1
|
Cai D, Liu T, Weng W, Zhu X. Research Progress on Extracellular Matrix-Based Composite Materials in Antibacterial Field. Biomater Res 2025; 29:0128. [PMID: 39822928 PMCID: PMC11735711 DOI: 10.34133/bmr.0128] [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: 08/23/2024] [Revised: 11/07/2024] [Accepted: 12/14/2024] [Indexed: 01/19/2025] Open
Abstract
Due to their exceptional cell compatibility, biodegradability, and capacity to trigger tissue regeneration, extracellular matrix (ECM) materials have drawn considerable attention in tissue healing and regenerative medicine. Interestingly, these materials undergo continuous degradation and release antimicrobial peptides (AMPs) while simultaneously promoting tissue regeneration, thereby exerting a potent antibacterial effect. On this basis, a variety of basic properties of ECM materials, such as porous adsorption, hydrophilic adsorption, group crosslinking, and electrostatic crosslinking, can be used to facilitate the integration of ECM materials and antibacterial agents through physical and chemical approaches in order to enhance the antibacterial efficacy. This article reviews the recent advancements in the study of ECM antibacterial materials, including the antibacterial function and antibacterial mechanism of free-standing ECM materials and ECM-based composite materials. In addition, the urgent challenges and future research prospects of ECM materials in the anti-infection industry are discussed.
Collapse
Affiliation(s)
- Dan Cai
- Department of Orthopedics, The First People’s Hospital of Huzhou,
First Affiliated Hospital of Huzhou University, Zhejiang 313000, China
| | - Tuoqin Liu
- Intensive Care Unit, People’s Hospital of Wuxing District, Wuxing District Maternal and Child Health Hospital, Huzhou, Zhejiang 313000, China
| | - Wei Weng
- Department of Orthopedics, The First People’s Hospital of Huzhou,
First Affiliated Hospital of Huzhou University, Zhejiang 313000, China
| | - Xinhong Zhu
- Department of Orthopedics, The First People’s Hospital of Huzhou,
First Affiliated Hospital of Huzhou University, Zhejiang 313000, China
| |
Collapse
|
2
|
Dini C, Borges MHR, Malheiros SS, Piazza RD, van den Beucken JJJP, de Avila ED, Souza JGS, Barão VAR. Progress in Designing Therapeutic Antimicrobial Hydrogels Targeting Implant-associated Infections: Paving the Way for a Sustainable Platform Applied to Biomedical Devices. Adv Healthc Mater 2025; 14:e2402926. [PMID: 39440583 DOI: 10.1002/adhm.202402926] [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: 08/06/2024] [Revised: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Implantable biomedical devices have found widespread use in restoring lost functions or structures within the human body, but they face a significant challenge from microbial-related infections, which often lead to implant failure. In this context, antimicrobial hydrogels emerge as a promising strategy for treating implant-associated infections owing to their tunable physicochemical properties. However, the literature lacks a comprehensive analysis of antimicrobial hydrogels, encompassing their development, mechanisms, and effect on implant-associated infections, mainly in light of existing in vitro, in vivo, and clinical evidence. Thus, this review addresses the strategies employed by existing studies to tailor hydrogel properties to meet the specific needs of each application. Furthermore, this comprehensive review critically appraises the development of antimicrobial hydrogels, with a particular focus on solving infections related to metallic orthopedic or dental implants. Then, preclinical and clinical studies centering on providing quantitative microbiological results associated with the application of antimicrobial hydrogels are systematically summarized. Overall, antimicrobial hydrogels benefit from the tunable properties of polymers and hold promise as an effective strategy for the local treatment of implant-associated infections. However, future clinical investigations, grounded on robust evidence from in vitro and preclinical studies, are required to explore and validate new antimicrobial hydrogels for clinical use.
Collapse
Affiliation(s)
- Caroline Dini
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Maria Helena Rossy Borges
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Samuel Santana Malheiros
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Rodolfo Debone Piazza
- Physical Chemistry Department, Institute of Chemistry, São Paulo State University (UNESP), Araraquara, São Paulo, 14800-900, Brazil
| | | | - Erica Dorigatti de Avila
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araçatuba, São Paulo State University (UNESP), Araçatuba, São Paulo, 16015-050, Brazil
| | - João Gabriel S Souza
- Dental Research Division, Guarulhos University (UNG), Guarulhos, São Paulo, 07023-070, Brazil
| | - Valentim A R Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| |
Collapse
|
3
|
Shevtsov M, Pitkin E, Combs SE, Yudintceva N, Nazarov D, Meulen GVD, Preucil C, Akkaoui M, Pitkin M. Biocompatibility Analysis of the Silver-Coated Microporous Titanium Implants Manufactured with 3D-Printing Technology. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1876. [PMID: 39683264 PMCID: PMC11643975 DOI: 10.3390/nano14231876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024]
Abstract
3D-printed microporous titanium scaffolds enjoy good biointegration with the residuum's soft and bone tissues, and they promote excellent biomechanical properties in attached prostheses. Implant-associated infection, however, remains a major clinical challenge. Silver-based implant coatings can potentially reduce bacterial growth and inhibit biofilm formation, thereby reducing the risk of periprosthetic infections. In the current study, a 1-µm thick silver coating was prepared on the surface of a 3D-printed microporous titanium alloy with physical vapor deposition (PVD), with a final silver content of 1.00 ± 02 mg/cm2. Cell viability was evaluated with an MTT assay of MC3T3-E1 osteoblasts and human dermal fibroblasts cultured on the surface of the implants, and showed low cytotoxicity for cells during the 14-day follow-up period. Quantitative real-time polymerase chain reaction (RT-PCR) analysis of the relative gene expression of the extracellular matrix components (fibronectin, vitronectin, type I collagen) and cell adhesion markers (α2, α5, αV, β1 integrins) in dermal fibroblasts showed that cell adhesion was not reduced by the silver coating of the microporous implants. An RT-PCR analysis of gene expression related to osteogenic differentiation, including TGF-β1, SMAD4, osteocalcin, osteopontin, and osteonectin in MC3T3-E1 osteoblasts, demonstrated that silver coating did not reduce the osteogenic activity of cells and, to the contrary, enhanced the activity of the TGF-β signaling pathway. For representative sample S5 on day 14, the gene expression levels were 7.15 ± 0.29 (osteonectin), 6.08 ± 0.12 (osteocalcin), and 11.19 ± 0.77 (osteopontin). In conclusion, the data indicate that the silver coating of the microporous titanium implants did not reduce the biointegrative or osteoinductive properties of the titanium scaffold, a finding that argues in favor of applying this coating in designing personalized osseointegrated implants.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany;
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia;
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Emil Pitkin
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany;
| | - Natalia Yudintceva
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia;
| | - Denis Nazarov
- Saint Petersburg State University, Universitetskaya Nab, 7/9, 199034 Saint Petersburg, Russia;
| | | | - Chris Preucil
- Movora, St. Augustine, FL 32095, USA; (G.V.D.M.); (C.P.)
| | | | - Mark Pitkin
- Department of Orthopaedics and Rehabilitation Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
- Poly-Orth International, Sharon, MA 02067, USA
| |
Collapse
|
4
|
Chen Y, Zhou L, Guan M, Jin S, Tan P, Fu X, Zhou Z. Multifunctionally disordered TiO 2 nanoneedles prevent periprosthetic infection and enhance osteointegration by killing bacteria and modulating the osteoimmune microenvironment. Theranostics 2024; 14:6016-6035. [PMID: 39346538 PMCID: PMC11426241 DOI: 10.7150/thno.98219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/01/2024] [Indexed: 10/01/2024] Open
Abstract
Rationale: Total hip arthroplasty (THA) and total knee arthroplasty (TKA) are effective interventions for end-stage osteoarthritis; however, periprosthetic infection is a devastating complication of arthroplasty. To safely prevent periprosthetic infection and enhance osteointegration, the surface modification strategy was utilized to kill bacteria, modulate the osteoimmune microenvironment, and improve new bone formation. Methods: We used the hydrothermal method to fabricate a bionic insect wing with the disordered titanium dioxide nanoneedle (TNN) coating. The mussel-inspired poly-dopamine (PDA) and antibacterial silver nanoparticles (AgNPs) were coated on TNN, named AgNPs-PDA@TNN, to improve the biocompatibility and long-lasting bactericidal capacity. The physicochemical properties of the engineered specimen were evaluated with SEM, AFM, XPS spectrum, and water contact assay. The biocompatibility, bactericidal ability, and the effects on macrophages and osteogenic differentiation were assessed with RT-qPCR, Western blotting, live/dead staining, immunofluorescent staining, etc. Results: The AgNPs-PDA@TNN were biocompatible with macrophages and exhibited immunomodulatory ability to promote M2 macrophage polarization. In addition, AgNPs-PDA@TNN ameliorated the cytotoxicity caused by AgNPs, promoted cell spreading, and increased osteogenesis and matrix deposition of BMSCs. Furthermore, AgNPs-PDA@TNN exhibited bactericidal ability against E. coli and S. aureus by the bionic nanostructure and coated AgNPs. Various imaging analyses indicated the enhanced bactericidal ability and improved new bone formation by AgNPs-PDA@TNN in vivo. H&E, Gram, and Masson staining, verified the improved bone formation, less inflammation, infection, and fibrosis encapsulation. The immunofluorescence staining confirmed the immunomodulatory ability of AgNPs-PDA@TNN in vivo. Conclusion: The bionic insect wing AgNPs-PDA@TNN coating exhibited bactericidal property, immunomodulatory ability, and enhanced osteointegration. Thus, this multidimensional bionic implant surface holds promise as a novel strategy to prevent periprosthetic infection.
