1
|
Gai T, Zhang H, Hu Y, Li R, Wang J, Chen X, Wang J, Chen Z, Jing Y, Wang C, Bai L, Wang X, Su J. Sequential construction of vascularized and mineralized bone organoids using engineered ECM-DNA-CPO-based bionic matrix for efficient bone regeneration. Bioact Mater 2025; 49:362-377. [PMID: 40144795 PMCID: PMC11937690 DOI: 10.1016/j.bioactmat.2025.02.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Given the limitations of allogeneic and artificial bone grafts, bone organoids have attracted extensive attention for their physiological properties that closely resemble natural bone, offering great potential to bone reconstruction for critical-sized bone defects. Although early-stage bone organoids such as osteo-callus organoids and woven bone organoids have been reported, functional bone organoids with vascularization and mineralization are currently unavailable due to the lack of bone-mimicking matrix and dynamic culture systems suitable for the long-term cultivation of mature bone organoids. Herein, a novel engineered bionic matrix hydrogels with multifunctional components and double network structure are developed by incorporating calcium phosphate oligomers (CPO) into a combination of bone-derived decellularized extracellular matrix (ECM) and salmon-derived deoxyribonucleic acid (DNA) via photo-crosslinking and dynamic self-assembly strategies. This kind of bionic matrix hydrogels facilitate recruitment, proliferation, osteogenesis and angiogenesis of bone marrow mesenchymal stromal cells (BMSCs). More importantly, vascularized and mineralized bone organoids are sequentially constructed using BMSCs-loaded engineered bionic matrix hydrogels via in vitro dynamic culture and in vivo heterotopic ossification. Meanwhile, this kind of engineered bionic matrix are capable of achieving efficient bone repair for cranial defect. These findings suggest that engineered bionic matrix hydrogels combined with such dynamic culture system, providing a promising strategy for functional bone organoids construction.
Collapse
Affiliation(s)
- Tingting Gai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ruiyang Li
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jian Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jianhua Wang
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhenhua Chen
- Yantai Zhenghai Bio-tech Co., Ltd, Yantai, 264006, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Chenglong Wang
- Yantai Zhenghai Bio-tech Co., Ltd, Yantai, 264006, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiuhui Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
2
|
Song Y, Li H, Yuan Y, Zhang D, Wang Z, Qi B, Jiang P, Yu A. Synergistic photothermal-sonodynamic therapy for antibacterial and immune reprogramming in chronic osteomyelitis. J Control Release 2025; 381:113612. [PMID: 40073945 DOI: 10.1016/j.jconrel.2025.113612] [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: 01/03/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
The development of antibiotic resistance and inadequate immune response in chronic inflammation pose significant challenges in treating chronic osteomyelitis. As accepted non-antibiotic antimicrobial therapies, sonodynamic therapy (SDT) and photothermal therapy (PTT) are recognized for their effectiveness in eliminating bacteria and promoting tissue repair, rendering them promising therapeutic strategies for treating bacterial infections and preventing the emergence of drug-resistant bacteria. However, the antimicrobial action and efficacy in promoting tissue repair depend on the activation status of the host immune system. In this study, by encapsulating horseradish peroxidase (HRP)-loaded gold/polydopamine (PDA) nanoparticles within DOTAP/DOPE cationic liposomes (DLPs), a novel multifunctional nanocatalyst, Au/PDA/HRP@DLP (APH@DLP), was developed to achieve antimicrobial effects and immunological reprogramming of chronic osteomyelitis through synergistic SDT and PTT. The impact on immune activation was investigated by assessing the anti-infective and healing effects in osteomyelitis rat models. The release of bacterial-associated antigens during treatment serves as an in situ vaccine, activating antigen-presenting cells and further stimulating adaptive immunity, while also inducing immune memory that significantly reduces the risk of recurrence. Additionally, macrophage phenotypic transformation during SDT and PTT facilitates tissue repair. This study highlights the role of immune activation in SDT/PTT-based antimicrobial therapy and suggests new strategies for treating chronic osteomyelitis.
Collapse
Affiliation(s)
- Yuchen Song
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Haimei Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan 430072, China
| | - Ying Yuan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Dong Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Baiwen Qi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Peng Jiang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan 430072, China.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
3
|
Li K, Xie E, Liu C, Hu J, Chen Q, Li J, Wang H, Meng Q, Liu D, Meng B, Liang T, Ma J, Yuan Z, Wang L, Shu W, Mao H, Han F, Li B. "Disguise strategy" to bacteria: A multifunctional hydrogel with bacteria-targeting and photothermal conversion properties for the repair of infectious bone defects. Bioact Mater 2025; 47:343-360. [PMID: 40026823 PMCID: PMC11870027 DOI: 10.1016/j.bioactmat.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/16/2025] [Accepted: 02/02/2025] [Indexed: 03/05/2025] Open
Abstract
Addressing the challenge of eliminating bacteria and stimulating osteogenesis in infectious bone defects, where cells and bacteria coexist within the microenvironment, presents a significant hurdle. In this study, a strategy of targeting bacteria is proposed to address this challenge. For this purpose, a methacrylated gelatin composite hydrogel containing zinc ion and D-type cysteine-modified polydopamine nanoparticles (PZC) is developed. The D-cysteine, involved in the metabolism of the bacterial peptidoglycan chain, allows PZC to specifically target bacteria, exhibiting a form of "disguise strategy". Through the targeting effect, this composite hydrogel can selectively kill bacteria and promote osteogenesis combing photothermal therapy with Zn2+ release, which showcases spatial controllability. Moreover, the antibacterial ability will be further improved after Near-infrared light irradiation. The multifunctional hydrogel containing Zn2+ modified nanoparticles can also promote osteogenic differentiation of bone marrow stem cells. Animal studies have revealed that the multifunctional hydrogel can inhibit bacteria growth and promote repair of infectious bone defects in rats. Findings from this study imply that endowing the nanoparticles with bacteria-targeting function can precisely control the events in cells and bacteria in the complex microenvironment, which can provide insights for the treatment of complex diseases with antibacterial requirements.
Collapse
Affiliation(s)
- Kexin Li
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - En Xie
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Chengyuan Liu
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Jie Hu
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Qianglong Chen
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Jiaying Li
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Huan Wang
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Qingchen Meng
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Dachuan Liu
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Bin Meng
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Ting Liang
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Jinjin Ma
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Zhangqin Yuan
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Lijie Wang
- Sanitation & Environment Technology Institute of Soochow University Ltd., Suzhou, Jiangsu, 215000, PR China
| | - Wenmiao Shu
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, G1 1QE, UK
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, PR China
| | - Fengxuan Han
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| |
Collapse
|
4
|
Zhou Z, Zhang P, Chen D, Kong N, Liu H, Liang J, Huang K, Wang H. Cecropin A-Derived Peptide for the Treatment of Osteomyelitis by Inhibiting the Growth of Multidrug-Resistant Bacteria and Eliminating Inflammation. ACS NANO 2025. [PMID: 40231707 DOI: 10.1021/acsnano.4c18858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Osteomyelitis poses substantial therapeutic challenges due to the prevalence of multidrug-resistant bacterial infections and associated inflammation. Current treatment regimens often rely on a combination of corticosteroids and antibiotics, which can lead to complications and impede effective bacterial clearance. In this study, we present CADP-10, a Cecropin A-derived peptide, designed to target methicillin-resistant Staphylococcus aureus (MRSA) and multidrug-resistant Escherichia coli (MRE), while simultaneously addressing inflammatory responses. CADP-10 self-assembles into nanobacterial net (NBacN) that selectively identify and bind to bacterial endotoxins (LPS and LTA), disrupting membrane integrity and depolarizing membrane potential, which culminates in bacterial death. Importantly, these NBacN are bound to LPS and LTA from dead bacteria, preventing their engagement with TLR receptors and effectively blocking downstream inflammatory pathways. Our assessments of CADP-10 demonstrate good biosafety in both in vitro and in vivo models. Notably, in a rabbit osteomyelitis model, CADP-10 eliminated MRSA-induced bone infections, mitigated inflammation, and promoted bone tissue regeneration. This research highlights the potential of CADP-10 as a multifunctional antimicrobial agent for the management of infectious inflammatory diseases.
Collapse
Affiliation(s)
- Ziao Zhou
- Department of Chemistry, Zhejiang University, Hangzhou 310027, Zhejiang Province, China
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Peng Zhang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Dinghao Chen
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Nan Kong
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Huayang Liu
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Juan Liang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Kai Huang
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Hangzhou 310012, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| |
Collapse
|
5
|
Chen F, Schiffer NE, Song J. Animal Models of Orthopedic Implant-Associated Infections and Revisions. ACS Biomater Sci Eng 2025; 11:2052-2068. [PMID: 40125564 DOI: 10.1021/acsbiomaterials.4c02331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Orthopedic implant-associated infections such as prosthetic joint infections (PJIs) lead to devastating complications for patients and impose significant financial burdens on the healthcare systems. Although the primary orthopedic implant associated infection rate is relatively low (0.3-9%), the reinfection rate after implant revisions can be as high as 20% to 40%. To evaluate novel therapeutic strategies for preventing and treating infections associated with primary and revision implants, it is essential to develop appropriate animal models that closely emulate clinical realities. Here we discuss existing animal models developed for orthopedic implant revision surgeries including small animal models in rats and mice, and larger animal models in rabbits, sheep, and mini-pigs. While larger animal models offer the advantage of more closely mimicking human surgical procedures, implant dimensions, and infection treatment protocols, rodent models are more cost-effective and better suited for screening experimental prophylaxes and therapeutics. Existing animal revision models have focused on primary infections established by Staphylococcal aureus (S. aureus) and revisions involving both one-stage and two-stage procedures. Further development of smaller animal implant revision models that implement more clinically relevant surgical procedures and recapitulate polymicrobial infections could facilitate the discovery and more rigorous evaluation of novel implant coating prophylaxes and therapeutics for reducing reinfection rates following implant revisions.
Collapse
Affiliation(s)
- Feiyang Chen
- Department of Orthopedics and Physical Rehabilitation, Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, Massachusetts 01655, United States
| | - Naomi E Schiffer
- Department of Orthopedics and Physical Rehabilitation, Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, Massachusetts 01655, United States
| | - Jie Song
- Department of Orthopedics and Physical Rehabilitation, Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, Massachusetts 01655, United States
| |
Collapse
|
6
|
Liu K, Liu Y, Wang Q, Nazaré M, Zhang L, Pan X, Hu HY. PaAP-Activatable NIR Probe for Diagnosing, Imaging, and Discovering Small-Molecule Therapeutics against Implant-Associated Biofilm Infections. J Med Chem 2025; 68:7827-7838. [PMID: 40138192 DOI: 10.1021/acs.jmedchem.5c00588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Biofilm formation on medical implants causes implant-associated infections (IAIs), leading to high morbidity and mortality. Developing molecular tools for precise biofilm detection, along with novel strategies and agents to target biofilm-related IAIs, is crucial for improving treatment options and patient outcomes. Pseudomonas aeruginosa aminopeptidase (PaAP), a key biofilm-associated virulence factor, is a promising target for combating infections. Here, we developed a PaAP-activatable near-infrared (NIR) fluorescent probe, Hcy-NEO-Leu, for real-time, specific, and sensitive detection of PaAP activity. This probe enables noninvasive imaging of the P. aeruginosa biofilm in vitro and in vivo. The probe also identified LY-58, a lycorine derivative that disrupts biofilm formation without affecting bacterial growth or mammalian cell viability, enhancing tobramycin penetration and overcoming antibiotic resistance. This study introduces LY-58 as a promising adjunctive therapy. In conclusion, the PaAP-activatable NIR imaging probe, combined with LY-58, offers innovative tools for the early diagnosis and effective treatment of IAIs.
Collapse
Affiliation(s)
- Kaixuan Liu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yang Liu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qinghua Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Marc Nazaré
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Leilei Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiandao Pan
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
7
|
Ake B, Yang H, Yang H, Liu H, Gui X, Liu T, Chen J, Liu J, Zhou W, Qu B, Zeng Z, Zhou C. Ultrasound-responsive smart biomaterials for bone tissue engineering. J Mater Chem B 2025; 13:4527-4543. [PMID: 40111085 DOI: 10.1039/d5tb00109a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Bone defects resulting from trauma, tumors, or other injuries significantly impact human health and quality of life. However, current treatments for bone defects are constrained by donor shortages and immune rejection. Bone tissue engineering has partially alleviated the limitations of traditional bone repair methods. The development of smart biomaterials that can respond to external stimuli to modulate the biofunctions has become a prominent area of research. Ultrasound technology is regarded as an optimal "remote controller" and "trigger" for bone repair biomaterials. This review reports the comprehensive and systematic overview of ultrasound-responsive bone repair smart biomaterials. It presents the fundamental theories of bone repair, the definition of ultrasound, and its applications. Furthermore, the review summarizes the ultrasound effect mechanisms of biomaterials and their roles in bone repair, including detailed studies on anti-inflammation, immunomodulation, and cell therapy. Finally, the advantages of ultrasound-responsive smart biomaterials and their future prospects in this field are discussed.
