 Bern, Switzerland
| | - Dieter D Bosshardt
- Department of Oral Surgery and Stomatology, University of Bern School of Dental Medicine, 3010 Bern, Switzerland
| | - Thomas E Van Dyke
- Department of Oral Medicine, Infection and Immunity, Harvard University School of Dental Medicine, Boston, MA 02115, USA
- Center of Clinical and Translational Research, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Michael J Kowolik
- Department of Periodontics, Indiana University School of Dentistry, Indianapolis, IN 46202, USA
| |
Collapse
|
46
|
Jiang Y, Xie Y, Chen Z, Li K, Zheng X. Preparation and characteristics of a novel oxygen-releasing coating for improved cell responses in hypoxic environment. J Biomed Mater Res A 2020; 109:248-261. [PMID: 32496645 DOI: 10.1002/jbm.a.37020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 12/16/2022]
Abstract
Affected by environmental factors such as oxygen deficiency, the secretion of growth factor was abnormal in bone injury sites, resulting in the poor responses of osteoblasts and prolonging the healing process. Herein, in this study, we reported an in situ oxygen-releasing porous titanium coating that combines the dual degradability of poly(lactic-co-glycolic acid) with the self-releasing oxygen capacity of the CaO2 core. The resulting formulation exhibited stable oxygen-releasing capacity as well as the ability to promote proliferation and differentiation of the MC3T3 cell line under hypoxia conditions. According to these results, oxygen-releasing coatings based on improved cellular microenvironment may be a promising bone repair material that would reduce the incidence of difficult bone healing in the future.
Collapse
Affiliation(s)
- Yuyin Jiang
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Youtao Xie
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Zhaoming Chen
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Kai Li
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Xuebin Zheng
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
47
|
Avery SJ, Ayre WN, Sloan AJ, Waddington RJ. Interrogating the Osteogenic Potential of Implant SurfacesIn Vitro: A Review of Current Assays. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:217-229. [DOI: 10.1089/ten.teb.2019.0312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Steven James Avery
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Wayne Nishio Ayre
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Alastair James Sloan
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Rachel Jane Waddington
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
48
|
Li X, Huang Q, Hu X, Wu D, Li N, Liu Y, Li Q, Wu H. Evaluating the osteoimmunomodulatory properties of micro-arc oxidized titanium surface at two different biological stages using an optimized in vitro cell culture strategy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110722. [DOI: 10.1016/j.msec.2020.110722] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 12/31/2019] [Accepted: 02/03/2020] [Indexed: 12/12/2022]
|
49
|
Andrukhov O, Behm C, Blufstein A, Wehner C, Gahn J, Pippenger B, Wagner R, Rausch-Fan X. Effect of implant surface material and roughness to the susceptibility of primary gingival fibroblasts to inflammatory stimuli. Dent Mater 2020; 36:e194-e205. [PMID: 32360041 DOI: 10.1016/j.dental.2020.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/04/2020] [Accepted: 04/13/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVES The impact of the implant surface material and roughness on inflammatory processes in peri-implantitis is not entirely clear. Hence, we investigated how titanium and zirconia surfaces with different roughness influence the susceptibility of primary human gingival fibroblasts to different inflammatory stimuli. METHODS Primary human gingival fibroblasts were isolated from 8 healthy individuals and cultured on following surfaces: smooth titanium machined surface (TiM), smooth zirconia machined surface (ZrM), moderately rough titanium surface (SLA), or moderately rough zirconia surface (ZLA). Subsequently, stimulation with one of the following stimuli was performed: Porphyromonas gingivalis lipopolysaccharide (LPS), tumor necrosis factor (TNF)-α, interleukin (IL)-1β. The resulting production of IL-6, IL-8, and monocyte chemoattractant protein (MCP)-1 was measured by qPCR and ELISA. RESULTS P. gingivalis LPS induced IL-6 and MCP-1 production was slightly higher on titanium surfaces compared to zirconia surfaces. IL-1β induced IL-6 production was not affected by any surface characteristic. The production of MCP-1 in response to IL-1β was higher on smooth compared to rough surfaces and was not affected by the material. The production of IL-6 and MCP-1 in response to TNF-α was most strongly affected by surface characteristics. Higher production of these cytokine was observed on smooth compared to rough surfaces and on titanium compared to zirconia surfaces. Surface characteristics had only minor effects on IL-8 production. SIGNIFICANCE The susceptibility of primary gingival fibroblasts to inflammation depends on various factors, such as surface material, surface roughness and the nature of inflammatory stimuli. All these factors might determine susceptibility to peri-implantitis.
Collapse
Affiliation(s)
- Oleh Andrukhov
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.
| | - Christian Behm
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Alice Blufstein
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Christian Wehner
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Johannes Gahn
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | | | | | - Xiaohui Rausch-Fan
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
50
|
Pachimalla PR, Mishra SK, Chowdhary R. Evaluation of hydrophilic gel made from Acemannan and Moringa oleifera in enhancing osseointegration of dental implants. A preliminary study in rabbits. J Oral Biol Craniofac Res 2020; 10:13-19. [PMID: 32025481 PMCID: PMC6997573 DOI: 10.1016/j.jobcr.2020.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 01/11/2020] [Accepted: 01/19/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Hydrophilic implant surface has gained increasing interest as a factor to stimulate osseointegration. PURPOSE The study was done to formulate hydrophilic gel to be applied on to the dental implant surface, to enhance bone to implant contact (BIC). MATERIALS AND METHODS In first part of study, Acemannan and Moringa oleifera hydro gel formulated in different proportions were coated on the titanium disk and 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide(MTT) assay was done to evaluate cell viability.Cytotoxicity of aqueous extracts of two plants were tested against UMR106 cells. In second part of study, the prototype titanium implants were placed in tibia and femur of 8 male rabbits. Hydrophilic gel formulated from Acemannan and Moringa oleifera were coated on the study groups of implants. Histomorphometric analysis was carried out of the enbloc sections specimens. Student's unpaired t-test was used to compare mean values between the two groups. RESULTS The alkaline phosphatase assay showed least cell inhibition for Acemannan and Moringa oleifera (2:1) as 4.45% and osteoblastic differentiation as 0.328 at 540 nm. Titamium disc coated with hydrogel of Acemannan and Moringa oliefera and seeded with Human MSC shows increased proliferation of osteoblast cells.Compare to study group implants, control group showed no new bone formation. CONCLUSIONS Hydrophilic implant surface showed new bone formation with increased bone to implant contact.There was absent of degenerative changes, necrotic changes, fibrosis, and inflammation at the new BIC.
Collapse
Affiliation(s)
- Praneeth Raj Pachimalla
- Department of Prosthodontics, Rajarajeswari Dental College and Hospital, Bengaluru, 560074, India
| | - Sunil Kumar Mishra
- Department of Prosthodontics, Peoples College of Dental Sciences & Research Centre, Bhopal, 462037, India
| | - Ramesh Chowdhary
- Department of Prosthodontics, Rajarajeswari Dental College and Hospital, Bengaluru, 560074, India
| |
Collapse
|