Collapse
Affiliation(s)
- Yangmengfan Chen
- Department of Orthopedics and Research Institute of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liqiang Zhou
- MOE Frontiers Science Center for Precision Oncology Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Ming Guan
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shue Jin
- Department of Orthopedics and Research Institute of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Peng Tan
- Department of Orthopedics and Research Institute of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoxue Fu
- Department of Orthopedics and Research Institute of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongke Zhou
- Department of Orthopedics and Research Institute of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
5
|
Chen F, Skelly JD, Chang SY, Song J. Triggered Release of Ampicillin from Metallic Implant Coatings for Combating Periprosthetic Infections. ACS APPLIED MATERIALS & INTERFACES 2024; 16:24421-24430. [PMID: 38690964 PMCID: PMC11099626 DOI: 10.1021/acsami.4c06002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Periprosthetic infections caused by Staphylococcus aureus (S. aureus) pose unique challenges in orthopedic surgeries, in part due to the bacterium's capacity to invade surrounding bone tissues besides forming recalcitrant biofilms on implant surfaces. We previously developed prophylactic implant coatings for the on-demand release of vancomycin, triggered by the cleavage of an oligonucleotide (Oligo) linker by micrococcal nuclease (MN) secreted by the Gram-positive bacterium, to eradicate S. aureus surrounding the implant in vitro and in vivo. Building upon this coating platform, here we explore the feasibility of extending the on-demand release to ampicillin, a broad-spectrum aminopenicillin β-lactam antibiotic that is more effective than vancomycin in killing Gram-negative bacteria that may accompany S. aureus infections. The amino group of ampicillin was successfully conjugated to the carboxyl end of an MN-sensitive Oligo covalently integrated in a polymethacrylate hydrogel coating applied to titanium alloy pins. The resultant Oligo-Ampicillin hydrogel coating released the β-lactam in the presence of S. aureus and successfully cleared nearby S. aureus in vitro. When the Oligo-Ampicillin-coated pin was delivered to a rat femoral canal inoculated with 1000 cfu S. aureus, it prevented periprosthetic infection with timely on-demand drug release. The clearance of the bacteria from the pin surface as well as surrounding tissue persisted over 3 months, with no local or systemic toxicity observed with the coating. The negatively charged Oligo fragment attached to ampicillin upon cleavage from the coating did diminish the antibiotic's potency against S. aureus and Escherichia coli (E. coli) to varying degrees, likely due to electrostatic repulsion by the anionic surfaces of the bacteria. Although the on-demand release of the β-lactam led to adequate killing of S. aureus but not E. coli in the presence of a mixture of the bacteria, strong inhibition of the colonization of the remaining E. coli on hydrogel coating was observed. These findings will inspire considerations of alternative broad-spectrum antibiotics, optimized drug conjugation, and Oligo linker engineering for more effective protection against polymicrobial periprosthetic infections.
Collapse
Affiliation(s)
- Feiyang Chen
- Department of Orthopedics and Physical Rehabilitation, Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Jordan D. Skelly
- Department of Orthopedics and Physical Rehabilitation, Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Shing-Yun Chang
- Department of Orthopedics and Physical Rehabilitation, Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Jie Song
- Department of Orthopedics and Physical Rehabilitation, Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
6
|
Eikani C, Hoyt A, Cho E, Levack AE. The State of Local Antibiotic Use in Orthopedic Trauma. Orthop Clin North Am 2024; 55:207-216. [PMID: 38403367 DOI: 10.1016/j.ocl.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Fracture-related infections are a challenging complication in orthopedic trauma that often necessitates multiple surgeries. Early administration of systemic antibiotics and surgical intervention remains the gold standard of care, but despite these measures, treatment failures can be as high as 35%. For these reasons, the introduction of local antibiotics at the site of at-risk fractures has increased over the past decade. This review looks at the various measures being used clinically including local antibiotic powder, polymethylmethacrylate, biodegradable substances, antibiotic-coated implants, and novel methods such as hydrogels and nanoparticles that have the potential for use in the future.
Collapse
Affiliation(s)
- Carlo Eikani
- Loyola University Stritch School of Medicine; Department of Orthopaedic Surgery & Rehabilitation, Loyola University Medical Center, 2160 South 1st Avenue, Maguire Suite 1700, Maywood, IL, USA.
| | - Aaron Hoyt
- Department of Orthopaedic Surgery & Rehabilitation, Loyola University Medical Center, 2160 South 1st Avenue, Maguire Suite 1700, Maywood, IL, USA
| | - Elizabeth Cho
- Department of Orthopaedic Surgery & Rehabilitation, Loyola University Medical Center, 2160 South 1st Avenue, Maguire Suite 1700, Maywood, IL, USA
| | - Ashley E Levack
- Loyola University Stritch School of Medicine; Department of Orthopaedic Surgery & Rehabilitation, Loyola University Medical Center, 2160 South 1st Avenue, Maguire Suite 1700, Maywood, IL, USA
| |
Collapse
|
7
|
Lu G, Zhao G, Wang S, Li H, Yu Q, Sun Q, Wang B, Wei L, Fu Z, Zhao Z, Yang L, Deng L, Zheng X, Cai M, Lu M. Injectable Nano-Micro Composites with Anti-bacterial and Osteogenic Capabilities for Minimally Invasive Treatment of Osteomyelitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306964. [PMID: 38234236 DOI: 10.1002/advs.202306964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/17/2023] [Indexed: 01/19/2024]
Abstract
The effective management of osteomyelitis remains extremely challenging due to the difficulty associated with treating bone defects, the high probability of recurrence, the requirement of secondary surgery or multiple surgeries, and the difficulty in eradicating infections caused by methicillin-resistant Staphylococcus aureus (MRSA). Hence, smart biodegradable biomaterials that provide effective and precise local anti-infection effects and can promote the repair of bone defects are actively being developed. Here, a novel nano-micro composite is fabricated by combining calcium phosphate (CaP) nanosheets with drug-loaded GelMA microspheres via microfluidic technology. The microspheres are covalently linked with vancomycin (Van) through an oligonucleotide (oligo) linker using an EDC/NHS carboxyl activator. Accordingly, a smart nano-micro composite called "CaP@MS-Oligo-Van" is synthesized. The porous CaP@MS-Oligo-Van composites can target and capture bacteria. They can also release Van in response to the presence of bacterial micrococcal nuclease and Ca2+, exerting additional antibacterial effects and inhibiting the inflammatory response. Finally, the released CaP nanosheets can promote bone tissue repair. Overall, the findings show that a rapid, targeted drug release system based on CaP@MS-Oligo-Van can effectively target bone tissue infections. Hence, this agent holds potential in the clinical treatment of osteomyelitis caused by MRSA.
Collapse
Affiliation(s)
- Guanghua Lu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Gang Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Shen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Hanqing Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Qiang Yu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Qi Sun
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Bo Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Li Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Zi Fu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Zhenyu Zhao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Linshan Yang
- Taikang Bybo Dental, Shanghai, 200001, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Xianyou Zheng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Min Lu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| |
Collapse
|
8
|
Hassani M, Kamankesh M, Rad-Malekshahi M, Rostamizadeh K, Rezaee F, Haririan I, Daghighi SM. Biomaterials coated with zwitterionic polymer brush demonstrated significant resistance to bacterial adhesion and biofilm formation in comparison to brush coatings incorporated with antibiotics. Colloids Surf B Biointerfaces 2024; 234:113671. [PMID: 38039822 DOI: 10.1016/j.colsurfb.2023.113671] [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: 07/01/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/03/2023]
Abstract
A critical problem with the use of biomaterial implants is associated with bacterial adhesion on the surface of implants and in turn the biofilm formation. Among different strategies that have been reported to resolve this dilemma, surface design combined with both antiadhesive and antimicrobial properties has proven to be highly effective. Physiochemical properties of polymer brush coatings possess non-adhesive capability against bacterial adhesion and create a niche for further functionalization. The current study aims to evaluate the effect of antibiotics incorporated into the polymer brush on bacterial adhesion and biofilm formation. Brushes made of zwitterionic polymers were synthesized, functionalized with vancomycin via both physical and chemical conjugation, and grafted onto the silicon rubber surfaces. Antibacterial and antiadhesive measurements of designed coated biomaterials were mediated through the use of a parallel plate flow chamber against biofilm growth developed by Staphylococcus aureus and Escherichia coli over a period of 24 h. The analysis of biofilm growth on designed coated biomaterials showed that the pristine coated zwitterionic brushes are significantly resistant to bacterial adhesion and biofilm formation but not in the polymer brush coating incorporated with antibiotics.