Collapse
Affiliation(s)
- Bicheng Ake
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Hongsheng Yang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Hao Yang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Hao Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Xingyu Gui
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 610064, Chengdu, China
| | - Taoyu Liu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 610064, Chengdu, China
| | - Jie Chen
- Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jia Liu
- The People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Wenzheng Zhou
- The People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Bo Qu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Zhimou Zeng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 610064, Chengdu, China
| |
Collapse
|
8
|
Li Z, Zhang Q, Lian X, Yin C, Lin Y, Wang Y, Han Z, Shen F, Xu Y, Wang H, Wang T. Value of metagenomic next-generation sequencing in the diagnosis of native pyogenic spinal infections: a multicenter, retrospective observational study. Spine J 2025; 25:640-648. [PMID: 39615696 DOI: 10.1016/j.spinee.2024.11.004] [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: 08/04/2024] [Revised: 10/21/2024] [Accepted: 11/05/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND CONTEXT The etiological diagnosis of pyogenic spinal infection is crucial for its precise antibiotic treatment. Traditional methods of detection are often slow and ineffective. In recent times, metagenomic next-generation sequencing (mNGS) has revolutionized pathogen detection, offering a more effective approach to disease management. PURPOSE Comparing mNGS with microbial culture to comprehensively explore the diagnostic value of mNGS in pyogenic spinal infections. DESIGN A multicenter, retrospective observational study. PATIENT SAMPLE In a multicenter retrospective observational study, we analyzed the data from 301 patients admitted in four selected hospitals with pyogenic spinal infections from December 2019 to February 2024. OUTCOME MEASURES Identification of pathogenic bacteria in patients. METHODS Obtain blood and lesion tissue or pus samples from the enrolled patients for microbial culture, serological and hematological laboratory tests, pathological examination, and mNGS analysis, followed by a comparative analysis of the results. RESULTS In our cohort of 301 cases of clinically diagnosed pyogenic spinal infections, 242 yielded etiological evidence. The most common gram-positive bacterium was Staphylococcus aureus, and the most common gram-negative bacterium was Escherichia coli. mNGS showed a significantly higher rate of detection (77.9%) compared with microbial culture (27.2%) with a notable difference (X² = 140.17, P<.001). In culture-negative samples, mNGS could detect pathogens in 73.1% of cases, and in culture-positive samples, it could detect pathogens in 91.5% of cases with 94.7% genus-level concordance. mNGS provided faster results (24-48 h) compared with the culture method (2-7 days). CONCLUSIONS mNGS serves as a valuable supplement to the culture method and shows potential in identifying the causative pathogen in native pyogenic spinal infections.
Collapse
Affiliation(s)
- Zhaohui Li
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, Shandong, China.
| | - Qiang Zhang
- Orthopedics Section II (Bone Infection), Public Health Clinical Center Affiliated to Shandong University, Shangdong, China
| | - Xiaofeng Lian
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuqiang Yin
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, Shandong, China
| | - Yuhan Lin
- Department of Spinal Surgery, Fuzhou Second General Hospital, Fujian, China
| | - Yuelei Wang
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, Shandong, China
| | - Zengshuai Han
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, Shandong, China
| | - Feng Shen
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, Shandong, China
| | - Yidan Xu
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, Shandong, China
| | - Huafeng Wang
- Department of Spinal Surgery, Fuzhou Second General Hospital, Fujian, China.
| | - Ting Wang
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, Shandong, China.
| |
Collapse
|
9
|
Wang Y, Li Z, Ji L, Sun J, Gao F, Yu R, Li K, Wang W, Zhao W, Zhong QZ, Ge S, Li J. Adhesive micro-liquid for efficient removal of bacterial biofilm infection. Mater Today Bio 2025; 31:101525. [PMID: 39958232 PMCID: PMC11830298 DOI: 10.1016/j.mtbio.2025.101525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/21/2025] [Accepted: 01/25/2025] [Indexed: 02/18/2025] Open
Abstract
Bacteria are common infectious pathogens that can cause invasive and potentially life-threatening infections. Ionic liquids have emerged as a novel class of alternatives to antibiotics, however their inherent hydrophobicity and immiscible in water exhibits poor adhesion to bacteria and diminishes its utilization and bioavailability for infection control. Herein, an adhesive metal phenolic encapsulated ionic liquid choline and geranate (CAGE@MPN) microcapsules is designed to address the aforementioned challenges and remove bacterial biofilm infections. The CAGE@MPN microcapsules are prepared through self-assembly of quercetin and ferrous ions on the interface of CAGE and water via metal-phenolic coordination. The MPN interface can stabilize the micro liquid and effectively adhere to bacterial surfaces. The microcapsules can disrupt bacterial cell walls to facilitate the release of cellular contents and destruct the biofilm, thereby exerting a pronounced bactericidal effect. The in vivo bactericidal effect of CAGE@MPN microcapsules is demonstrated in a murine model of Staphylococcus aureus (S. aureus) skin infection. The proposed adhesive micro-liquid system offers a promising strategy for noninvasive and efficient removal of bacterial biofilm infection.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Zhibang Li
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Lingli Ji
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Jiao Sun
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Fei Gao
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Ruiqing Yu
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Kai Li
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Wenjun Wang
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Weiwei Zhao
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Qi-Zhi Zhong
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Shaohua Ge
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Jianhua Li
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| |
Collapse
|
10
|
Wang Z, Li M, Li W, He L, Wang L, Cai K, Zhao X, Chen Y, Li D. Hybrid Outer Membrane Vesicles with Genetically Engineering for Treatment of Implant-Associated Infections and Relapse Prevention Through Host Immunomodulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415379. [PMID: 39950746 PMCID: PMC11984893 DOI: 10.1002/advs.202415379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/01/2025] [Indexed: 04/12/2025]
Abstract
Implant-associated infections (IAIs) are refractory to elimination, and the local immunosuppressive microenvironment (IME) exacerbates therapeutic difficulties, ultimately causing persistence and relapse. Therefore, exploring immunostrengthening treatments holds great promise for reversing IME and thoroughly eradicating chronic or repetitive infections. Bacterial outer membrane vesicles (OMVs) have emerged as potential immunostimulatory candidates; however, they lack active targeting capabilities and cause non-specific inflammatory side effects. In this study, bone marrow-derived mesenchymal stem cells (BMSCs) are genetically engineered to overexpress CXCR4 and isolated cell membranes (mBMSCCXCR4) for hybridization with OMVs derived from Escherichia coli (E. coli) to produce nanovesicles (mBMSCCXCR4@OMV). The resulting mBMSCCXCR4@OMV nanovesicles demonstrate excellent bone marrow targeting capability and are effectively taken up by bone marrow-derived macrophages, triggering the efficient transition to pro-inflammatory M1 status through TLR/NF-κB pathway. This alteration promotes innate bactericidal capacity and antigen presentation. Subsequent activation of T and B cells and inhibition of myeloid-derived suppressor cells (MDSCs) facilitated in vivo adaptive immunity in mouse models. Additionally, mBMSCCXCR4@OMV boosted memory B cell and bacteria-specific antibody responses. Together, these data highlight the potential of mBMSCCXCR4@OMV to eradicate complicated IAIs and provide whole-stage protection against postsurgical relapse, thus marking a significant immunotherapeutic advancement in the post-antibiotic era.
Collapse
Affiliation(s)
- Zhichao Wang
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Mingfei Li
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Medical 3D Printing CenterThe First Affiliated Hospital of Zhengzhou UniversityHenan Institute of Advanced Technology of Zhengzhou UniversityZhengzhou450052China
| | - Wenshuai Li
- Medical 3D Printing CenterThe First Affiliated Hospital of Zhengzhou UniversityHenan Institute of Advanced Technology of Zhengzhou UniversityZhengzhou450052China
| | - Liuliang He
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Long Wang
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Kehan Cai
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190China
| | - Yazhou Chen
- Medical 3D Printing CenterThe First Affiliated Hospital of Zhengzhou UniversityHenan Institute of Advanced Technology of Zhengzhou UniversityZhengzhou450052China
| | - Daifeng Li
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| |
Collapse
|
11
|
Zhang H, Qiao W, Liu Y, Yao X, Zhai Y, Du L. Addressing the challenges of infectious bone defects: a review of recent advances in bifunctional biomaterials. J Nanobiotechnology 2025; 23:257. [PMID: 40158189 PMCID: PMC11954225 DOI: 10.1186/s12951-025-03295-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025] Open
Abstract
Infectious bone defects present a substantial clinical challenge due to the complex interplay between infection control and bone regeneration. These defects often result from trauma, autoimmune diseases, infections, or tumors, requiring a nuanced approach that simultaneously addresses infection and promotes tissue repair. Recent advances in tissue engineering and materials science, particularly in nanomaterials and nano-drug formulations, have led to the development of bifunctional biomaterials with combined osteogenic and antibacterial properties. These materials offer an alternative to traditional bone grafts, minimizing complications such as multiple surgeries, high antibiotic dosages, and lengthy recovery periods. This review examines the repair mechanisms in the infectious microenvironment and highlights various bifunctional biomaterials that foster both anti-infective and osteogenic processes. Emerging design strategies are also discussed to provide a forward-looking perspective on treating infectious bone defects with clinically significant outcomes.
Collapse
Affiliation(s)
- Huaiyuan Zhang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Wenyu Qiao
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yu Liu
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Xizhou Yao
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yonghua Zhai
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Longhai Du
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
12
|
Inverardi N, Serafim MF, Sekar A, Fujino K, Ferreira M, Marzouca A, Nagler E, Muratoglu OK, Oral E. Wear-resistant antibacterial UHMWPE-based implant materials obtained by radiation crosslinking. Biomater Sci 2025. [PMID: 40131351 DOI: 10.1039/d4bm01663g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
The crosslinking of ultrahigh molecular weight polyethylenes (UHMWPEs) by irradiation has been employed for decades to enhance the wear resistance of these materials when used as a load-bearing implant component for joint arthroplasty. This surgical procedure can restore the mobility of patients affected by severe arthritis by the implantation of an artificial joint made of an articulating pair and a bearing component. While the surgery is usually successful, one of the most severe complications is peri-prosthetic joint infection (PJI), which can be extremely difficult to treat and eradicate. The use of UHMWPEs as a platform for the local delivery of antibiotics in addition to their structural function could be extremely beneficial for the improvement in the outcome of PJIs. In this study, we investigated whether irradiation can be used to sterilize and crosslink antibiotic-loaded UHMWPEs, and its effect on the drug eluting and antibacterial properties of these materials. We found that the antibiotics gentamicin sulfate and vancomycin hydrochloride were stable in irradiated UHMWPEs and did not hinder crosslinking of the UHMWPE matrix. Effective crosslinking led to optimal wear resistance, which was comparable to that of clinically available UHMWPEs. Sustained drug release was observed for an extended duration (up to six months) and both the drug eluents and eluted material surfaces showed antibacterial activity against Staphylococcus aureus, the most common causative bacterium for PJIs.
Collapse
Affiliation(s)
- Nicoletta Inverardi
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
- Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Maria F Serafim
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | - Amita Sekar
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
- Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Keita Fujino
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | - Matheus Ferreira
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | - Anthony Marzouca
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | - Emma Nagler
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | - Orhun K Muratoglu
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
- Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Ebru Oral
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
- Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
13
|
Xu W, Lin Z, Cortez-Jugo C, Qiao GG, Caruso F. Antimicrobial Phenolic Materials: From Assembly to Function. Angew Chem Int Ed Engl 2025; 64:e202423654. [PMID: 39905990 DOI: 10.1002/anie.202423654] [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: 12/04/2024] [Indexed: 02/06/2025]
Abstract
Infectious diseases pose considerable challenges to public health, particularly with the rise of multidrug-resistant pathogens that globally cause high mortality rates. These pathogens can persist on surfaces and spread in public and healthcare settings. Advances have been made in developing antimicrobial materials to reduce the transmission of pathogens, including materials composed of naturally sourced polyphenols and their derivatives, which exhibit antimicrobial potency, broad-spectrum activity, and a lower likelihood of promoting resistance. This review provides an overview of recent advances in the fabrication of antimicrobial phenolic biomaterials, where natural phenolic compounds act as active antimicrobial agents or encapsulate other antimicrobial agents (e.g., metal ions, antimicrobial peptides, natural biopolymers). Various forms of phenolic biomaterials synthesized through these two strategies, including antimicrobial particles, capsules, hydrogels, and coatings, are summarized, with a focus on their application in wound healing, bone repair and regeneration, oral health, and antimicrobial coatings for medical devices. The potential of these advanced phenolic biomaterials provides a promising therapeutic approach for combating antimicrobial-resistant infections and reducing microbial transmission.