Collapse
Affiliation(s)
- Maryam Hassani
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Kamankesh
- Department of Polymer Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Kobra Rostamizadeh
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Farhad Rezaee
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Mojtaba Daghighi
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Pedroni MA, Ribeiro VST, Cieslinski J, Lopes APDA, Kraft L, Suss PH, Tuon FF. Different concentrations of vancomycin with gentamicin loaded PMMA to inhibit biofilm formation of Staphylococcus aureus and their implications. J Orthop Sci 2024; 29:334-340. [PMID: 36526520 DOI: 10.1016/j.jos.2022.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND This study aimed to evaluate different concentrations of vancomycin and/or gentamicin loaded polymethylmethacrylate (PMMA) against biofilm formation of Staphylococcus aureus. METHODS Biofilm production of S. aureus in PMMA loaded with different concentrations of vancomycin and gentamicin were evaluated by quantitative analysis of biofilm cells, scanning electronic microscopy, viability assay, Fourier transform infrared spectroscopy, and checkerboard. Statistical analysis was performed by Mann Whitney test. The difference in colony forming units per mL was significant when p < 0.05. RESULTS All loaded PMMA presented a reduction in the number of colony forming units per mL (p < 0.05). The gentamicin-loaded PMMA could inhibits the grown of sessile cells (p < 0.05), where the group vancomycin 4 g + gentamicin 500 mg presented a better result. The Fourier transform infrared spectra showed no significant differences, and checkerboard of vancomycin and gentamicin showed synergism. CONCLUSION Effects against adherence and bacterial development in PMMA loaded with antibiotics were mainly seen in the group vancomycin 4 g + gentamicin 500 mg, and synergic effect can be applied in antibiotic-loaded cement.
Collapse
Affiliation(s)
- Marco Antonio Pedroni
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Victoria Stadler Tasca Ribeiro
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Juliette Cieslinski
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Ana Paula de Andrade Lopes
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Letícia Kraft
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Paula Hansen Suss
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Felipe Francisco Tuon
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil.
| |
Collapse
|
10
|
Wang S, He W, Wang H, Liu D, Wang M, Yang H, Pan G, Li B. Hematoma-like dynamic hydrogelation through natural glycopeptide molecular recognition for infected bone fracture repair. Bioact Mater 2023; 30:73-84. [PMID: 37575878 PMCID: PMC10413008 DOI: 10.1016/j.bioactmat.2023.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/03/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023] Open
Abstract
Infected bone fractures remain a major clinical challenge for orthopedic surgeons. From a tissue regeneration perspective, biomaterial scaffolds with antibacterial and osteoinductive activities are highly desired, while advanced materials capable of mimicking the pathological microenvironment during the healing process of infected tissues remain an area deserving more research. Hematoma, the gel-like blood coagulum, plays an essential role in bone fracture repair because of its ability to serve as a dynamic and temporary scaffold with cytokines for both pathogen elimination and tissue healing. In light of this, we designed a dynamic hydrogel with hematoma-like antimicrobial or reparative performance for infected bone fracture repair in this study. The proposed dynamic hydrogel network was based on the reversible recognition of a natural glycopeptide antibiotic vancomycin (Van) and its target dipeptide D-Ala-D-Ala (AA), which could serve as a hematoma-like scaffold for obliterating bacteria in the fracture region and promoting bone repair by introducing an endogenous osteogenic peptide (OGP). In vivo experiments demonstrated that the hydrogel could rapidly eradicate bacteria, improve bone regeneration and restore the local inflammatory microenvironment. Together, findings from this study imply that the use of hematoma-like dynamic hydrogel could lead to a biomimetic revolution in surgical strategies against susceptible bone fractures.
Collapse
Affiliation(s)
- Shenghao Wang
- Orthopedic Institute, Department of Orthopaedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Wenbo He
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Huan Wang
- Orthopedic Institute, Department of Orthopaedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Dachuan Liu
- Orthopedic Institute, Department of Orthopaedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Miao Wang
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Huilin Yang
- Orthopedic Institute, Department of Orthopaedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Bin Li
- Orthopedic Institute, Department of Orthopaedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215006, China
| |
Collapse
|
11
|
Jiang P, Zhang Y, Hu R, Shi B, Zhang L, Huang Q, Yang Y, Tang P, Lin C. Advanced surface engineering of titanium materials for biomedical applications: From static modification to dynamic responsive regulation. Bioact Mater 2023; 27:15-57. [PMID: 37035422 PMCID: PMC10074421 DOI: 10.1016/j.bioactmat.2023.03.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
Titanium (Ti) and its alloys have been widely used as orthopedic implants, because of their favorable mechanical properties, corrosion resistance and biocompatibility. Despite their significant success in various clinical applications, the probability of failure, degradation and revision is undesirably high, especially for the patients with low bone density, insufficient quantity of bone or osteoporosis, which renders the studies on surface modification of Ti still active to further improve clinical results. It is discerned that surface physicochemical properties directly influence and even control the dynamic interaction that subsequently determines the success or rejection of orthopedic implants. Therefore, it is crucial to endow bulk materials with specific surface properties of high bioactivity that can be performed by surface modification to realize the osseointegration. This article first reviews surface characteristics of Ti materials and various conventional surface modification techniques involving mechanical, physical and chemical treatments based on the formation mechanism of the modified coatings. Such conventional methods are able to improve bioactivity of Ti implants, but the surfaces with static state cannot respond to the dynamic biological cascades from the living cells and tissues. Hence, beyond traditional static design, dynamic responsive avenues are then emerging. The dynamic stimuli sources for surface functionalization can originate from environmental triggers or physiological triggers. In short, this review surveys recent developments in the surface engineering of Ti materials, with a specific emphasis on advances in static to dynamic functionality, which provides perspectives for improving bioactivity and biocompatibility of Ti implants.
Collapse
Affiliation(s)
- Pinliang Jiang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
- State Key Lab of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Yanmei Zhang
- State Key Lab of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Ren Hu
- State Key Lab of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Bin Shi
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, China
| | - Lihai Zhang
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, China
| | - Qiaoling Huang
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, College of Physical Science and Technology, Xiamen University, Xiamen, 361005, China
| | - Yun Yang
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, College of Physical Science and Technology, Xiamen University, Xiamen, 361005, China
| | - Peifu Tang
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, China
| | - Changjian Lin
- State Key Lab of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
12
|
Skelly JD, Chen F, Chang SY, Ujjwal RR, Ghimire A, Ayers DC, Song J. Modulating On-Demand Release of Vancomycin from Implant Coatings via Chemical Modification of a Micrococcal Nuclease-Sensitive Oligonucleotide Linker. ACS APPLIED MATERIALS & INTERFACES 2023; 15:37174-37183. [PMID: 37525332 PMCID: PMC10421633 DOI: 10.1021/acsami.3c05881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Periprosthetic infections are one of the most serious complications in orthopedic surgeries, and those caused by Staphylococcus aureus (S. aureus) are particularly hard to treat due to their tendency to form biofilms on implants and their notorious ability to invade the surrounding bones. The existing prophylactic local antibiotic deliveries involve excessive drug loading doses that could risk the development of drug resistance strains. Utilizing an oligonucleotide linker sensitive to micrococcal nuclease (MN) cleavage, we previously developed an implant coating capable of releasing covalently tethered vancomycin, triggered by S. aureus-secreted MN, to prevent periprosthetic infections in the mouse intramedullary (IM) canal. To further engineer this exciting platform to meet broader clinical needs, here, we chemically modified the oligonucleotide linker by a combination of 2'-O-methylation and phosphorothioate modification to achieve additional modulation of its stability/sensitivity to MN and the kinetics of MN-triggered on-demand release. We found that when all phosphodiester bonds within the oligonucleotide linker 5'-carboxy-mCmGTTmCmG-3-acrydite, except for the one between TT, were replaced by phosphorothioate, the oligonucleotide (6PS) stability significantly increased and enabled the most sustained release of tethered vancomycin from the coating. By contrast, when only the peripheral phosphodiester bonds at the 5'- and 3'-ends were replaced by phosphorothioate, the resulting oligonucleotide (2PS) linker was cleaved by MN more rapidly than that without any PS modifications (0PS). Using a rat femoral canal periprosthetic infection model where 1000 CFU S. aureus was inoculated at the time of IM pin insertion, we showed that the prophylactic implant coating containing either 0PS- or 2PS-modified oligonucleotide linker effectively eradicated the bacteria by enabling the rapid on-demand release of vancomycin. No bacteria were detected from the explanted pins, and no signs of cortical bone changes were detected in these treatment groups throughout the 3 month follow-ups. With an antibiotic tethering dose significantly lower than conventional antibiotic-bearing bone cements, these coatings also exhibited excellent biocompatibility. These chemically modified oligonucleotides could help tailor prophylactic anti-infective coating strategies to meet a range of clinical challenges where the risks for S. aureus prosthetic infections range from transient to long-lasting.
Collapse
Affiliation(s)
- Jordan D Skelly
- Department of Orthopedics and Physical Rehabilitation, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
| | - Feiyang Chen
- Department of Orthopedics and Physical Rehabilitation, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
| | - Shing-Yun Chang
- Department of Orthopedics and Physical Rehabilitation, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
| | - Rewati R Ujjwal
- Department of Orthopedics and Physical Rehabilitation, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
| | - Ananta Ghimire
- Department of Orthopedics and Physical Rehabilitation, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
| | - David C Ayers
- Department of Orthopedics and Physical Rehabilitation, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
| | - Jie Song
- Department of Orthopedics and Physical Rehabilitation, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
| |
Collapse
|
13
|
Wang X, Zhang M, Zhu T, Wei Q, Liu G, Ding J. Flourishing Antibacterial Strategies for Osteomyelitis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206154. [PMID: 36717275 PMCID: PMC10104653 DOI: 10.1002/advs.202206154] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/05/2022] [Indexed: 06/18/2023]
Abstract
Osteomyelitis is a destructive disease of bone tissue caused by infection with pathogenic microorganisms. Because of the complex and long-term abnormal conditions, osteomyelitis is one of the refractory diseases in orthopedics. Currently, anti-infective therapy is the primary modality for osteomyelitis therapy in addition to thorough surgical debridement. However, bacterial resistance has gradually reduced the benefits of traditional antibiotics, and the development of advanced antibacterial agents has received growing attention. This review introduces the main targets of antibacterial agents for treating osteomyelitis, including bacterial cell wall, cell membrane, intracellular macromolecules, and bacterial energy metabolism, focuses on their mechanisms, and predicts prospects for clinical applications.
Collapse
Affiliation(s)
- Xukai Wang
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Mingran Zhang
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
| | - Tongtong Zhu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
| | - Qiuhua Wei
- Department of Disinfection and Infection ControlChinese PLA Center for Disease Control and Prevention20 Dongda StreetBeijing100071P. R. China
| | - Guangyao Liu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
14
|
Talebian S, Mendes B, Conniot J, Farajikhah S, Dehghani F, Li Z, Bitoque D, Silva G, Naficy S, Conde J, Wallace GG. Biopolymeric Coatings for Local Release of Therapeutics from Biomedical Implants. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207603. [PMID: 36782094 PMCID: PMC10131825 DOI: 10.1002/advs.202207603] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Indexed: 06/18/2023]
Abstract
The deployment of structures that enable localized release of bioactive molecules can result in more efficacious treatment of disease and better integration of implantable bionic devices. The strategic design of a biopolymeric coating can be used to engineer the optimal release profile depending on the task at hand. As illustrative examples, here advances in delivery of drugs from bone, brain, ocular, and cardiovascular implants are reviewed. These areas are focused to highlight that both hard and soft tissue implants can benefit from controlled localized delivery. The composition of biopolymers used to achieve appropriate delivery to the selected tissue types, and their corresponding outcomes are brought to the fore. To conclude, key factors in designing drug-loaded biopolymeric coatings for biomedical implants are highlighted.
Collapse
Affiliation(s)
- Sepehr Talebian
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Bárbara Mendes
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - João Conniot
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Syamak Farajikhah
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Fariba Dehghani
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Zhongyan Li
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
| | - Diogo Bitoque
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Gabriela Silva
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Sina Naficy
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - João Conde
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Gordon G. Wallace
- Intelligent Polymer Research InstituteARC Centre of Excellence for Electromaterials ScienceAIIM FacilityUniversity of WollongongSydneyNSW2522Australia
| |
Collapse
|
15
|
Chen X, Zhou J, Qian Y, Zhao L. Antibacterial coatings on orthopedic implants. Mater Today Bio 2023; 19:100586. [PMID: 36896412 PMCID: PMC9988588 DOI: 10.1016/j.mtbio.2023.100586] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023] Open
Abstract
With the aging of population and the rapid improvement of public health and medical level in recent years, people have had an increasing demand for orthopedic implants. However, premature implant failure and postoperative complications frequently occur due to implant-related infections, which not only increase the social and economic burden, but also greatly affect the patient's quality of life, finally restraining the clinical use of orthopedic implants. Antibacterial coatings, as an effective strategy to solve the above problems, have been extensively studied and motivated the development of novel strategies to optimize the implant. In this paper, a variety of antibacterial coatings recently developed for orthopedic implants were briefly reviewed, with the focus on the synergistic multi-mechanism antibacterial coatings, multi-functional antibacterial coatings, and smart antibacterial coatings that are more potential for clinical use, thereby providing theoretical references for further fabrication of novel and high-performance coatings satisfying the complex clinical needs.
Collapse
Affiliation(s)
- Xionggang Chen
- Institute of Physics & Optoelectronics Technology, Baoji Advanced Titanium Alloys and Functional Coatings Cooperative Innovation Center, Baoji University of Arts and Sciences, Baoji, 721016, PR China
| | - Jianhong Zhou
- Institute of Physics & Optoelectronics Technology, Baoji Advanced Titanium Alloys and Functional Coatings Cooperative Innovation Center, Baoji University of Arts and Sciences, Baoji, 721016, PR China
| | - Yu Qian
- Institute of Physics & Optoelectronics Technology, Baoji Advanced Titanium Alloys and Functional Coatings Cooperative Innovation Center, Baoji University of Arts and Sciences, Baoji, 721016, PR China
| | - LingZhou Zhao
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, 100142, PR China
| |
Collapse
|
16
|
Wang H, Mu N, He Y, Zhang X, Lei J, Yang C, Ma L, Gao Y. Ultrasound-controlled MXene-based Schottky heterojunction improves anti-infection and osteogenesis properties. Theranostics 2023; 13:1669-1683. [PMID: 37056559 PMCID: PMC10086208 DOI: 10.7150/thno.81511] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/25/2023] [Indexed: 04/15/2023] Open
Abstract
Background: The current clinical treatment of osteomyelitis is limited by the emergence of drug-resistant bacteria, which often leads to the failure of traditional antibiotic treatment and large bone defects. Sonodynamic therapy (SDT) is a new strategy that is widely used to overcome the problem of bacterial resistance to antibiotic therapy as well as poor tissue penetration using near-infrared light in photodynamic therapy (PDT). Therefore, it is necessary to develop a new sonosensitizer that can kill bacteria and promote bone repair. Methods: Herein, we developed a sonosensitizer, porphyrin metal-organic framework (HNTM), with a Schottky junction modified by Ti3C2 nanosheets (HN-Ti3C2) for highly efficient sonodynamic therapy of osteomyelitis and bone regeneration. Results: Ti3C2 greatly improves the acoustic catalytic performance by rapidly transferring the charge carriers generated by HNTM under ultrasound (US) irradiation, which killing drug-resistant bacteria through the generation of large amounts of reactive oxygen species (ROS). And HN-Ti3C2 shows excellent 99.75% antibacterial effectiveness against MRSA. In addition, HN-Ti3C2 generates a sonocurrent under low-intensity US to promote the repair of bone defects for a long time period. Mechanistic research using CCK-8 and RNA-seq showed that HN-Ti3C2 nanocomposites can promote the proliferation of stem cells by regulating the cell cycle, DNA replication, and apoptosis. In addition, after low-intensity US irradiation, HN-Ti3C2 promotes osteogenic differentiation via some key signaling pathways, including the calcium, Wnt, and TGF-beta signaling pathways, according to the Kyoto Encyclopedia of Genes and Genomes (KEGG). In a MRSA-infected rat tibial osteomyelitis model, HN-Ti3C2 successfully eliminated the infection and significantly improved bone regeneration under US irradiation. Conclusion: This study indicates that engineered HN-Ti3C2 is a distinctive nanocomposite for successful osteomyelitis treatment.
Collapse
Affiliation(s)
- Hongchuan Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Na Mu
- College of Agronomy, Xinjiang Agriculture University, Urumqi, Xinjiang, China
| | - Yaqi He
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoguang Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jie Lei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yong Gao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
17
|
Antimicrobial-Loaded Polyacrylamide Hydrogels Supported on Titanium as Reservoir for Local Drug Delivery. Pathogens 2023; 12:pathogens12020202. [PMID: 36839473 PMCID: PMC9962340 DOI: 10.3390/pathogens12020202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/11/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Arthroplasty is a highly successful treatment to restore the function of a joint. The contamination of the implant via bacterial adhesion is the first step toward the development of device-associated infections. The emerging concern about antimicrobial resistance resulted in a growing interest to develop alternative therapeutic strategies. Thus, the increment in the incidence of bacterial periprosthetic infections, the complexity of treating infections caused by organisms growing in biofilms, together with the rise in antibiotic resistant bacteria, expose the need to design novel surfaces that provide innovative solutions to these rising problems. The aim of this work is to develop a coating on titanium (Ti) suitable for inhibiting bacterial adhesion and proliferation, and hence, biofilm formation on the surface. We have successfully prepared polyacrylamide hydrogels containing the conventional antibiotic ampicillin (AMP), silver nanoparticles (AgNPs), and both, AMP and AgNPs. The release of the antibacterial agents from the gelled to aqueous media resulted in an excellent antibacterial action of the loaded hydrogels against sessile S. aureus. Moreover, a synergic effect was achieved with the incorporation of both AMP and AgNPs in the hydrogel, which highlights the importance of combining antimicrobial agents having different targets. The polyacrylamide hydrogel coating on the Ti surface was successfully achieved, as it was demonstrated by FTIR, contact angle, and AFM measurements. The modified Ti surfaces having the polyacrylamide hydrogel film containing AgNPs and AMP retained the highest antibacterial effect against S. aureus as it was found for the unsupported hydrogels. The modified surfaces exhibit an excellent cytocompatibility, since healthy, flattened MC3T3-E1 cells spread on the surfaces were observed. In addition, similar macrophage RAW 264.7 adhesion was found on all the surfaces, which could be related to a low macrophage activation. Our results indicate that AMP and AgNP-loaded polyacrylamide hydrogel films on Ti are a good alternative for designing efficient antibacterial surfaces having an excellent cytocompatibility without inducing an exacerbated immune response. The approach emerges as a superior alternative to the widely used direct adsorption of therapeutic agents on surfaces, since the antimicrobial-loaded hydrogel coatings open the possibility of modulating the concentration of the antimicrobial agents to enhance bacterial killing, and then, reducing the risk of infections in implantable materials.