Collapse
Affiliation(s)
- Wanjun Xu
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| | - Zhixing Lin
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| | - Christina Cortez-Jugo
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| | - Greg G Qiao
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne Parkville, Victoria, 3010, Australia
| |
Collapse
|
14
|
Zhou X, Xie J, Zhou X, Ma T, Lu Y, Yang Y, Xie Z, Ling H, Xu R, Wu M, Wang J, Wang W, Kong D, Xu P, Wan X, Wu H, Tong P, Xia H. Single-atom Zr doped heterojunction enhanced piezocatalysis for implant infection therapy through synergistic metal immunotherapy with sonodynamic and physical puncture. J Nanobiotechnology 2025; 23:243. [PMID: 40128749 PMCID: PMC11931772 DOI: 10.1186/s12951-025-03309-x] [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: 09/26/2024] [Accepted: 03/08/2025] [Indexed: 03/26/2025] Open
Abstract
Recent common clinical treatments for implant bacterial infections involve replacing inert implants and using antibiotics. However, these methods remain limited in their effectiveness for pathogen clearance, immune regulation, and osteogenesis. In this study, we developed a Zr-doped heterointerface of SrTiO3 and Hap (SrTiZrO3/Hap) heterojunction coating with single-atom Zr doping and heterogeneous interfaces designed for ultrasound-responsive antimicrobial activity and bone formation. Under ultrasound, the mechanical force exerted by SrTiZrO3/Hap enhances its physical puncture and sonodynamic activity, synergizing with the metalloimmunotherapy effect of Zr4+ for efficient antimicrobial activity. The primary mechanism enhancing sonodynamic activity involves local interfacial polarization from single-atom Zr doping, achieving piezoelectric catalysis in conjunction with electronic polarization from the built-in electric field. SrTiZrO3/Hap achieved a 99.3% antibacterial rate against S. aureus and 99.7% against E. coli under ultrasound. Additionally, SrTiZrO3/Hap promoted osteogenic differentiation after ultrasound irradiation by activating the PI3K/Akt pathway via its piezoelectric, needle-like topological surface and the release of functional ions, thus accelerating bone repair.
Collapse
Affiliation(s)
- Xing Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Shangcheng District, Hangzhou, 310006, China
| | - Jingbo Xie
- Department of Orthopaedics, The People's Hospital of Fengcheng City, Fengcheng, Jiangxi, China
- The Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Xingchen Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Tianyou Ma
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yichen Lu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yiwen Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhefei Xie
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Houfu Ling
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Rui Xu
- Department of Orthopaedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Mo Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jinglei Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Weixiang Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Derong Kong
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, P.R. China
| | - Pengchao Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xuan Wan
- Department of Orthopaedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- University Medicine Rostock, University of Rostock, Parkstr. 6, 18057, Rostock, Germany
| | - Hongbo Wu
- Department of Rehabilitation Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510700, China.
| | - Peijian Tong
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Hanting Xia
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- Department of Orthopaedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China.
| |
Collapse
|
15
|
Zegre M, Barros J, David AB, Fialho L, Ferraz MP, Monteiro FJ, Caetano LA, Gonçalves L, Bettencourt A. Dual-Loaded Chitosan-Based Nanoparticles: A Novel approach for treating polymicrobial osteomyelitis. Int J Pharm 2025; 674:125480. [PMID: 40097053 DOI: 10.1016/j.ijpharm.2025.125480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
Developing innovative approaches to target osteomyelitis caused by polymicrobial infections remains a significant therapeutic challenge. In this study, monodispersed chitosan nanoparticles co-loaded with antibacterial (minocycline) and antifungal (voriconazole) agents were successfully prepared. Minocycline presented higher encapsulation efficiency as compared to voriconazole. Thermostability analysis suggested interactions between the co-loaded drugs within the dual-delivery system, potentially limiting voriconazole release. The dual-loaded chitosan nanoparticles exhibited significant in vitro anti-biofilm activity, achieving up to a 90% reduction in polymicrobial biofilms of S. aureus and C. albicans. Additionally, the nanoparticles showed cytocompatibility with a human osteoblast cell line. These findings highlight the potential of this dual-delivery chitosan-based nanoparticle system to address a critical gap in osteomyelitis treatment by targeting both bacterial and fungal pathogens.
Collapse
Affiliation(s)
- M Zegre
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; H&TRC - Centro de Investigação em Saúde e Tecnologia, ESTeSL - Escola Superior de Tecnologia da Saúde de Lisboa, IPL - Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal
| | - J Barros
- i3S - Instituto de Investigação e Inovação em Saúde - Associação, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
| | - A B David
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - L Fialho
- i3S - Instituto de Investigação e Inovação em Saúde - Associação, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
| | - M P Ferraz
- i3S - Instituto de Investigação e Inovação em Saúde - Associação, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; Departamento de Engenharia Mecânica, Faculdade de Engenharia, Universidade do Porto, s/n, R. Dr. Roberto Frias 4200-465, Portugal
| | - F J Monteiro
- i3S - Instituto de Investigação e Inovação em Saúde - Associação, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; Departamento de Engenharia Mecânica, Faculdade de Engenharia, Universidade do Porto, s/n, R. Dr. Roberto Frias 4200-465, Portugal
| | - L A Caetano
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; H&TRC - Centro de Investigação em Saúde e Tecnologia, ESTeSL - Escola Superior de Tecnologia da Saúde de Lisboa, IPL - Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal
| | - L Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - A Bettencourt
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
16
|
Zuo J, Kong F, Wang X, Wang T, Zhao J, Zhao Z. Antimicrobial photodynamic therapy with 5-aminolevulinic acid plus antibiotics: a promising treatment for tibial osteomyelitis caused by drug-resistant bacteria. Front Pharmacol 2025; 16:1566744. [PMID: 40129941 PMCID: PMC11931076 DOI: 10.3389/fphar.2025.1566744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Osteomyelitis is a severely destructive bone disease caused by microbial infections, and currently, no available treatment effectively controls the infection. 5-Aminolevulinic acid is a second-generation endogenous photosensitizer. This study investigated the efficacy of 5-aminolevulinic acid-mediated photodynamic therapy (ALA-PDT) in combination with antibiotics in the treatment of tibial osteomyelitis in rabbits. The results illustrated that ALA-PDT alone and in combination of antibiotics displayed significant efficacy in treating osteomyelitis. Animals in the photodynamic antimicrobial chemotherapy (PACT) + antibiotics group exhibited a higher survival rate, an improved overall mental status, a lower localized infection rate, and reduced Tang Hui and Norden scores (P < 0.05), indicating less severe bone destruction. Histologically, more strips of lamellar new bone formation and more pronounced periosteal hyperplasia were noted in the PACT + antibiotics group. Micro-computed tomography illustrated that the structural integrity of cortical bone and cancellous bone structure had better continuity and clearer display in the PACT + antibiotics group than in the other groups, and the periosteal reaction in the modeling area was the most obvious. Bone parameter analysis indicated that trabecular thickness, bone volume, and trabeculae volume were significantly higher in the PACT + antibiotics group than in the model and antibiotics groups (P < 0.05). Additionally, trabecular separation was significantly lower in the PACT + antibiotic group than in the other groups (P < 0.05). These findings suggest that the combination of ALA-PDT and antibiotics has a sensitizing therapeutic effect, offering a promising strategy for the clinical treatment of osteomyelitis.
Collapse
Affiliation(s)
- Ju Zuo
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
- School of Clinical Medicine, Hebei University, Baoding, China
| | - Feiyan Kong
- School of Basic Medicine, Hebei University, Baoding, China
| | - Xiyu Wang
- School of Clinical Medicine, Hebei University, Baoding, China
| | - Tianyu Wang
- School of Basic Medicine, Hebei University, Baoding, China
| | - Jianxi Zhao
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Zhanjuan Zhao
- School of Basic Medicine, Hebei University, Baoding, China
| |
Collapse
|
17
|
Nguyen Ngoc D, Latalski M, Danielewicz A, Szponder T, Wessely-Szponder J, Mazur E. Application of Antimicrobial Peptides (AMPs) in Treatment of Osteomyelitis in Human and Veterinary Orthopedics. J Funct Biomater 2025; 16:90. [PMID: 40137369 PMCID: PMC11943317 DOI: 10.3390/jfb16030090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/24/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025] Open
Abstract
Osteomyelitis, a severe bone infection, poses a significant therapeutic challenge in both human and veterinary medicine, especially due to the increasing prevalence of antibiotic-resistant pathogens like methicillin-resistant Staphylococcus aureus (MRSA). Conventional treatments, including surgical debridement and systemic antibiotics, often prove inadequate due to the ability of bacteria to form biofilms and evade host immune responses. Antimicrobial peptides (AMPs), such as LL-37 and β-defensins, have emerged as a promising alternative therapeutic strategy. AMPs exhibit broad-spectrum antimicrobial activity, including efficacy against resistant strains, and possess immunomodulatory properties that can promote bone regeneration. This article comprehensively reviews AMP applications in treating osteomyelitis across both human and veterinary medicine. We discuss diverse therapeutic approaches, including free AMPs, their conjugation with biomaterials such as collagen and chitosan to enhance delivery and stability, and the development of AMP-based nanoparticles. Furthermore, we analyze preclinical and clinical findings, highlighting the efficacy and safety of AMPs in combating osteomyelitis in both human and animal patients. Finally, we explore future perspectives and challenges, such as optimizing delivery, stability, and efficacy, while minimizing cytotoxicity, and in translating AMP-based therapies into clinical practice to effectively manage this debilitating disease.
Collapse
Affiliation(s)
- Dominika Nguyen Ngoc
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland; (D.N.N.); (E.M.)
| | - Michał Latalski
- Children’s Orthopaedic Department, Medical University of Lublin, 20-093 Lublin, Poland; (M.L.)
| | - Anna Danielewicz
- Children’s Orthopaedic Department, Medical University of Lublin, 20-093 Lublin, Poland; (M.L.)
| | - Tomasz Szponder
- Department of Diagnostics and Clinical Sciences, Faculty of Veterinary Medicine, University of Agriculture in Krakow, 31-120 Cracow, Poland
| | - Joanna Wessely-Szponder
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland; (D.N.N.); (E.M.)
| | - Ewa Mazur
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland; (D.N.N.); (E.M.)
| |
Collapse
|
18
|
Fornari V, Accardo G, Lupia T, De Rosa FG, Corcione S. Suppressive antibiotic treatment (SAT) in the era of MDRO infections: a narrative review. Expert Rev Anti Infect Ther 2025:1-13. [PMID: 40016121 DOI: 10.1080/14787210.2025.2473077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/02/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
INTRODUCTION Antibiotics were originally developed to treat acute bacterial infections, and research studies focus their efforts on safety and efficacy in the short term; however, prolonged course of antibiotics has been documented in multiple clinical settings. The aim of this narrative review is to provide a new perspective on SAT and to discuss new therapeuticpossibilities. AREAS COVERED We discuss new clinical scenarios in which SAT could be considered. We provided a broad discussion about long-acting agents and new or repurposed oral agents as well as the use of OPAT with elastomeric pumps and an overview of the pipeline of new antifungals. Limitations of SAT are presented in this review and especially patients' adherence issues, possible spread of MDROs, possible rising of the incidence of Clostridioides difficile infections, drug-to-drug interactions and drug-related problems, cost-effectiveness evaluation issues. EXPERT OPINION Many research gaps are evident and further studies are needed. Above all, the efficacy and safety of SAT in the different clinical scenarios. Discovery of new molecules against MDROs and ongoing research on PK/PD variables as well as a better understanding of the relationship between SAT and the emergence of resistance could improve SAT usage and reduce the impact of DRPs.
Collapse
Affiliation(s)
- Valentina Fornari
- Department of Medical Sciences, Infectious Diseases, University of Turin, Turin, Italy
| | - Guido Accardo
- Department of Medical Sciences, Infectious Diseases, University of Turin, Turin, Italy
| | - Tommaso Lupia
- Infectious Diseases Unit, A.O.U. Città della Salute e della Scienza di Torino, Presidio Molinette, Turin, Italy
| | | | - Silvia Corcione
- Department of Medical Sciences, Infectious Diseases, University of Turin, Turin, Italy
- Division of Geographic Medicine and Infectious Diseases, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
19
|
Wang Z, Chu Y, Du J, Hu Y, Wang H, Liu H, Yang C, Wang M, Ran J, Yu A. Accelerating repair of infected bone defects through post-reinforced injectable hydrogel mediated antibacterial/immunoregulatory microenvironment at bone-hydrogel interface. Carbohydr Polym 2025; 351:123082. [PMID: 39779005 DOI: 10.1016/j.carbpol.2024.123082] [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: 09/14/2024] [Revised: 11/04/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025]
Abstract
Functional injectable hydrogel (IH) is promising for infected bone defects (IBDs) repair, but how to endow it with desired antibacterial/immunoregulatory functions as well as avoid mechanical failures during its manipulation has posed as main challenges. Herein, rosmarinic acid (RosA), a natural product with antibacterial/immunoregulatory activities, was utilized to develop a FCR IH through forming phenylboronic acid ester bonds with 4-formylphenyl phenylboronic acid (4-FPBA) grafted chitosan (CS) (FC). After being applied to the IBD site, the FCR IH was then injected with tobramycin (Tob) solution, another alkaline antibacterial drug, to induce in situ crystallization of the FC, endowing the resultant FCRT hydrogel with adaptively enhanced mechanical strength and structural stability. Owing to the specific structural composition, the FCRT hydrogel could sustainedly release Tob and RosA molecules at the IBD interface, effectively eliminating in situ bacterial infection. In addition, the released RosA molecules also induced the M2 polarization of in situ macrophages (Mφ), which was identified to be related to the NF-κB and PI3K-AKT pathways, therefore promoting the osteogenic differentiation of in situ bone marrow stromal cells (BMSCs). Due to the simultaneous antibacterial/osteo-immunoregulatory microenvironment at the IBD interface, the repair of IBDs was proved to be greatly accelerated by the FCRT hydrogel.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan 430000, China
| | - Ying Chu
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Jingyi Du
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yan Hu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan 430000, China
| | - Huimin Wang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Hanghang Liu
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Changying Yang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Man Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Jiabing Ran
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Medicine & Engineering & Informatics Fusion and Transformation Key Laboratory of LuZhou City, Luzhou, China. 646000.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China; Hubei Clinical Medical Research Center of Trauma and Microsurgery, Wuhan 430000, China.
| |
Collapse
|
20
|
Yu Z, Wang M, Li J, Xu H, Zhang W, Xing F, Li J, Yang J, Xiong Y. A Fused Membrane-Camouflaged Biomimetic Nanosystem for Dual-Targeted Therapy of Septic Arthritis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410710. [PMID: 39828630 DOI: 10.1002/smll.202410710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/31/2024] [Indexed: 01/22/2025]
Abstract
Due to the inherent aseptic and enclosed characteristics of joint cavity, septic arthritis (SA) almost inevitably leads to intractable infections and rapidly progressing complex pathological environments. Presently, SA faces not only the deficient effectiveness of the gold-standard systemic antibiotic therapy but also the scarcity of effective localized targeted approaches and standardized animal models. Herein, an ingenious multifunctional nanosystem is designed, which involves the methylation of hyaluronic acid (HA), copolymerization with DEGDA, loading with vancomycin (VAN), and then coating with fused macrophage-platelet membrane (denoted as FM@HA@VAN). Upon intra-articular administration, FM@HA@VAN nanoparticles exhibit sustained retention and selectively targeting to infected sites, leveraging macrophage-mediated inflammation homing and platelet-directed bacteria targeting. The acidic microenvironment triggers responsive release of vancomycin, leading to potent bactericidal effects. Subsequently, the exposed HA@VAN nanoparticles are efficiently internalized by activated macrophages, releasing HA to alleviate oxidative stress and achieve chondroprotection by inhibiting pro-inflammatory cytokines, neutralizing ROS and upregulating macrophage M2 polarization. In vivo model and experiments confirm the efficacy of this dual-targeting antibacterial approach, demonstrating its precision in eradicating bacterial infections and alleviating associated pathological processes, including synovial hyperplasia and cartilage erosion. The dual-targeting therapeutic nanosystem, coordinated with fused-membranes, holds promise for enhancing the treatment efficacy of SA.
Collapse
Affiliation(s)
- Zeping Yu
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengxian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junqiao Li
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Xu
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenli Zhang
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fei Xing
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jian Li
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiaojiao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yan Xiong
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
21
|
Shang S, Zheng F, Tan W, Xing Z, Chen S, Peng F, Lv X, Wang D, Zhu X, Wu J, Zhou Z, Zhang X, Yang X. Piezoelectric Biomaterial with Advanced Design for Tissue Infection Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413105. [PMID: 39887897 PMCID: PMC11905007 DOI: 10.1002/advs.202413105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/25/2024] [Indexed: 02/01/2025]
Abstract
Bacterial infection has become the most dangerous factor in tissue repair, which strongly affects the tissue regeneration efficiency and wellness of patients. Piezoelectric materials exhibit the outstanding advantage of producing electrons without external power supply. The ability of electron enrichment and reactive oxygen species generation through noninvasive stimulations enables piezoelectric materials the potential applications of antibacterial. Many studies have proved the feasibility of piezoelectric materials as a functional addition in antibacterial biomaterial. In fact, numerous piezoelectric materials with ingenious designs are reported to be effective in antibacterial processes. This review summarizes the antibacterial mechanisms of piezoelectric, illuminating their potential in combating bacteria. Recent advancement in the design and construction of piezoelectric biomaterial including defect engineering, heterojunction, synergy with metal and the composite scaffold configuration are thoroughly reviewed. Moreover, the applications and therapeutic effects of piezoelectric materials in common tissues with antibacterial requirements are introduced, such as orthopedics, dental, and wound healing. Finally, the development prospects and points deserving further exploration are listed. This review is expected to provide valuable insight into the relationship between antibacterial processes and piezoelectric materials, further inspiring constructive development in this emerging scientific discipline.
Collapse
Affiliation(s)
- Siyuan Shang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Fuyuan Zheng
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, 610065, China
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wen Tan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Zhengyi Xing
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Siyu Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fuli Peng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Xiang Lv
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Duan Wang
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, 610065, China
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Jiagang Wu
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065, China
- College of Physics, Sichuan University, Chengdu, 610065, China
| | - Zongke Zhou
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
22
|
Liu Y, Yuan K, Lin Y, Yang Y, Kong W, Shan J, Niu H, Kong W, Li F, Yue X, Du Y, Liang Y, Chang H, Yu Z, Wang J, Yang G, Cao L, Huang K, Yang S, Tang T. Directional Freeze-Casting Cryogel Loaded with Quaternized Chitosan Modified Gallium Metal-Organic Frameworks to Capture and Eradicate the Resistant Bacteria for Guided Regeneration in Infected Bone Defects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414437. [PMID: 39846310 DOI: 10.1002/adma.202414437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/16/2024] [Indexed: 01/24/2025]
Abstract
Antimicrobial resistance and impaired bone regeneration are the great challenges in treating infected bone defects. Its recurrent and resistant nature, high incidence rate, long-term hospitalization, and high medical costs have driven the efforts of the scientific community to develop new therapies to improve the situation. Considering the complex microenvironment and persistent mechanisms mediated by resistant bacteria, it is crucial to develop an implant with enhanced osseointegration and sustained and effective infection clearance effects. Here, a positively charged quaternized chitosan (QCS) coated gallium-based metal-organic framework (GaMOF) is designed, to capture the antibiotic-resistant bacteria (Methicillin-resistant Staphylococcus aureus, MRSA) as a "captor", and rejuvenate Methicillin (Me) via disturbing the tricarboxylic acid (TCA) cycle. Then, a radially oriented porous cryogel loaded with the Me and QCSGaMOF is fabricated by the directional freeze-casting method. The oriented porous structure has an enhanced osseointegration effect by guiding the ingrowth of osteogenic cells. In vitro and in vivo experiments prove the advantages of as-prepared Me/QCSGa-MOF@Cryogel in combating resistant bacteria and guiding bone regeneration in infected bone defects.
Collapse
Affiliation(s)
- Yihao Liu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Kai Yuan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China
| | - Yixuan Lin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Yiqi Yang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd, Hangzhou, 310003, P. R. China
| | - Weiqing Kong
- Department of Orthopedic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, 199Jiefang South Road, Xuzhou, 221009, P. R. China
| | - Jing Shan
- School of Pharmacy, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Haoyi Niu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Weize Kong
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Fupeng Li
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Xiaokun Yue
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Yun Du
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Yakun Liang
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, P. R. China
| | - Haishuang Chang
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, P. R. China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Guangzheng Yang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Lingyan Cao
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Kai Huang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| |
Collapse
|
23
|
Yang G, Pan H, Wei Y, Yang J, Zhang Z, Chen S, Wan W. Directional Mushroom-Derived Scaffold for Microenvironment Regulation in Infected Bone Defects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407730. [PMID: 39961061 DOI: 10.1002/adma.202407730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/11/2024] [Indexed: 03/27/2025]
Abstract
Infected bone defects are a common clinical condition, but conventional treatments often fail to achieve the desired outcomes, including addressing antibiotic resistance and preventing nonunion complications. In the presented study, a functionalized decellularized mushroom stem scaffold is developed composed of its naturally aligned channels, Zn2+/curcumin MOFs, hydroxyapatite minerals, and icariin. In vitro, It is found that functionalized acellular mushroom stem scaffold can control bacterial infections through Zn2+/curcumin MOFs. The naturally aligned channels guide bone mesenchymal stem cells (BMSCs) migration, and the components adsorbed on the acellular substrate further promote the migration of BMSCs. Moreover, these functional components further accelerated the polarization of M2 macrophage and osteogenic differentiation of BMSCs. In vivo, the functionalized decellularized mushroom stem scaffold cleared infected bacteria within 3 days, induced extracellular matrix secretion and alignment, and promoted new bone formation to cover defects within 8 weeks. The functionalized decellularized mushroom stem scaffold provides a promising strategy for treating infectious bone defects.
Collapse
Affiliation(s)
- Ganghua Yang
- Department of Orthopaedic Surgery, Institute of Orthopedics of Jiangxi Province and Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Hao Pan
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Yuxuan Wei
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Jianqiu Yang
- Department of Orthopaedic Surgery, Institute of Orthopedics of Jiangxi Province and Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zihan Zhang
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Shixuan Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Wenbing Wan
- Department of Orthopaedic Surgery, Institute of Orthopedics of Jiangxi Province and Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
24
|
Kimura H, Aiba H, Saito S, Sakai T, Murakami H. Long-term follow-up of chronic osteomyelitis after bone tumor resection. J Infect Chemother 2025; 31:102597. [PMID: 39710164 DOI: 10.1016/j.jiac.2024.102597] [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: 07/26/2023] [Revised: 08/20/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
The standard treatment for chronic osteomyelitis after trauma is affected bone resection and bone and soft tissue defect reconstruction. However, few reports exist regarding chronic osteomyelitis after bone tumor surgery. We retrospectively reviewed five cases of chronic infection after bone tumor surgery, including their treatment strategy and clinical course. We reviewed three cases of giant cell tumors of bone and two cases of osteosarcoma. Reconstruction was performed after tumor resection in all cases. Despite careful management, fistula formation and chronic infection occurred. Two patients underwent radical surgery for chronic infection. After the infection subsided, reconstruction was performed again. However, in one case, the infection recurred, and consequently, amputation was performed. When radical surgery is performed, implant replacement is essential due to biofilm formation. Controlling soft tissue infection, besides bone and implant infection, is important. In some cases, however, radical surgery is undesirable, and patients choose to live with the chronic infection instead. Even when methicillin-resistant S. aureus (MRSA) was detected, anti-MRSA drugs were used only in the early stages, after which the infection was managed by switching to oral antibiotics, such as minocycline and sulfamethoxazole-trimethoprim combination drugs. Careful follow-up is necessary due to the risk of fistula cancer in conservative management.