Collapse
|
18
|
Recent Advancements in Metallic Drug-Eluting Implants. Pharmaceutics 2023; 15:pharmaceutics15010223. [PMID: 36678852 PMCID: PMC9862589 DOI: 10.3390/pharmaceutics15010223] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Over the past decade, metallic drug-eluting implants have gained significance in orthopedic and dental applications for controlled drug release, specifically for preventing infection associated with implants. Recent studies showed that metallic implants loaded with drugs were substituted for conventional bare metal implants to achieve sustained and controlled drug release, resulting in a desired local therapeutic concentration. A number of secondary features can be provided by the incorporated active molecules, including the promotion of osteoconduction and angiogenesis, the inhibition of bacterial invasion, and the modulation of host body reaction. This paper reviews recent trends in the development of the metallic drug-eluting implants with various drug delivery systems in the past three years. There are various types of drug-eluting implants that have been developed to meet this purpose, depending on the drug or agents that have been loaded on them. These include anti-inflammatory drugs, antibiotics agents, growth factors, and anti-resorptive drugs.
Collapse
|
19
|
Cheng Y, Zhang Y, Zhao Z, Li G, Li J, Li A, Xue Y, Zhu B, Wu Z, Zhang X. Guanidinium-Decorated Nanostructure for Precision Sonodynamic-Catalytic Therapy of MRSA-Infected Osteomyelitis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2206646. [PMID: 36245331 DOI: 10.1002/adma.202206646] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/23/2022] [Indexed: 06/16/2023]
Abstract
Osteomyelitis caused by methicillin-resistant Staphylococcus aureus (MRSA) biofilm infection is difficult to eradicate and can even be life-threatening. Given that the infection is persistent and deep-seated in the bone tissue, controlled and efficient treatment of osteomyelitis remains challenging. Herein, an activatable nanostructure (Au/TNT@PG) is presented for synergistic sonodynamic-catalytic therapy of MRSA-infected osteomyelitis. The Au/TNT@PG backbone is obtained by conjugating a guanidinium-rich polymer (PG), a component that penetrates the biofilm matrix, onto ultrasound (US)-absorbing gold-doped titanate nanotubes (Au/TNTs). Under deep-penetrating US irradiation, the nanocomposite generates 1 O2 for sonodynamic therapy and catalyzes the decomposition of endogenous H2 O2 into toxic •OH in the acidic infection microenvironment for catalytic therapy, leading to bacterial cell death. Its robust antibacterial effectiveness is attributable to its bacteria-capturing ability, the biofilm penetrability of positively charged guanidinium, and the subsequent synergistic effect of sonodynamic-catalytic action of Au/TNT. Such a remotely controlled approach potentiates the polarization of macrophages to M2-type while suppressing the M1-type, leading to topical inflammation resolution and enhanced osteoblast proliferation and differentiation to inhibit bone loss. Therefore, this study provides a generic nanotherapeutic approach for efficient sonodynamic-catalytic therapy with respect to osteomyelitis.
Collapse
Affiliation(s)
- Yijie Cheng
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Zhe Zhao
- Department of Surgery of Traditional Chinese Medicine, Tianjin Hospital, Tianjin, 300211, P. R. China
| | - Gang Li
- No. 2 Department Radiology, Tianjin Hospital, Tianjin, 300211, P. R. China
| | - Jie Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Anran Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Yun Xue
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Baolin Zhu
- College of Chemistry, National Demonstration Center for Experimental Chemistry Education (Nankai University), The Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education) and Tianjin Key Lab of Metal and Molecule-based Material Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Zhongming Wu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, P. R. China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
20
|
Abstract
Degradable and environmentally responsive polymers have been actively developed for drug delivery and regenerative medicine applications, yet inadequate consideration of their compatibility with terminal sterilization presents notable barriers to clinical translation. This Review discusses industry-established terminal sterilization methods and aseptic processing and contrasts them with innovative approaches aimed at preserving the integrity of polymeric implants. Regulatory guidelines, fiscal considerations, and potential pitfalls are discussed to encourage early integration of sterility regulatory considerations in material designs.
Collapse
Affiliation(s)
- Chloe K Herczeg
- Department of Orthopedics and Physical Rehabilitation, Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
| | - Jie Song
- Department of Orthopedics and Physical Rehabilitation, Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, United States
| |
Collapse
|
21
|
Moriarty TF, Metsemakers WJ, Morgenstern M, Hofstee MI, Vallejo Diaz A, Cassat JE, Wildemann B, Depypere M, Schwarz EM, Richards RG. Fracture-related infection. Nat Rev Dis Primers 2022; 8:67. [PMID: 36266296 DOI: 10.1038/s41572-022-00396-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 11/09/2022]
Abstract
Musculoskeletal trauma leading to broken and damaged bones and soft tissues can be a life-threating event. Modern orthopaedic trauma surgery, combined with innovation in medical devices, allows many severe injuries to be rapidly repaired and to eventually heal. Unfortunately, one of the persisting complications is fracture-related infection (FRI). In these cases, pathogenic bacteria enter the wound and divert the host responses from a bone-healing course to an inflammatory and antibacterial course that can prevent the bone from healing. FRI can lead to permanent disability, or long courses of therapy lasting from months to years. In the past 5 years, international consensus on a definition of these infections has focused greater attention on FRI, and new guidelines are available for prevention, diagnosis and treatment. Further improvements in understanding the role of perioperative antibiotic prophylaxis and the optimal treatment approach would be transformative for the field. Basic science and engineering innovations will be required to reduce infection rates, with interventions such as more efficient delivery of antibiotics, new antimicrobials, and optimizing host defences among the most likely to improve the care of patients with FRI.
Collapse
Affiliation(s)
- T Fintan Moriarty
- AO Research Institute Davos, Davos, Switzerland.,Center for Musculoskeletal Infections, Department of Orthopaedic and Trauma Surgery, University Hospital Basel, Basel, Switzerland
| | - Willem-Jan Metsemakers
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Mario Morgenstern
- Center for Musculoskeletal Infections, Department of Orthopaedic and Trauma Surgery, University Hospital Basel, Basel, Switzerland
| | | | - Alejandro Vallejo Diaz
- Department of Orthopedics and Traumatology, Hospital Alma Mater de Antioquia, Medellín, Colombia.,Department of Orthopedics and Traumatology, Universidad Pontificia Bolivariana, Medellín, Colombia
| | - James E Cassat
- Department of Paediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Britt Wildemann
- Experimental Trauma Surgery, Department of Trauma, Hand and Reconstructive Surgery, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Melissa Depypere
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Bacteriology and Mycology, KU Leuven, Leuven, Belgium
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - R Geoff Richards
- AO Research Institute Davos, Davos, Switzerland. .,School of Veterinary Science, Aberystwyth University, Aberystwyth, UK.
| |
Collapse
|
22
|
Jin L, Zheng Y, Liu X, Zhang Y, Li Z, Liang Y, Zhu S, Jiang H, Cui Z, Wu S. Magnetic Composite Rapidly Treats Staphylococcus aureus-Infected Osteomyelitis through Microwave Strengthened Thermal Effects and Reactive Oxygen Species. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2204028. [PMID: 36089666 DOI: 10.1002/smll.202204028] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/17/2022] [Indexed: 06/15/2023]
Abstract
It is difficult to effectively treat bacterial osteomyelitis using photothermal therapy or photodynamic therapy due to poor penetration of light. Here, a microwave (MW)-excited magnetic composite of molybdenum disulfide (MoS2 ) / iron oxide (Fe3 O4 ) is reported for the treatment of bacteria-infected osteomyelitis. In in vitro and in vivo experiments, MoS2 /Fe3 O4 is shown to effectively eradicate bacteria-infected mouse tibia osteomyelitis, due to MW thermal enhancement and reactive oxygen species (ROS) (1 O2 and ·O2 - ) production under MW radiation. In addition, the mechanism of MW heat generation is proposed by MW network vector analysis. By the density functional theory and finite element method, the ROS generation mechanism is proposed. The synergy or conductive network between dielectric MoS2 and magnetic Fe3 O4 can reach both enhancement of the dielectric and magnetic attenuation capability. In addition, abundant interfaces are generated to enhance the attenuation of electromagnetic waves by MoS2 and Fe3 O4, introducing multiple reflections and interfacial polarization. Therefore, MoS2 /Fe3 O4 has excellent MW absorption ability based on the synergy or conductive network between MoS2 and magnetic Fe3 O4 as well as multiple dielectric reflections and interfacial polarization.