Collapse
Affiliation(s)
- Hiroaki Kimura
- Department of Orthopedic Surgery, Nagoya City University, Nagoya, Japan.
| | - Hisaki Aiba
- Department of Orthopedic Surgery, Nagoya City University, Nagoya, Japan
| | - Shiro Saito
- Department of Orthopedic Surgery, Nagoya City University, Nagoya, Japan
| | - Takao Sakai
- Department of Orthopedic Surgery, Nagoya City University, Nagoya, Japan
| | - Hideki Murakami
- Department of Orthopedic Surgery, Nagoya City University, Nagoya, Japan
| |
Collapse
|
25
|
Wang Y, Wang L, Guo D, Liu X, Xu Y, Wang R, Sun Y, Liu Q, Guan J, Liu D, Wang B, Zhao Y, Yan M. Targeting ClpP: Unlocking a novel therapeutic approach of isochlorogenic acid A for methicillin-resistant Staphylococcus aureus-infected osteomyelitis. Microbiol Res 2025; 292:128042. [PMID: 39756139 DOI: 10.1016/j.micres.2024.128042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
A medical predicament has led to extensive drug resistance in methicillin-resistant Staphylococcus aureus (MRSA), and the complexity of treatment has increased exponentially with the induction of osteomyelitis. In view of the severe situation and the potential of bacterial antivirulence strategies, this study focused on the key virulence factor caseinolytic protease (ClpP) of S. aureus to identify new strategies against MRSA-induced osteomyelitis. As the main protein "quality control" system of S. aureus, ClpP is indispensable for coordinating drug resistance, regulating adhesion, and acting on numerous virulence targets. Through fluorescence resonance energy transfer (FRET), we successfully identified isochlorogenic acid A (I-A), a polyphenol derivative, as an efficient inhibitor of ClpP, with an IC50 value of 24.89 μg/mL. Further analysis revealed that I-A can effectively inhibit the expression of virulence factors of MRSA and significantly reduce its adhesion to fibrinogen. Molecular docking revealed the potential binding sites of ClpP and I-A, namely, ILE-81, LYS-109, GLU-156, ARG-157, and GLY-184. At the cellular level, I-A can alleviate the death and increased secretion of inflammatory factors caused by MRSA USA300 in MC3T3-E1 cells. Moreover, it downregulates the activity of ClpP and reduces the response of bacteria to environmental stress. In vivo experiments have confirmed that I-A shows significant efficacy in both rat osteomyelitis models and Galleria mellonella infection models. This study provides new insights into the field of treatment strategies targeting virulence and provides a solid foundation for further exploration of the potential of I-A in combating drug-resistant S. aureus.
Collapse
Affiliation(s)
- Yueying Wang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Li Wang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China; Clinical Medical College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Dongbin Guo
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xinyao Liu
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yueshan Xu
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Rong Wang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yun Sun
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Quan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun 130062, China
| | - Jiyu Guan
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun 130062, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Bingmei Wang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China; Clinical Medical College, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Yicheng Zhao
- Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong 519000, China; State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun 130062, China.
| | - Ming Yan
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China.
| |
Collapse
|
26
|
Scourfield LEA, Nardo-Marino A, Williams TN, Rees DC. Infections in sickle cell disease. Haematologica 2025; 110:546-561. [PMID: 39568431 PMCID: PMC11873697 DOI: 10.3324/haematol.2024.285066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024] Open
Abstract
Sickle cell disease (SCD) is one of the commonest severe inherited disorders in the world. Infection accounts for a significant amount of the morbidity and mortality, particularly in sub-Saharan Africa, but is relatively poorly studied and characterized. Patients with SCD have significant immunodeficiency and are more likely to suffer severe and life-threatening complications of infection, and additionally infections can trigger complications of SCD itself. Those with more severe forms of SCD have functional asplenia from a very early age, which accounts for much of the morbidity in young children, particularly invasive infections from encapsulated bacteria including Streptococcus pneumoniae, Haemophilus influenzae, Salmonella typhi and meningococcal disease. Additionally, there are other defects in immune function in SCD, associated with anemia, tissue infarction and impaired adaptive immunity. Complications of infections in SCD include acute chest syndrome, acute painful episodes, osteomyelitis, meningitis, urinary tract infections, overwhelming sepsis and death. Viral infections cause significant morbidity, particularly severe anemia associated with parvovirus, and to a lesser extent other infections such as influenza and coronavirus disease 2019. The relationship between malaria and SCD is complicated and discussed in this review. Unlike many of the genetic risk factors for poor outcomes in SCD, it is theoretically possible to modify the risks associated with infections with established public health measures. These include the provision of vaccination, prophylactic antibiotics and access to clean water and mosquito avoidance, although current financial restraints and political priorities have made this difficult.
Collapse
Affiliation(s)
- Lily E. A. Scourfield
- Department of Haematological Medicine, King’s College Hospital NHS Foundation Trust, London, UK
- Red Cell Haematology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| | | | - Thomas N. Williams
- Department of Epidemiology, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Institute for Global Health Innovation, Imperial College, London, UK
| | - David C. Rees
- Department of Haematological Medicine, King’s College Hospital NHS Foundation Trust, London, UK
- Red Cell Haematology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| |
Collapse
|
27
|
Han X, Wang W, Shen Z, Lv L, Lin B, Ren H, Liu Y, Guo Q, Kai H, Wang X. Comparative study of the inhibitory effects of different antibiotic administration routes on bone healing in a rat tibial infection model. Front Cell Infect Microbiol 2025; 15:1529692. [PMID: 40093534 PMCID: PMC11907719 DOI: 10.3389/fcimb.2025.1529692] [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: 11/17/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Objective This study aimed to evaluate the effectiveness of intravenous versus oral antibiotic treatments in managing bone infections, particularly osteomyelitis, using a rat tibial infection model. Methods A tibial bone infection model was established in twelve-week-old Wistar rats via injection of Staphylococcus aureus at a cortical defect site. After six weeks, rats were treated with vancomycin (intravenous), cefazolin (intravenous), ciprofloxacin (oral), or ciprofloxacin combined with rifampin (oral). Microbial analysis, blood analysis for pro-inflammatory cytokines, micro-computed tomography (μCT), histological analysis, and osteoclast activity were used to assess the efficacy of each treatment. Results Blood analysis showed significant reductions in white blood cell count and pro-inflammatory cytokines in the intravenous treatment groups, especially with vancomycin. μCT imaging revealed better preservation of bone structure in intravenous treatment groups, while oral treatments resulted in more pronounced structural deterioration. Microbial analysis confirmed a lower bacterial load in the intravenous groups, particularly vancomycin, compared to oral treatments. Histological analysis revealed reduced inflammation, lower fibrosis, and minimal bacterial presence in intravenous groups. Osteoclast activity was notably reduced in the vancomycin and cefazolin groups, indicating better control of bone resorption. Conclusion Intravenous administration of vancomycin demonstrated superior efficacy in controlling bone infection, reducing inflammation, and preserving bone structure compared to oral treatments. While ciprofloxacin and the ciprofloxacin-rifampin combination showed some efficacy, they were less effective than intravenous vancomycin, likely due to lower bioavailability and insufficient drug penetration in bone tissue.
Collapse
Affiliation(s)
- Xiaoyu Han
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Wei Wang
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zengli Shen
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Lisong Lv
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Bingyuan Lin
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Haiyong Ren
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yiyang Liu
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Qiaofeng Guo
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Huang Kai
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Xiang Wang
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
28
|
Liu X, Wang C, Wang H, Wang G, Zhang Y, Zhang Y. Calcium phosphate-based anti-infective bone cements: recent trends and future perspectives. Front Pharmacol 2025; 16:1522225. [PMID: 40078285 PMCID: PMC11897017 DOI: 10.3389/fphar.2025.1522225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Bone infection remains a challenging condition to fully eradicate due to its intricate nature. Traditional treatment strategies, involving long-term and high-dose systemic antibiotic administration, often encounter difficulties in achieving therapeutic drug concentrations locally and may lead to antibiotic resistance. Bone cement, serving as a local drug delivery matrix, has emerged as an effective anti-infective approach validated in clinical settings. Calcium phosphate cements (CPCs) have garnered widespread attention and application in the local management of bone infections due to their injectable properties, biocompatibility, and degradability. The interconnected porous structure of calcium phosphate particles, not only promotes osteoconductivity and osteoinductivity, but also serves as an ideal carrier for antibacterial agents. Various antimicrobial agents, including polymeric compounds, antibiotics, antimicrobial peptides, therapeutic inorganic ions (TIIs) (and their nanoparticles), graphene, and iodine, have been integrated into CPC matrices in numerous studies aimed at treating bone infections in diverse applications such as defect filling, preparation of metal implant surface coatings, and coating of implant surfaces. Additionally, for bone defects and nonunions resulting from chronic bone infections, the utilization of calcium phosphate-calcium sulfate composite multifunctional cement loaded with antibacterial agents serves to efficiently deal with infection, stimulate new bone formation, and attain an optimal degradation rate of the bone cement matrix. This review briefly delves into various antibacterial strategies based on calcium phosphate cement for the prevention and treatment of bone infections, while also discussing the application of calcium phosphate-calcium sulfate composites in the development of multifunctional bone cement against bone infections.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Orthopaedics, Second Affiliated Hospital, Air Force Medical University, Xi’an, China
| | - Chaoli Wang
- Department of Pharmacy, Air Force Medical University, Xi’an, China
| | - Han Wang
- Department of Orthopaedics, Second Affiliated Hospital, Air Force Medical University, Xi’an, China
| | - Guoliang Wang
- Department of Orthopaedics, Second Affiliated Hospital, Air Force Medical University, Xi’an, China
| | - Yong Zhang
- Department of Orthopaedics, Second Affiliated Hospital, Air Force Medical University, Xi’an, China
| | - Yunfei Zhang
- Department of Orthopaedics, Second Affiliated Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
29
|
Hu Q, Wu C, Wang L, Cao D, Wang J, Du Y, Liu M, Li K. Multifunctional metal-phenolic nanoparticles with antibacterial and anti-inflammatory effects for osteomyelitis management. J Mater Chem B 2025; 13:3067-3079. [PMID: 39899347 DOI: 10.1039/d4tb02649g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Osteomyelitis is a serious inflammatory disease mostly caused by bacterial infections. It is necessary to simultaneously eradicate bacterial cells and inhibit inflammation in treating osteomyelitis. Herein, we design an innovative zinc ion (Zn2+)-based nano delivery system for the management of osteomyelitis. Taking advantage of the coordination self-assembly of Zn2+, quercetin (QU), and ε-poly-L-lysine (EPL), Zn2+-containing nanoparticles (denoted as ZQE NPs) are prepared. ZQE NPs are spherical nanoparticles with amorphous structures. They are stable in the physiological neutral environment but can be dissociated in an acidic microenvironment of infection sites. Since Zn2+ is encapsulated into ZQE NPs by coordination interaction, the deactivation of Zn2+ by proteins can be effectively avoided. Therefore, ZQE NPs can maintain excellent bactericidal activity in a protein-rich environment, while dissociative Zn2+ doesn't exhibit obvious bactericidal ability. Meanwhile, ZQE NPs are highly effective at scavenging intracellular reactive oxygen species (ROS) and inhibiting pro-inflammatory cytokines, due to the strong anti-inflammatory effects of QU and Zn2+. The in vivo therapeutic efficacy of ZQE NPs is assessed using a rat model of methicillin-resistant Staphylococcus aureus (MRSA)-induced osteomyelitis. Results demonstrate that ZQE NPs effectively eradicate bacterial cells and reduce inflammation in vivo, thereby promoting osteogenesis and recovery of osteomyelitis.
Collapse
Affiliation(s)
- Qinsheng Hu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- Department of Orthopedic Surgery, Ya'an People's Hospital, Ya'an 625000, China
| | - Chengcheng Wu
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Ling Wang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Dan Cao
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Junchao Wang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Yangrui Du
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Miao Liu
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, P. R. China.
| | - Kaijun Li
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China.
| |
Collapse
|
30
|
Yang X, Fang R, Li X, Kong W, Jin Y, Jiao R, Liu Z, Zhang M, Peng Q, Zhang Y, Song N. Engineered Nanovesicles for the Precise and Noninvasive Treatment of Deep Osteomyelitis Caused by MRSA Infection with Enhanced Immune Response. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11795-11810. [PMID: 39945439 DOI: 10.1021/acsami.4c20893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The clinical treatment of hospital-acquired persistent osteomyelitis caused by methicillin-resistant Staphylococcus aureus (MRSA) presents two major challenges: ineffective drug delivery into deep tissues and counteracting the rapid establishment of an immunosuppressive microenvironment. Indeed, MRSA can evade immunosurveillance and undermine both innate and adaptive immune responses. Herein, the engineered nanovesicles, functioning by combining sonodynamic therapy (SDT) with immune modulation, were constructed for the precise and noninvasive removal of MRSA in deep tissue and activation of the antimicrobial immune response using a newly engineered nanovesicle. Macrophage-derived M1 phenotypic microvesicles (M1-MW) internalized vancomycin-cross-linked micelles with the acoustic sensitizer indocyanine green (ICG) (VCG micelles). The vesicles of M1-MW were grafted with PEGylated mannose, allowing for targeted accumulation at the infection site. The VCG micelles were responsive to the highly reducing environment and released ICG to generate ROS after exposure to ultrasounds. This effect was combined with the presence of vancomycin to kill MRSA. In an osteomyelitis infection model, we observed an improved survival rate and reprogramming of macrophages to a pro-inflammatory M1 phenotype. The latter promoted T-cell activation and immune defense against MRSA-camouflaged homologous cell-transferred infections. Thus, our study presents a noninvasive and efficient treatment (VCG@MMW) for deep osteomyelitis with improved bacterial clearance and reduced risk of recurrence with enhanced immune response.
Collapse
Affiliation(s)
- Xingyue Yang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ren Fang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Xiaotian Li
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Weihao Kong
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yubao Jin
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ruohan Jiao
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Zhenggong Liu
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Meiqi Zhang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Qixian Peng
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300350, P. R. China
| | - Ningning Song
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| |
Collapse
|
31
|
Wang S, Zhao T, Sun Y, Li S, Lu D, Qiu M, Yan B, Yang J, Shao Z, Yin Y, Li S, Fu T. In Situ Application of Berberine-Loaded Liposomes on the Treatment of Osteomyelitis. ACS OMEGA 2025; 10:7350-7361. [PMID: 40028119 PMCID: PMC11866016 DOI: 10.1021/acsomega.4c11198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/20/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025]
Abstract
Osteomyelitis is a major challenge in global healthcare, as it requires the simultaneous management of bone defects and bacterial infections, which poses considerable difficulties for orthopedic clinicians. In this study, we developed berberine liposome-modified bone cement specifically aimed at treating osteomyelitis induced by Staphylococcus aureus. We characterized the physical properties of this modified bone cement, conducted in vitro antibacterial assays to evaluate its efficacy in eradicating Staphylococcus aureus biofilm, established an in vivo rat model of osteomyelitis, and performed histopathological assessments alongside micro-CT analysis of bone parameters. The results indicated that the berberine liposome-modified bone cement exhibited favorable biodegradability and sustained-release characteristics, with a drug release rate of more than 90% within 14 days, while effectively eliminating bacterial biofilm with a biofilm eradication rate of up to 80% and facilitating bone repair with a bone volume fraction of 80%. This innovative treatment demonstrated both safety and efficacy in addressing tibial osteomyelitis in rats, thereby offering novel insights and methodologies for clinical interventions against osteomyelitis.
Collapse
Affiliation(s)
- Siting Wang
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tianlong Zhao
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuping Sun
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sipan Li
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Danya Lu
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengmeng Qiu
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Baofei Yan
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jingwen Yang
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhitao Shao
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuqi Yin
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shaoguang Li
- Microsurgery
Department of Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Tingming Fu
- State
Key Laboratory on Technologies for Chinese Medicine Pharmaceutical
Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
32
|
Rasquel-Oliveira FS, Ribeiro JM, Martelossi-Cebinelli G, Costa FB, Nakazato G, Casagrande R, Verri WA. Staphylococcus aureus in Inflammation and Pain: Update on Pathologic Mechanisms. Pathogens 2025; 14:185. [PMID: 40005560 PMCID: PMC11858194 DOI: 10.3390/pathogens14020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterium of significant clinical importance, known for its versatility and ability to cause a wide array of infections, such as osteoarticular, pulmonary, cardiovascular, device-related, and hospital-acquired infections. This review describes the most recent evidence of the pathogenic potential of S. aureus, which is commonly part of the human microbiota but can lead to severe infections. The prevalence of pathogenic S. aureus in hospital and community settings contributes to substantial morbidity and mortality, particularly in individuals with compromised immune systems. The immunopathogenesis of S. aureus infections involves intricate interactions with the host immune and non-immune cells, characterized by various virulence factors that facilitate adherence, invasion, and evasion of the host's defenses. This review highlights the complexity of S. aureus infections, ranging from mild to life-threatening conditions, and underscores the growing public health concern posed by multidrug-resistant strains, including methicillin-resistant S. aureus (MRSA). This article aims to provide an updated perspective on S. aureus-related infections, highlighting the main diseases linked to this pathogen, how the different cell types, virulence factors, and signaling molecules are involved in the immunopathogenesis, and the future perspectives to overcome the current challenges to treat the affected individuals.
Collapse
Affiliation(s)
- Fernanda S. Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Jhonatan Macedo Ribeiro
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Fernanda Barbosa Costa
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| |
Collapse
|
33
|
Lv H, Yang M, Yang Y, Tang Z, Guo Y, Zhou J, Gui Y, Huang R, Cai J, Yu B, Yang J, Bao Y, Zhang Z, Zhang D, Hou T. Metal Ion and Antibiotic Co-loaded Nanoparticles for Combating Methicillin-Rresistant Staphylococcus aureus-Induced Osteomyelitis. ACS NANO 2025; 19:5253-5268. [PMID: 39886847 DOI: 10.1021/acsnano.4c11956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) causes osteomyelitis (OM), which seriously threatens public health due to its antimicrobial resistance. To increase the sensitivity of antibiotics and eradicate intracellular bacteria, a Zn2+ and vancomycin (Van) codelivered nanotherapeutic (named Man-Zn2+/Van NPs) was fabricated and characterized via mannose (Man) modification. Man-Zn2+/Van NPs exhibit significant inhibitory activity against extra- and intracellular MRSA and obviously decrease the minimum inhibitory concentration of Van. Man-Zn2+/Van NPs can be easily internalized by MRSA-infected macrophages and significantly accumulated in infected bone via Man-mediated targeting. In vivo experiments in a mouse OM model verified that Man-Zn2+/Van NPs significantly reduce the extra- and intracellular MRSA burden, improve gait patterns, increase bone mass, and decrease inflammatory cytokine expression. The antibacterial mechanism of Man-Zn2+/Van NPs includes destruction of the MRSA membrane, degeneration of intracellular proteins and DNA, inhibition of MRSA glycolysis, and intervention in the energy metabolism of bacteria. Overall, this metal-antibiotic nanotherapeutics strategy provides new insight for combating extra- and intracellular infections caused by MRSA-induced OM.
Collapse
Affiliation(s)
- Hui Lv
- Department of Emergency and trauma orthopedics, the 958th Hospital of Chinese People's Liberation Army, Army Medical University (Third Military Medical University), Chongqing 400023, China
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ming Yang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yusheng Yang
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhenzhen Tang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuan Guo
- Department of Emergency and trauma orthopedics, the 958th Hospital of Chinese People's Liberation Army, Army Medical University (Third Military Medical University), Chongqing 400023, China
| | - Jiangling Zhou
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yingtao Gui
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Rong Huang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Juan Cai
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Bo Yu
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jing Yang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ying Bao
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhongrong Zhang
- Department of Emergency and trauma orthopedics, the 958th Hospital of Chinese People's Liberation Army, Army Medical University (Third Military Medical University), Chongqing 400023, China
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Tianyong Hou
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
34
|
Li W, Huang Y, Gu D, Peng S, Zhang B, Peng F, Zhang D, Li M, Xiao J, Jia Z, Qiu L. Ascorbate-loaded MgFe layered double hydroxide for osteomyelitis treatment. J Control Release 2025; 378:1045-1060. [PMID: 39740696 DOI: 10.1016/j.jconrel.2024.12.072] [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: 08/03/2024] [Revised: 12/15/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
Bacterial infections evoke considerable apprehension in orthopedics. Traditional antibiotic treatments exhibit cytotoxic effects and foster bacterial resistance, thereby presenting an ongoing and formidable obstacle in the realm of therapeutic interventions. Achieving bacterial eradication and osteogenesis are critical requirements for bone infection treatment. Herein, we design and fabricate a nanoenzyme-mimicking drug through the co-precipitation process, integrating MgFe layered double hydroxide with ascorbic acid (AA@LDH), to facilitate the simultaneous presence of these two unique functionalities. Within a bacterial acidic milieu, the degradation of the AA@LDH nanosystem prompts ascorbic acid to undergo a pro-oxidative transformation, generating an abundance of reactive oxygen species (ROS). These ROS overwhelm bacterial cellular processes, including nucleic acid replication, cell wall construction, virulence factor production, biosynthetic pathways, and energy generation. This disruption culminates in substantial bacterial mortality, as substantiated by RNA sequencing data. Hence, the AA@LDH nano system exhibits an in vitro antibacterial rate of approximately 100 % and 99 %, against S.aureus and E. coli, respectivaly. Additionally, the AA@LDH could directly accelerate osteogenic differentiation in vitro, evidenced by a 50 % increase in alkaline phosphatase activity and a 270 % improvement in extracellular matrix mineralization capability. Moreover, it enhances osteointegration process in vivo by favorably reshaping the osteogenic immune microenvironment. This innovative nanosystem for delivery offers new strategies that concurrently combat bacterial infections, mitigate inflammation, and induce tissue regeneration, marking a significant advancement in the realm of advanced materials and its applications.
Collapse
Affiliation(s)
- Wei Li
- Department of Sports Medicine, Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yuliang Huang
- Department of Traumatology and Orthopaedic Surgery, Huizhou Central People's Hospital, Huizhou 516001, China; Hui Zhou-Hong Kong Bone Health Joint Research Center, Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou 516001, China
| | - Dongqiang Gu
- Department of Sports Medicine, Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Sijun Peng
- Department of Traumatology and Orthopaedic Surgery, Huizhou Central People's Hospital, Huizhou 516001, China; Hui Zhou-Hong Kong Bone Health Joint Research Center, Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou 516001, China
| | - Bao Zhang
- Department of Orthopedics, Guangyuan Central Hospital, Guangyuan 628000, China
| | - Feng Peng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Dongdong Zhang
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mingjun Li
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China
| | - Jin Xiao
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| | - Zhiwei Jia
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101100, China.
| | - Longhai Qiu
- Department of Traumatology and Orthopaedic Surgery, Huizhou Central People's Hospital, Huizhou 516001, China; Hui Zhou-Hong Kong Bone Health Joint Research Center, Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou 516001, China.
| |
Collapse
|
35
|
Figueiredo J, Lindo J, Chaves C, Nogueira C. Clinical and epidemiological differences in staphylococcal osteoarticular infections: insights for developing hospital-based infection control interventions. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2025; 35:66. [PMID: 39921754 PMCID: PMC11807055 DOI: 10.1007/s00590-025-04184-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/25/2025] [Indexed: 02/10/2025]
Abstract
PURPOSE Osteoarticular infections (OAI) are serious clinical conditions with Staphylococcus aureus and Coagulase-negative Staphylococcus (CoNS) responsible for up to two-thirds of cases. This work aimed to compare the epidemiological, clinical, and microbiological characteristics of OAI caused by S. aureus versus CoNS to aid in clinical management and infection control strategies. METHODS A single-centre retrospective study was performed at the Centro Hospitalar e Universitário de Coimbra for the period of January 2011 to December 2021. A total of 458 cases of OAI were gathered. Data was retrieved from medical records and statistical analysis was performed with SPSS. RESULTS S. aureus accounted for 60.7% of infections, followed by S. epidermidis (29.9%). Independent risk factors for S. aureus infections included being male (p < 0.001; OR = 0.47) and a history of osteomyelitis (p < 0.001; OR = 0.18). In contrast, CoNS infections were associated with older age (p = 0.018), carrying a prosthetic device (p < 0.001; OR = 2.92), and a prior periprosthetic infection (p = 0.023; OR = 1.86). Both groups exhibited significant antimicrobial resistance, with CoNS showing greater resistance to gentamicin, linezolid, teicoplanin and trimethoprim-sulfamethoxazole, while S. aureus was more commonly resistant to clindamycin. CONCLUSION Our findings show the distinct characteristics of OAI caused by S. aureus and CoNS, highlighting the need for targeted risk factor management and tailored empiric antibiotic therapy to reduce incidence and improve outcomes.
Collapse
Affiliation(s)
- João Figueiredo
- FMUC-Faculty of Medicine, University Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Jorge Lindo
- FMUC-Faculty of Medicine, University Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- CNC-UC - Centre for Neuroscience and Cell Biology, University Coimbra, Coimbra, Portugal
- CiBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Catarina Chaves
- Microbiology Laboratory, Pathology Unit, Centro Hospitalar e Universitário de Coimbra, Portugal, Coimbra, Portugal
| | - Célia Nogueira
- FMUC-Faculty of Medicine, University Coimbra, Coimbra, Portugal.
- CNC-UC - Centre for Neuroscience and Cell Biology, University Coimbra, Coimbra, Portugal.
- CiBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
36
|
Yang Y, Zhou H, Li F, Zhang Y, Yang J, Shen Y, Hu N, Zou Q, Qin L, Zeng H, Huang W. Staphylococcus aureus induces mitophagy via the HDAC11/IL10 pathway to sustain intracellular survival. J Transl Med 2025; 23:156. [PMID: 39905391 DOI: 10.1186/s12967-025-06161-7] [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: 09/18/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND The immune evasion and prolonged survival of Staphylococcus aureus (S. aureus) within macrophages are key factors contributing to the difficulty in curing osteomyelitis. Although macrophages play a vital role as innate immune cells, the mechanisms by which S. aureus survives within them and suppresses host immune functions remain incompletely understood. METHODS This study employed confocal microscopy, flow cytometry, ELISA, and siRNA technology to assess the survival capacity of S. aureus within macrophages and the impact of inflammatory cytokines on its persistence. Proteomics was used to investigate the potential mechanisms and differential proteins involved in S. aureus intracellular survival. Additionally, confocal microscopy, flow cytometry, Mdivi-1 intervention, and Western blot were utilized to validate the role of mitophagy in supporting S. aureus survival. The study further explored how the HDAC11/IL10 axis enhances mitophagy to promote intracellular S. aureus survival by using HDAC11 overexpression, siRNA, and rapamycin intervention combined with confocal microscopy and flow cytometry. RESULTS The findings demonstrated that IL10 promotes mitophagy to clear mitochondrial reactive oxygen species (mtROS), thereby enhancing the intracellular survival of S. aureus within macrophages. Additionally, we discovered that the transcriptional repressor of IL10, HDAC11, was significantly downregulated during S. aureus infection. Overexpression of HDAC11 and the use of the autophagy activator rapamycin further validated that the HDAC11/IL10 axis regulates mitophagy via the mTOR pathway, which is essential for supporting S. aureus intracellular survival. CONCLUSION This study reveals that S. aureus enhances IL10 production by inhibiting HDAC11, thereby promoting mitophagy and mtROS clearance, which supports its survival within macrophages. These findings offer new insights into the intracellular survival mechanisms of S. aureus and provide potential therapeutic approaches for the clinical management of osteomyelitis.
Collapse
Affiliation(s)
- Yaji Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Haotian Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Feilong Li
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
- Department of Orthopaedics, The People's Hospital of Dazu, Chongqing, 402360, China
| | - Yanhao Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Jianye Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Yidong Shen
- Department of Orthopaedics, The First people's Hospital of Yancheng, Yancheng, Jiangsu, 224006, China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Leilei Qin
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China.
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
37
|
Dong Y, Hu Y, Hu X, Wang L, Shen X, Tian H, Li M, Luo Z, Cai C. Synthetic nanointerfacial bioengineering of Ti implants: on-demand regulation of implant-bone interactions for enhancing osseointegration. MATERIALS HORIZONS 2025; 12:694-718. [PMID: 39480512 DOI: 10.1039/d4mh01237b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Titanium and its alloys are the most commonly used biometals for developing orthopedic implants to treat various forms of bone fractures and defects, but their clinical performance is still challenged by the unfavorable mechanical and biological interactions at the implant-tissue interface, which substantially impede bone healing at the defects and reduce the quality of regenerated bones. Moreover, the impaired osteogenesis capacity of patients under certain pathological conditions such as diabetes and osteoporosis may further impair the osseointegration of Ti-based implants and increase the risk of treatment failure. To address these issues, various modification strategies have been developed to regulate the implant-bone interactions for improving bone growth and remodeling in situ. In this review, we provide a comprehensive analysis on the state-of-the-art synthetic nanointerfacial bioengineering strategies for designing Ti-based biofunctional orthopedic implants, with special emphasis on the contributions to (1) promotion of new bone formation and binding at the implant-bone interface, (2) bacterial elimination for preventing peri-implant infection and (3) overcoming osseointegration resistance induced by degenerative bone diseases. Furthermore, a perspective is included to discuss the challenges and potential opportunities for the interfacial engineering of Ti implants in a translational perspective. Overall, it is envisioned that the insights in this review may guide future research in the area of biometallic orthopedic implants for improving bone repair with enhanced efficacy and safety.
Collapse
Affiliation(s)
- Yilong Dong
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325016, China.
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Xinqiang Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Lingshuang Wang
- School of Life Sciences, Chongqing University, Chongqing, 400044, China.
| | - Xinkun Shen
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325016, China.
| | - Hao Tian
- Kairui Stomatological Hospital, Chengdu 610211, China
| | - Menghuan Li
- School of Life Sciences, Chongqing University, Chongqing, 400044, China.
| | - Zhong Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
- School of Life Sciences, Chongqing University, Chongqing, 400044, China.
| | - Chunyuan Cai
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325016, China.
| |
Collapse
|
38
|
Liang W, Li Y, Ji Y, Kang R, Zhang K, Su X, Li J, Ji M, Wu T, Cao X, Chen J, Huo J. Exosomes derived from bone marrow mesenchymal stem cells induce the proliferation and osteogenic differentiation and regulate the inflammatory state in osteomyelitis in vitro model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1695-1705. [PMID: 39168906 DOI: 10.1007/s00210-024-03357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024]
Abstract
Chronic osteomyelitis is a chronic bone infection characterized by progressive osteonecrosis and dead bone formation, which is closely related to persistent infection and chronic inflammation. Exosomes derived from bone marrow-derived mesenchymal stem cells (BMSC) play an important role in bone tissue regeneration and the modulation of inflammatory processes. However, their role and mechanism of action in osteomyelitis have not been reported so far. This paper explores the potential effect of BMSC-derived exosomes on osteomyelitis in vitro model with the aim of providing a theoretical basis for the treatment of osteomyelitis in the future. In this study, exosomes were isolated and extracted from BMSCs and identified. MC3T3-E1 cells were treated with Staphylococcal protein A (SPA) to establish an in vitro model of osteomyelitis. Next, the effects of BMSC-derived exosomes on cell proliferation, apoptosis, angiogenesis, and autophagy in MC3T3-E1 cells treated with SPA were evaluated. Results showed that the proliferation ability of MC3T3-E1 cells increased after co-culture with BMSC-derived exosomes. Moreover, exosomes induced autophagy and osteogenic differentiation in MC3T3-E1 cells. The mRNA and protein levels of factors related to proliferation, differentiation, apoptosis, autophagy, and angiogenesis including β-Catenin, Runx2, Bcl-2, VEGFA, and Beclin-1 upregulated in SPA-treated MC3T3-E1 cells, whereas the levels of inflammatory cytokines including TNF-α, IL-1β, and IL-6 decreased in the supernatant. The results showed that exosomes derived from BMSCs may participate in the attenuation of osteomyelitis by inducing proliferation and osteogenic differentiation and regulating the inflammatory state in bone cells.
Collapse
Affiliation(s)
- Wei Liang
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Yangui Li
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Yihua Ji
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Renjie Kang
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Kaixi Zhang
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Xueyuan Su
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Jiangbo Li
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Mingming Ji
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Taiyong Wu
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Xinjie Cao
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China
| | - Jianrui Chen
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China.
| | - Jianzhong Huo
- Department of Orthopaedics, Taiyuan Hospital of Peking University First Hospital (Taiyuan Central Hospital), No. 1, East Sandao Lane, Jiefang Road, Taiyuan, 030000, Shanxi Province, China.
| |
Collapse
|
39
|
Cheng Q, Yang Y, Li F, Li X, Qin L, Huang W. Dual-Energy Computed Tomography Iodine Maps: Application in the Diagnosis of Periprosthetic Joint Infection in Total Hip Arthroplasty. J Arthroplasty 2025; 40:499-505. [PMID: 39128781 DOI: 10.1016/j.arth.2024.08.007] [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: 03/05/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND The challenge of early and rapid diagnosis of periprosthetic joint infection (PJI) remains important. This study aimed to assess the efficacy of dual-energy computed tomography (DECT) iodine maps for diagnosing PJI in total hip arthroplasty. METHODS We prospectively enrolled 68 patients who had postoperative joint pain after hip arthroplasty. All patients underwent preoperative DECT iodine imaging to quantify iodine concentration (IC) in periprosthetic tissues during arterial and venous phases. The diagnostic efficacy of DECT iodine maps was evaluated by constructing receiver operating characteristic curves according to the Musculoskeletal Infection Society criteria. RESULTS Compared with erythrocyte sedimentation rate (area under the curve [AUC] = 0.837), polymorphonuclear cell percentage (AUC = 0.703), and C-reactive protein (AUC = 0.837), periprosthetic tissue venous-phase IC (AUC = 0.970) and arterial-phase IC (AUC = 0.964) exhibited outstanding discriminative capability between PJI and aseptic failure. The PJI cut-off point was venous IC = 1.225 mg/mL, with a sensitivity of 92.31% and specificity of 90.48%; for arterial IC = 1.065 mg/mL, the sensitivity was 96.15% and specificity was 90.70%. CONCLUSIONS This study demonstrates the great potential of DECT iodine maps for the diagnosis of PJI around hip arthroplasty, which helps to differentiate between periprosthetic infection and aseptic failure after hip arthroplasty.
Collapse
Affiliation(s)
- Qiang Cheng
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaji Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feilong Li
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Orthopaedics, The People's Hospital of Dazu, Chongqing, China
| | - Xiaobin Li
- Changdu People's Hospital of Xizang, Xizang, China
| | - Leilei Qin
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
40
|
Vanvelk N, de Mesy Bentley KL, Verhofstad MHJ, Metsemakers WJ, Moriarty TF, Siverino C. Development of an ex vivo model to study Staphylococcus aureus invasion of the osteocyte lacuno-canalicular network. J Orthop Res 2025; 43:446-456. [PMID: 39380444 DOI: 10.1002/jor.25988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/08/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024]
Abstract
Staphylococcus aureus has multiple mechanisms to evade the host's immune system and antibiotic treatment. One such mechanism is the invasion of the osteocyte lacuno-canalicular network (OLCN), which may be particularly important in recurrence of infection after debridement and antibiotic therapy. The aim of this study was to develop an ex vivo model to facilitate further study of S. aureus invasion of the OLCN and early-stage testing of antibacterial strategies against bacteria in this niche. The diameter of the canaliculi of non-infected human, sheep, and mouse bones was measured microscopically on Schmorl's picrothionin stained sections, showing a large overlap in canalicular diameter. S. aureus successfully invaded the OLCN in all species in vitro as revealed by presence in osteocyte lacunae in Brown and Brenn-stained sections and by scanning electron microscopy. Murine bones were then selected for further experiments, and titanium pins with either a wild-type or ΔPBP4 mutant S. aureus USA300 were placed trans-cortically and incubated for 2 weeks in tryptic soy broth. Wild-type S. aureus readily invaded the osteocyte lacunae in mouse bones while the ΔPBP4 showed a significantly lower invasion of the OLCN (p = 0.0005). Bone specimens were then treated with gentamicin, sitafloxacin, R14 bacteriophages, or left untreated. Gentamicin (p = 0.0027) and sitafloxacin (p = 0.0280) significantly reduced the proportion of S. aureus-occupied lacunae, whilst bacteriophage treatment had no effect. This study shows that S. aureus is able to invade the OLCN in an ex vivo model. This ex vivo model can be used for future early-stage studies before proceeding to in vivo studies.
Collapse
Affiliation(s)
- Niels Vanvelk
- AO Research Institute Davos, Davos, Switzerland
- Trauma Research Unit, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Center for Advanced Research Technology (CART), University of Rochester Medical Center Rochester, Rochester, New York, USA
| | - Michael H J Verhofstad
- Trauma Research Unit, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Willem-Jan Metsemakers
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven-University of Leuven, Leuven, Belgium
| | - Thomas F Moriarty
- AO Research Institute Davos, Davos, Switzerland
- Center for Muskuloskeletal Infections (ZMSI), University Hospital Basel, Basel, Switzerland
| | | |
Collapse
|
41
|
Ge M, Zhu W, Mei J, Hu T, Yang C, Lin H, Shi J. Piezoelectric-Enhanced Nanocatalysts Trigger Neutrophil N1 Polarization against Bacterial Biofilm by Disrupting Redox Homeostasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2409633. [PMID: 39350533 DOI: 10.1002/adma.202409633] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/18/2024] [Indexed: 02/13/2025]
Abstract
Strategies of manipulating redox signaling molecules to inhibit or activate immune signals have revolutionized therapeutics involving reactive oxygen species (ROS). However, certain diseases with dual resistance barriers to the attacks by both ROS and immune cells, such as bacterial biofilm infections of medical implants, are difficult to eradicate by a single exogenous oxidative stimulus due to the diversity and complexity of the redox species involved. Here, this work demonstrates that metal-organic framework (MOF) nanoparticles capable of disrupting the bacterial ROS-defense system can dismantle bacterial redox resistance and induce potent antimicrobial immune responses in a mouse model of surgical implant infection by simultaneously modulating redox homeostasis and initiating neutrophil N1 polarization in the infection microenvironment. Mechanistically, the piezoelectrically enhanced MOF triggers ROS production by tilting the band structure and acts synergistically with the aurintricarboxylic acid loaded within the MOF, which inhibits the activity of the cystathionine γ-cleaving enzyme. This leads to biofilm structure disruption and antigen exposure through homeostatic imbalance and synergistic activation of neutrophil N1 polarization signals. Thus, this study provides an alternative but promising strategy for the treatment of antibiotic-resistant biofilm infections.