Collapse
Affiliation(s)
- Liguo Jin
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, P. R. China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, P. R. China
| | - Xiangmei Liu
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, P. R. China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Zhongshan 2nd Road 106#, Guangzhou, 510080, P. R. China
| | - Zhaoyang Li
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, P. R. China
| | - Yanqin Liang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, P. R. China
| | - Shengli Zhu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, P. R. China
| | - Hui Jiang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, P. R. China
| | - Zhenduo Cui
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, P. R. China
| | - Shuilin Wu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, P. R. China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, P. R. China
| |
Collapse
|
23
|
Polymer–Metal Composite Healthcare Materials: From Nano to Device Scale. JOURNAL OF COMPOSITES SCIENCE 2022. [DOI: 10.3390/jcs6080218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Metals have been investigated as biomaterials for a wide range of medical applications. At nanoscale, some metals, such as gold nanoparticles, exhibit plasmonics, which have motivated researchers’ focus on biosensor development. At the device level, some metals, such as titanium, exhibit good physical properties, which could allow them to act as biomedical implants for physical support. Despite these attractive features, the non-specific delivery of metallic nanoparticles and poor tissue–device compatibility have greatly limited their performance. This review aims to illustrate the interplay between polymers and metals, and to highlight the pivotal role of polymer–metal composite/nanocomposite healthcare materials in different biomedical applications. Here, we revisit the recent plasmonic engineered platforms for biomolecules detection in cell-free samples and highlight updated nanocomposite design for (1) intracellular RNA detection, (2) photothermal therapy, and (3) nanomedicine for neurodegenerative diseases, as selected significant live cell–interactive biomedical applications. At the device scale, the rational design of polymer–metallic medical devices is of importance for dental and cardiovascular implantation to overcome the poor physical load transfer between tissues and devices, as well as implant compatibility under a dynamic fluidic environment, respectively. Finally, we conclude the treatment of these innovative polymer–metal biomedical composite designs and provide a future perspective on the aforementioned research areas.
Collapse
|
24
|
Chang B, Zhang L, Wu S, Sun Z, Cheng Z. Engineering single-atom catalysts toward biomedical applications. Chem Soc Rev 2022; 51:3688-3734. [PMID: 35420077 DOI: 10.1039/d1cs00421b] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Due to inherent structural defects, common nanocatalysts always display limited catalytic activity and selectivity, making it practically difficult for them to replace natural enzymes in a broad scope of biologically important applications. By decreasing the size of the nanocatalysts, their catalytic activity and selectivity will be substantially improved. Guided by this concept, the advances of nanocatalysts now enter an era of atomic-level precise control. Single-atom catalysts (denoted as SACs), characterized by atomically dispersed active sites, strikingly show utmost atomic utilization, precisely located metal centers, unique metal-support interactions and identical coordination environments. Such advantages of SACs drastically boost the specific activity per metal atom, and thus provide great potential for achieving superior catalytic activity and selectivity to functionally mimic or even outperform natural enzymes of interest. Although the size of the catalysts does matter, it is not clear whether the guideline of "the smaller, the better" is still correct for developing catalysts at the single-atom scale. Thus, it is clearly a new, urgent issue to address before further extending SACs into biomedical applications, representing an important branch of nanomedicine. This review begins by providing an overview of recent advances of synthesis strategies of SACs, which serve as a basis for the discussion of emerging achievements in improving the enzyme-like catalytic properties at an atomic level. Then, we carefully compare the structures and functions of catalysts at various scales from nanoparticles, nanoclusters, and few-atom clusters to single atoms. Contrary to conventional wisdom, SACs are not the most catalytically active catalysts in specific reactions, especially those requiring multi-site auxiliary activities. After that, we highlight the unique roles of SACs toward biomedical applications. To appreciate these advances, the challenges and prospects in rapidly growing studies of SACs-related catalytic nanomedicine are also discussed in this review.
Collapse
Affiliation(s)
- Baisong Chang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China.
| | - Liqin Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China.
| | - Shaolong Wu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China.
| | - Ziyan Sun
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China.
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China. .,Bohai rim Advanced Research Institute for Drug Discovery, Yantai, 264000, China.,Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University, California 94305, USA
| |
Collapse
|
25
|
Feng X, Lei J, Ma L, Ouyang Q, Zeng Y, Liang H, Lei C, Li G, Tan L, Liu X, Yang C. Ultrasonic Interfacial Engineering of MoS 2 -Modified Zn Single-Atom Catalysts for Efficient Osteomyelitis Sonodynamic Ion Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105775. [PMID: 34889522 DOI: 10.1002/smll.202105775] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/12/2021] [Indexed: 05/23/2023]
Abstract
Osteomyelitis is considered as the most serious bone infection, which can lead to the bone destruction or fatal sepsis. Clinical treatments through frequent antibiotics administration and surgical debridement bring inevitable side effects including drug-resistance and disfigurements. It is urgent to develop an antibiotics-free and rapid strategy to treat osteomyelitis. Herein, a bifunctional sonosensitizer that consists of porphyrin-like Zn single-atom catalysts (g-ZnN4 ) and MoS2 quantum dots is developed, which exhibits excellent sonodynamic antibacterial efficiency and osteogenic ability. It is found that the construction of heterogeneous interfaces of g-ZnN4 -MoS2 fully activates the adsorbed O2 due to the increased interface charge transfer, enhanced spin-flip, and reduced activation energy of O2 . The generated 1 O2 can kill methicillin-resistant Staphylococcus aureus (MRSA) with an antibacterial efficiency of 99.58% under 20 min of ultrasound (US) irradiation. The Zn single atoms immobilized in g-ZnN4 can be released steadily in the form of Zn2+ for 28 days within safe concentration, realizing the great osteoinductive ability of such a sonosensitizer. For the treatment of MRSA-infected osteomyelitis, the inflammation and bone loss can be significantly suppressed through sonodynamic ion therapy. This work provides another strategy for developing high efficiency sonosensitizer through ultrasound interfacial engineering.
Collapse
Affiliation(s)
- Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jie Lei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qunle Ouyang
- Biomedical Materials Engineering Research Center, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan, 430062, China
| | - Yuxuan Zeng
- Biomedical Materials Engineering Research Center, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan, 430062, China
| | - Hang Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chunchi Lei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lei Tan
- Biomedical Materials Engineering Research Center, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan, 430062, China
| | - Xiangmei Liu
- School of Life Science and Health Engineering, Hebei University of Technology, Xiping Avenue 5340, Beichen District, Tianjin, 300401, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
26
|
Fitting pieces into the puzzle: The impact of titanium-based dental implant surface modifications on bacterial accumulation and polymicrobial infections. Adv Colloid Interface Sci 2021; 298:102551. [PMID: 34757285 DOI: 10.1016/j.cis.2021.102551] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/09/2021] [Accepted: 10/17/2021] [Indexed: 12/12/2022]
Abstract
Polymicrobial infection is the main cause of dental implant failure. Although numerous studies have reported the ability of titanium (Ti) surface modifications to inhibit microbial adhesion and biofilm accumulation, the majority of solutions for the utilization of Ti antibacterial surfaces have been testedin in vitro and animal models, with only a few developed surfaces progressing into clinical research. Motivated by this huge gap, we critically reviewed the scientific literature on the existing antibacterial Ti surfaces to help understand these surfaces' impact on the "puzzle" of undesirable dental implant-related infections. This manuscript comprises three main sections: (i) a narrative review on topics related to oral biofilm formation, bacterial-implant surface interactions, and on how implant-surface modifications can influence microbial accumulation; (ii) a critical evidence-based review to summarize pre-clinical and clinical studies in an attempt to "fit pieces into the puzzle" to unveil the best way to reduce microbial loads and control polymicrobial infection around dental implants showed by the current in vivo evidence; and (iii) discussion and recommendations for future research testing emerging antibacterial implant surfaces, connecting basic science and the requirements for future clinical translation. The findings of the present review suggest no consensus regarding the best available Ti surface to reduce bacterial colonization on dental implants. Smart release or on-demand activation surface coatings are a "new piece of the puzzle", which may be the most effective alternative for reducing microbial colonization on Ti surfaces, and future studies should focus on these technologies.
Collapse
|
27
|
Cai D, Chen S, Wu B, Chen J, Tao D, Li Z, Dong Q, Zou Y, Chen Y, Bi C, Zu D, Lu L, Fang B. Construction of multifunctional porcine acellular dermal matrix hydrogel blended with vancomycin for hemorrhage control, antibacterial action, and tissue repair in infected trauma wounds. Mater Today Bio 2021; 12:100127. [PMID: 34585135 PMCID: PMC8452890 DOI: 10.1016/j.mtbio.2021.100127] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/29/2021] [Accepted: 08/16/2021] [Indexed: 12/18/2022] Open
Abstract
Prevention of bacterial infection and reduction of hemorrhage, the primary challenges posed by trauma before hospitalization, are essential steps in prolonging the patient's life until they have been transported to a trauma center. Extracellular matrix (ECM) hydrogel is a promising biocompatible material for accelerating wound closure. However, due to the lack of antibacterial properties, this hydrogel is difficult to be applied to acute contaminated wounds. This study formulates an injectable dermal extracellular matrix hydrogel (porcine acellular dermal matrix (ADM)) as a scaffold for skin defect repair. The hydrogel combines vancomycin, an antimicrobial agent for inducing hemostasis, expediting antimicrobial activity, and promoting tissue repair. The hydrogel possesses a porous structure beneficial for the adsorption of vancomycin. The antimicrobial agent can be timely released from the hydrogel within an hour, which is less than the time taken by bacteria to infest an injury, with a cumulative release rate of approximately 80%, and thus enables a relatively fast bactericidal effect. The cytotoxicity investigation demonstrates the biocompatibility of the ADM hydrogel. Dynamic coagulation experiments reveal accelerated blood coagulation by the hydrogel. In vivo antibacterial and hemostatic experiments on a rat model indicate the healing of infected tissue and effective control of hemorrhaging by the hydrogel. Therefore, the vancomycin-loaded ADM hydrogel will be a viable biomaterial for controlling hemorrhage and preventing bacterial infections in trauma patients.
Collapse
Affiliation(s)
- D Cai
- Department of Spine Surgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - S Chen
- Department of Spine Surgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - B Wu
- Department of Spine Surgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - J Chen
- Bacterial Laboratory, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - D Tao
- Pathology Department, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - Z Li
- Pathology Department, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - Q Dong
- Department of Spine Surgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - Y Zou
- Department of Spine Surgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - Y Chen
- Department of Spine Surgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China.,School of Medcine, Shaoxing University, Shaoxing, China
| | - C Bi
- Department of Spine Surgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China.,School of Medcine, Shaoxing University, Shaoxing, China
| | - D Zu
- Central Laboratory, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - L Lu
- Department of Spine Surgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| | - B Fang
- Department of Spine Surgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, China
| |
Collapse
|
28
|
Jiménez T, Botero J, Otaegui D, Calvo J, Hernandez FJ, San Sebastian E. Rational Design and Experimental Analysis of Short-Oligonucleotide Substrate Specificity for Targeting Bacterial Nucleases. J Med Chem 2021; 64:12855-12864. [PMID: 34460263 PMCID: PMC8436206 DOI: 10.1021/acs.jmedchem.1c00884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
An undecamer oligonucleotide probe
based on a pair of deoxythymidines
flanked by several modified nucleotides is a specific and highly efficient
biosensor for micrococcal nuclease (MNase), an endonuclease produced
by Staphylococcus aureus. Herein, the
interaction mode and cleavage process on such oligonucleotide probes
are identified and described for the first time. Also, we designed
truncated pentamer probes as the minimum-length substrates required
for specific and efficient biosensing. By means of computational (virtual
docking) and experimental (ultra-performance liquid chromatography–mass
spectrometry and matrix-assisted laser desorption ionization time-of-flight)
techniques, we perform a sequence/structure–activity relationship
analysis, propose a catalytically active substrate–enzyme complex,
and elucidate a novel two-step phosphodiester bond hydrolysis mechanism,
identifying the cleavage sites and detecting and quantifying the resulting
probe fragments. Our results unravel a picture of both the enzyme–biosensor
complex and a two-step cleavage/biosensing mechanism, key to the rational
oligonucleotide design process.
Collapse
Affiliation(s)
- Tania Jiménez
- Somaprobes S.L, Mikeletegi Pasealekua, 83, 20009 Donostia, Gipuzkoa, Spain
| | - Juliana Botero
- Somaprobes S.L, Mikeletegi Pasealekua, 83, 20009 Donostia, Gipuzkoa, Spain.,Applied Chemistry Department, University of the Basque Country (UPV/EHU), 20018 San Sebastián, Spain
| | - Dorleta Otaegui
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastian 20014, Spain
| | - Javier Calvo
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastian 20014, Spain
| | - Frank J Hernandez
- Wallenberg Center for Molecular Medicine (WCMM), 58185 Linköping, Sweden.,Department of Physics, Chemistry and Biology, Linköping University, 58185 Linköping, Sweden
| | - Eider San Sebastian
- Applied Chemistry Department, University of the Basque Country (UPV/EHU), 20018 San Sebastián, Spain
| |
Collapse
|
29
|
Yu Y, Tan L, Li Z, Liu X, Zheng Y, Feng X, Liang Y, Cui Z, Zhu S, Wu S. Single-Atom Catalysis for Efficient Sonodynamic Therapy of Methicillin-Resistant Staphylococcus aureus-Infected Osteomyelitis. ACS NANO 2021; 15:10628-10639. [PMID: 34086433 DOI: 10.1021/acsnano.1c03424] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Osteomyelitis, as a severe bone disease caused by bacterial infection, can result in lifelong disability or fatal sepsis. Considering that the infection is stubborn and deep-sited in bone tissue, in situ and rapid treatments for osteomyelitis remain a significant challenge. Herein, we prepare an ultrasound (US)-activated single-atom catalyst that consists of a Au nanorod (NRs)-actuated single-atom-doped porphyrin metal-organic framework (HNTM-Pt@Au) and red cell membrane (RBC), which can efficiently treat methicillin-resistant Staphylococcus aureus (MRSA)-infected osteomyelitis under US. Besides the outstanding performance in the field of photocatalysis, we find that single atoms (such as Pt, Au, Cu) also improve the sonocatalytic ability of the sonosensitizer. Due to the strong electron-trapping and oxygen adsorption capacity, the Pt single atom endows RBC-HNTM-Pt@Au with an excellent sonocatalytic activity. It shows an excellent antibacterial performance with an antibacterial efficiency of 99.9% toward MRSA under 15 min of US irradiation. Meanwhile, the RBC-HNTM-Pt@Au can be propelled directionally under US and thus dynamically neutralize the secreted toxins. The MRSA-infected osteomyelitis in rat tibia was successfully treated, which shows negligible bone loss, reduced inflammation response, and great biocompatibility. This work presents an efficient sonodynamic therapy for the treatment of deep tissue infections via a multifunctional single-atom catalyst.
Collapse
Affiliation(s)
- Yi Yu
- Biomedical Materials Engineering Research Center, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Lei Tan
- Biomedical Materials Engineering Research Center, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Zhaoyang Li
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Xiangmei Liu
- School of Life Science and Health Engineering, Hebei University of Technology, Xiping Avenue 5340, Beichen District, Tianjin 300401, China
| | - Yufeng Zheng
- State Key Laboratory for Turbulence and Complex System and Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing 0087, China
| | - Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanqin Liang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Shengli Zhu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Shuilin Wu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| |
Collapse
|
30
|
Peng J, Liu P, Peng W, Sun J, Dong X, Ma Z, Gan D, Liu P, Shen J. Poly(hexamethylene biguanide) (PHMB) as high-efficiency antibacterial coating for titanium substrates. JOURNAL OF HAZARDOUS MATERIALS 2021; 411:125110. [PMID: 33858091 DOI: 10.1016/j.jhazmat.2021.125110] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 06/12/2023]
Abstract
Bacterial associated infection is a remaining urgent challenge in clinic application of metallic implants and devices. Here, we developed a new strategy to combat the bacterial associated infection of titanium alloy (TC4). Novel phosphonate/active ester block polymers (pDEMMP-b-pNHSMA) with identical phosphonate segments (DP = 29) as the metal anchorable ligand but varied active ester segments (DPs = 7, 29, and 64) as the conjugation site for poly(hexamethylene biguanide) (PHMB) were precisely prepared. Through a facile two-step process, the polymeric coating were successfully constructed on TC4 substrates as evidenced by water contact angle and XPS measurements. Through systematical in vitro antibacterial evaluations, robust relationship between the chemical structure of coating polymer and the antibacterial property endowed to the TC4 substrates has been established. Results showed that the block polymer, bearing an active ester segment of 64 repeat units, enabled dense packing of PHMB coating on the TC4 surface, which is able to kill 100% of both S. aureus and E. coli. that seeded without compromising the cytocompatibility of TC4 substrates. Furthermore, PHMB coating could significantly inhibit the colony of the bacteria and consequently reduce the bacterial associated inflammatory reaction as verified by a subcutaneous infection model on rat.
Collapse
Affiliation(s)
- Jiangmei Peng
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Peiming Liu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Wan Peng
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Jin Sun
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Xiaohan Dong
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Zhuangzhuang Ma
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Donglin Gan
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Pingsheng Liu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China.
| | - Jian Shen
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China; Jiangsu Engineering Research Center of Interfacial Chemistry, Nanjing University, Nanjing 210093, PR China.
| |
Collapse
|
31
|
Ghimire A, Song J. Anti-Periprosthetic Infection Strategies: From Implant Surface Topographical Engineering to Smart Drug-Releasing Coatings. ACS APPLIED MATERIALS & INTERFACES 2021; 13:20921-20937. [PMID: 33914499 PMCID: PMC8130912 DOI: 10.1021/acsami.1c01389] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Despite advanced implant sterilization and aseptic surgical techniques, periprosthetic bacterial infection remains a major challenge for orthopedic and dental implants. Bacterial colonization/biofilm formation around implants and their invasion into the dense skeletal tissue matrices are difficult to treat and could lead to implant failure and osteomyelitis. These complications require major revision surgeries and extended antibiotic therapies that are associated with high treatment cost, morbidity, and even mortality. Effective preventative measures mitigating risks for implant-related infections are thus in dire need. This review focuses on recent developments of anti-periprosthetic infection strategies aimed at either reducing bacterial adhesion, colonization, and biofilm formation or killing bacteria directly in contact with and/or in the vicinity of implants. These goals are accomplished through antifouling, quorum-sensing interfering, or bactericidal implant surface topographical engineering or surface coatings through chemical modifications. Surface topographical engineering of lotus leaf mimicking super-hydrophobic antifouling features and cicada wing-mimicking, bacterium-piercing nanopillars are both presented. Conventional physical coating/passive release of bactericidal agents is contrasted with their covalent tethering to implant surfaces through either stable linkages or linkages labile to bacterial enzyme cleavage or environmental perturbations. Pros and cons of these emerging anti-periprosthetic infection approaches are discussed in terms of their safety, efficacy, and translational potentials.
Collapse
Affiliation(s)
- Ananta Ghimire
- Department of Orthopedics and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jie Song
- Department of Orthopedics and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
32
|
Garg D, Matai I, Sachdev A. Toward Designing of Anti-infective Hydrogels for Orthopedic Implants: From Lab to Clinic. ACS Biomater Sci Eng 2021; 7:1933-1961. [PMID: 33826312 DOI: 10.1021/acsbiomaterials.0c01408] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
An alarming increase in implant failure incidence due to microbial colonization on the administered orthopedic implants has become a horrifying threat to replacement surgeries and related health concerns. In essence, microbial adhesion and its subsequent biofilm formation, antibiotic resistance, and the host immune system's deficiency are the main culprits. An advanced class of biomaterials termed anti-infective hydrogel implant coatings are evolving to subdue these complications. On this account, this review provides an insight into the significance of anti-infective hydrogels for preventing orthopedic implant associated infections to improve the bone healing process. We briefly discuss the clinical course of implant failure, with a prime focus on orthopedic implants. We identify the different anti-infective coating strategies and hence several anti-infective agents which could be incorporated in the hydrogel matrix. The fundamental design criteria to be considered while fabricating anti-infective hydrogels for orthopedic implants will be discussed. We highlight the different hydrogel coatings based on the origin of the polymers involved in light of their antimicrobial efficacy. We summarize the relevant patents reported in the prevention of implant infections, including orthopedics. Finally, the challenges concerning the clinical translation of the aforesaid hydrogels are described, and considerable solutions for improved clinical practice and better future prospects are proposed.
Collapse
Affiliation(s)
- Deepa Garg
- Central Scientific Instruments Organisation (CSIR-CSIO), Chandigarh-160030, India.,Academy of Scientific and Innovative Research, CSIR-CSIO, Chandigarh-160030, India
| | - Ishita Matai
- Central Scientific Instruments Organisation (CSIR-CSIO), Chandigarh-160030, India.,Academy of Scientific and Innovative Research, CSIR-CSIO, Chandigarh-160030, India
| | - Abhay Sachdev
- Central Scientific Instruments Organisation (CSIR-CSIO), Chandigarh-160030, India.,Academy of Scientific and Innovative Research, CSIR-CSIO, Chandigarh-160030, India
| |
Collapse
|
33
|
Souza JGS, Bertolini MM, Costa RC, Nagay BE, Dongari-Bagtzoglou A, Barão VAR. Targeting implant-associated infections: titanium surface loaded with antimicrobial. iScience 2021; 24:102008. [PMID: 33490916 PMCID: PMC7811145 DOI: 10.1016/j.isci.2020.102008] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Implant devices have = proven a successful treatment modality in reconstructive surgeries. However, increasing rates of peri-implant diseases demand further examination of their pathogenesis. Polymicrobial biofilm formation on titanium surfaces has been considered the main risk factor for inflammatory processes on tissues surrounding implant devices, which often lead to implant failure. To overcome microbial accumulation on titanium surfaces biofilm targeting strategies have been developed to modify the surface and incorporate antimicrobial coatings. Because antibiotics are widely used to treat polymicrobial infections, these agents have recently started to be incorporated on titanium surface. This review discusses the biofilm formation on titanium dental implants and key factors to be considered in therapeutic and preventative strategies. Moreover, a systematic review was conducted on coatings developed for titanium surfaces using different antibiotics. This review will also shed light on potential alternative strategies aiming to reduce microbial loads and control polymicrobial infection on implanted devices.
Collapse
Affiliation(s)
- João Gabriel Silva Souza
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
- Dental Research Division, Guarulhos University, Guarulhos, SP 07023-070, Brazil
- Dentistry Science School (Faculdade de Ciências Odontológicas - FCO), Montes Claros, Minas Gerais, 39401-303, Brazil
| | - Martinna Mendonça Bertolini
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Raphael Cavalcante Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| | - Bruna Egumi Nagay
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| | - Anna Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Valentim Adelino Ricardo Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| |
Collapse
|
34
|
Zhang S, Wang X, Shi X, Tan H. Susceptibility of Mature Staphylococcus Biofilms to Chinese Herbal Decoction Sanhuang Jiedu: An In Vitro Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7473942. [PMID: 33062698 PMCID: PMC7539100 DOI: 10.1155/2020/7473942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/18/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND External socking and washing with the Chinese herbal Sanhuang Jiedu decoction (SHJD) can effectively control local limb infections with bone and implant exposure. However, the antibiofilm activities of this decoction in vitro have not yet been investigated. Therefore, the aim of this study was to examine the effects and characteristics of SHJD on the mature biofilms of multidrug-resistant staphylococci on a titanium surface. METHODS Biofilm-forming methicillin-resistant Staphylococcus epidermidis ATCC 35984 and S. aureus ATCC 43330, and non-biofilm-forming S. epidermidis ATCC 12228 were selected as the experimental strains. The mature biofilms were prepared on titanium surfaces. The five experimental groups were based on dilution concentrations (DC) of SHJD: the control group (biofilm incubated with 0.85% NaCl solution), the SHJD (DC:1/8) group (initial SHJD solution was diluted 1/8), the SHJD (DC:1/4) group, the SHJD (DC:1/2) group, and the SHJD (DC:1/1) group (initial SHJD solution). The effects of SHJD on the mature biofilms were observed with the bacterial spread plate method, crystal violet (CV) staining, scanning electron microscopy, and confocal laser scanning microscopy. RESULTS After culture in tryptic soy broth for 72 h, ATCC 43300 and ATCC 35984 produced mature biofilms and ATCC 12228 did not. The optical density value of ATCC 12228 was 0.11 ± 0.02, significantly lower than that of ATCC 35984 (0.42 ± 0.05) or ATCC 43300 (0.41 ± 0.03) (P < 0.05). The mature biofilms of ATCC 43300 and ATCC 35984 clearly disintegrated when incubated for 12-24 h with SHJD (DC:1/1) or SHJD (DC:1/2), showing only scattered bacterial adhesion. In the SHJD (DC:1/4) group, although many residual bacterial colonies still clustered together, presenting a biofilm structure, it was very looser than that in the SHJD (DC:1/8) group in which the biofilm was similar to that in the control group. For ATCC 12228, only colony adhesion was observed, and the number of colonies decreased as the concentration of SHJD or the culture period increased. The quantitative results for the bacterial spread plate and CV staining showed significant differences between the SHJD groups (P < 0.05). CONCLUSION SHJD has antibiofilm activity against multidrug-resistant Staphylococcus strains. It weakens or disrupts already-formed mature biofilms on titanium surfaces in a concentration- and incubation time-dependent manner.
Collapse
Affiliation(s)
- Shaoe Zhang
- Henan Orthopedic Institute, Henan Luoyang Orthopedic-Traumatological Hospital (Henan Orthopedic Hospital), Luoyang 471000, China
| | - Xiao Wang
- Henan Orthopedic Institute, Henan Luoyang Orthopedic-Traumatological Hospital (Henan Orthopedic Hospital), Luoyang 471000, China
| | - Xiaotao Shi
- Henan Orthopedic Institute, Henan Luoyang Orthopedic-Traumatological Hospital (Henan Orthopedic Hospital), Luoyang 471000, China
| | - Honglue Tan
- Henan Orthopedic Institute, Henan Luoyang Orthopedic-Traumatological Hospital (Henan Orthopedic Hospital), Luoyang 471000, China
| |
Collapse
|
35
|
Zhang B, Skelly JD, Braun BM, Ayers DC, Song J. Surface-grafted zwitterionic polymers improve the efficacy of a single antibiotic injection in suppressing S. aureus periprosthetic infections. ACS APPLIED BIO MATERIALS 2020; 3:5896-5904. [PMID: 34368642 DOI: 10.1021/acsabm.0c00600] [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] [Indexed: 12/13/2022]
Abstract
Implant-associated bacterial infections are difficult to treat due to the tendency of biofilm formation on implant surfaces, which protects embedded pathogens from host defense and impedes antibiotic penetration, rendering systemic antibiotic injections ineffective. Here, we test the hypothesis that implant coatings that reduce bacterial colonization would make planktonic bacteria within the periprosthetic environment more susceptible to conventional systemic antibiotic treatment. We covalently grafted zwitterionic polymer brushes poly(sulfobetaine methacryate) from Ti6Al4V surface to increase the substrate surface hydrophilicity and reduce staphylococcus aureus (S. aureus) adhesion. Using a mouse femoral intramedullary (IM) canal infection model, we showed that the anti-fouling coating applied to Ti6Al4V IM implants, when combined with a single vancomycin systemic injection, significantly suppressed both bacterial colonization on implant surfaces and the periprosthetic infections, outperforming either treatment alone. This work supports the hypothesis that grafting anti-fouling polymers to implant surfaces improves the efficacy of systemic antibiotic injections to combat periprosthetic infections.
Collapse
Affiliation(s)
- Ben Zhang
- Department of Orthopedics & Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jordan D Skelly
- Department of Orthopedics & Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Benjamin M Braun
- Department of Orthopedics & Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - David C Ayers
- Department of Orthopedics & Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jie Song
- Department of Orthopedics & Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
36
|
Dhar Y, Han Y. Current developments in biofilm treatments: Wound and implant infections. ENGINEERED REGENERATION 2020. [DOI: 10.1016/j.engreg.2020.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|