Collapse
Affiliation(s)
- Min Ge
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, China
| | - Wanbo Zhu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Jiawei Mei
- Department of Orthopaedics, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
| | - Tingting Hu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, China
| | - Chuang Yang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Han Lin
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, China
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200331, China
| | - Jianlin Shi
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, China
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200331, China
| |
Collapse
|
42
|
Fan M, Ren Y, Zhu Y, Zhang H, Li S, Liu C, Lv H, Chu L, Hou Z, Zhang Y, Pan H, Cui X, Chen W. Borosilicate bioactive glass synergizing low-dose antibiotic loaded implants to combat bacteria through ATP disruption and oxidative stress to sequentially achieve osseointegration. Bioact Mater 2025; 44:184-204. [PMID: 39502840 PMCID: PMC11535878 DOI: 10.1016/j.bioactmat.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Bone infection is a catastrophe in clinical orthopedics. Despite being the standard therapy for osteomyelitis, antibiotic-loaded polymethyl methacrylate (PMMA) cement has low efficiency against bacteria in biofilms. Furthermore, high-dose antibiotic-loaded implants carry risks of bacterial resistance, tissue toxicity, and impairment of local tissue healing. By incorporating borosilicate bioactive glass (BSG) into low-dose gentamicin sulfate (GS)-loaded PMMA cement, an intelligent strategy that synergistically eradicates bacteria and sequentially promotes osseointegration, was devised. Results showed that BSG did not compromises the handling properties of the cement, but actually endowed it with an ionic and alkaline microenvironment, thereby damaging the integrity of bacterial cell walls and membranes, inhibiting ATP synthesis by disrupting the respiratory chain in cell membranes and glycogen metabolism, and elevating reactive oxygen species (ROS) levels by weakening antioxidant components (peroxisomes and carotenoids). These antibacterial characteristics of BSG synergistically reinforced the effectiveness of GS, which was far below the actual clinical dosage, achieving efficient bacterial killing and biofilm clearance by binding to the 30S subunit of ribosomes. Furthermore, the released GS and the ionic and alkaline microenvironment from the implants fostered the osteogenic activity of hBMSCs in vitro and coordinately enhanced osseointegration in vivo. Collectively, this study underscores that BSG incorporation offers a promising strategy for reducing antibiotic dosage while simultaneously enhancing the antibacterial activity and osteogenesis of implants. This approach holds potential for resolving the conflict between bacterial resistance and bone infection.
Collapse
Affiliation(s)
- Mengke Fan
- Department of Orthopaedic Surgery the Hebei Medical University Third Hospital, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Engineering Research Center of Orthopedic Minimally Invasive Intelligent Equipment of Ministry of Education, Key Laboratory of Biomechanics of Hebei Province, No.139 Ziqiang Road, Shijiazhuang, 050051, PR China
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Youliang Ren
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong Distinct, Chongqing, 400010, PR China
- Department of Orthopaedics, Guizhou Provincial People's Hospital, Guiyang, 550000, PR China
| | - Yanbin Zhu
- Department of Orthopaedic Surgery the Hebei Medical University Third Hospital, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Engineering Research Center of Orthopedic Minimally Invasive Intelligent Equipment of Ministry of Education, Key Laboratory of Biomechanics of Hebei Province, No.139 Ziqiang Road, Shijiazhuang, 050051, PR China
| | - Hao Zhang
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen, PR China
| | - Shuaijie Li
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Chunyu Liu
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen, PR China
| | - Hongzhi Lv
- Department of Orthopaedic Surgery the Hebei Medical University Third Hospital, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Engineering Research Center of Orthopedic Minimally Invasive Intelligent Equipment of Ministry of Education, Key Laboratory of Biomechanics of Hebei Province, No.139 Ziqiang Road, Shijiazhuang, 050051, PR China
| | - Lei Chu
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong Distinct, Chongqing, 400010, PR China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery the Hebei Medical University Third Hospital, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Engineering Research Center of Orthopedic Minimally Invasive Intelligent Equipment of Ministry of Education, Key Laboratory of Biomechanics of Hebei Province, No.139 Ziqiang Road, Shijiazhuang, 050051, PR China
| | - Yingze Zhang
- Department of Orthopaedic Surgery the Hebei Medical University Third Hospital, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Engineering Research Center of Orthopedic Minimally Invasive Intelligent Equipment of Ministry of Education, Key Laboratory of Biomechanics of Hebei Province, No.139 Ziqiang Road, Shijiazhuang, 050051, PR China
| | - Haobo Pan
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen, PR China
| | - Xu Cui
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen, PR China
| | - Wei Chen
- Department of Orthopaedic Surgery the Hebei Medical University Third Hospital, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Engineering Research Center of Orthopedic Minimally Invasive Intelligent Equipment of Ministry of Education, Key Laboratory of Biomechanics of Hebei Province, No.139 Ziqiang Road, Shijiazhuang, 050051, PR China
| |
Collapse
|
43
|
Shi T, Wu Q, Ruan Z, Luo Z, Wang W, Guo Z, Ma Y, Wang X, Chu G, Lin H, Ge M, Chen Y. Resensitizing β-Lactams by Reprogramming Purine Metabolism in Small Colony Variant for Osteomyelitis Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410781. [PMID: 39656854 PMCID: PMC11791937 DOI: 10.1002/advs.202410781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/27/2024] [Indexed: 12/17/2024]
Abstract
Small colony variant (SCV) is strongly linked to antibiotic resistance and the persistence of osteomyelitis. However, the intrinsic phenotypic instability of SCV has hindered a thorough investigation of its pathogenic mechanisms. In this study, phenotypically stable SCV strains are successfully recovered from clinical specimens, characterized by elevated drug resistance and reduced immunogenicity. Multi-omics analysis revealed that the acquired high drug resistance is associated with altered flux in the purine metabolism pathway, attributable to mutations in the hypoxanthine phosphoribosyltransferase (hpt) gene. Furthermore, this study innovatively discovered that lonidamine, an inhibitor of cellular energy metabolism, can effectively mitigate SCV resistance to β-lactam antibiotics, thereby facilitating its eradication. The underlying mechanism involves the reprogramming of purine metabolism. Therefore, a co-delivery system for lonidamine and oxacillin is constructed with amino-modified dendritic mesoporous silica as a carrier, which showed high efficacy and safety in combating SCV both in vitro and in vivo experiments. Overall, this study elucidated the pathogenic mechanisms of a class of clinically isolated SCV isolates with hpt mutations and provided a paradigm for treating SCV-associated osteomyelitis by reprogramming purine metabolism.
Collapse
Affiliation(s)
- Tingwang Shi
- Department of Orthopedic SurgeryShanghai Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RoadShanghai200233China
| | - Qiong Wu
- Department of Laboratory MedicineShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RoadShanghai200233China
| | - Zesong Ruan
- Department of Orthopedic SurgeryShanghai Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RoadShanghai200233China
| | - Zhiyuan Luo
- Department of Orthopedic SurgeryShanghai Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RoadShanghai200233China
| | - Wenbo Wang
- Department of Orthopedic SurgeryShanghai Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RoadShanghai200233China
| | - Zhao Guo
- Department of Orthopedic SurgeryShanghai Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RoadShanghai200233China
| | - Yihong Ma
- Department of Orthopedic SurgeryShanghai Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RoadShanghai200233China
| | - Xin Wang
- Department of Orthopedic SurgeryShanghai Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RoadShanghai200233China
| | - Guangyu Chu
- Department of Orthopedic SurgerySpine LabThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
- Joslin‐Beth Israel Deaconess Foot Center and The Rongxiang XuMDCenter for Regenerative TherapeuticsBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA02215USA
| | - Han Lin
- Shanghai Institute of Ceramics Chinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical SciencesShanghai200050China
| | - Min Ge
- Department of Electrical and Electronic EngineeringThe University of Hong KongPokfulam RoadHong Kong999077China
| | - Yunfeng Chen
- Department of Orthopedic SurgeryShanghai Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine600 Yishan RoadShanghai200233China
| |
Collapse
|
44
|
Li Y, Li J, Zhong Y, Zhang Q, Wu Y, Huang J, Pang K, Zhou Y, Xiao T, Wu Z, Sun W, He C. pH-responsive and nanoenzyme-loaded artificial nanocells relieved osteomyelitis efficiently by synergistic chemodynamic and cuproptosis therapy. Biomaterials 2025; 313:122762. [PMID: 39178559 DOI: 10.1016/j.biomaterials.2024.122762] [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: 04/29/2024] [Revised: 08/03/2024] [Accepted: 08/17/2024] [Indexed: 08/26/2024]
Abstract
Osteomyelitis is an osseous infectious disease that primarily affects children and the elderly with high morbidity and recurrence. The conventional treatments of osteomyelitis contain long-term and high-dose systemic antibiotics with debridements, which are not effective and lead to antibiotic resistance with serious side/adverse effects in many cases. Hence, developing novel antibiotic-free interventions against osteomyelitis (especially antibiotic-resistant bacterial infection) is urgent and anticipated. Here, a bone mesenchymal stem cell membrane-constructed nanocell (CFE@CM) was fabricated against osteomyelitis with the characteristics of acid-responsiveness, hydrogen peroxide self-supplying, enhanced chemodynamic therapeutic efficacy, bone marrow targeting and cuproptosis induction. Notably, mRNA sequencing was applied to unveil the underlying biological mechanisms and found that the biological processes related to copper ion binding, oxidative phosphorylation, peptide biosynthesis and metabolism, etc., were disturbed by CFE@CM in bacteria. This work provided an innovative antibiotic-free strategy against osteomyelitis through copper-enhanced Fenton reaction and distinct cuproptosis, promising to complement the current insufficient therapeutic regimen in clinic.
Collapse
Affiliation(s)
- Yuanhui Li
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Jian Li
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Yuxuan Zhong
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Qingshun Zhang
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Yuchun Wu
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Jinpeng Huang
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Kaicheng Pang
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Yuanyue Zhou
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Tong Xiao
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Zenghui Wu
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
| | - Wei Sun
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
| | - Chao He
- Department of Orthopedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Provincial Engineering Research Center for Biomedical Engineering, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
45
|
Cojutti PG, Tedeschi S, Zamparini E, Fornaro G, Zagarrigo M, De Paolis M, Viale P, Pea F. Could a Reduced Dose of 8 g of Continuous Infusion Fosfomycin Be Considered as Effective as and Safer than a Standard 16 g Dose When Combined with High-Dose Daptomycin in the Treatment of Staphylococcal osteoarticular Infections? Antibiotics (Basel) 2025; 14:139. [PMID: 40001383 PMCID: PMC11851862 DOI: 10.3390/antibiotics14020139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Daptomycin plus fosfomycin combination therapy is a valuable strategy for treating staphylococcal osteoarticular infections (OIs), but hypernatremia and hypokalemia due to sodium overload are important issues. The aim of this study was to assess the likelihood of attaining a pharmacokinetic/pharmacodynamic (PK/PD) target of AUC/MIC > 66.6 and/or of 70%t > MIC with continuous infusion (CI) fosfomycin at the recommended vs. reduced dose in patients with OIs receiving combination therapy with high-dose daptomycin. Adverse events were also evaluated. Methods: Patients with OIs treated with 8-10 mg/kg daily daptomycin plus CI fosfomycin, and who had a ≥1 TDM assessment of CI fosfomycin, were retrospectively included in the high-dose (16 g daily) or reduced-dose (<16 g daily) groups. The attainment of the PK/PD targets of 70%t > MIC and AUC/MIC > 66.6 up to an MIC of 32 mg/L was calculated. A CART analysis was used to identify a cut-off of fosfomycin AUC that indicated occurrence of hypernatremia and/or hypokalemia. Results: A total of 44 and 39 patients were included in the high- and reduced-dose groups, respectively. The two groups did not differ in terms of demographic characteristics, underlying infectious diseases and microbiological isolates. No differences between groups in attaining both PK/PD targets up to an MIC of 32 mg/L and in C-reactive protein reduction at the end of treatment were observed. Fosfomycin AUC > 8245 mg × h/L and >8326 mg × h/L were associated with hypernatremia and hypokalemia, respectively. Conclusions: CI fosfomycin at 8 g daily may reach optimal PK/PD target attainment with better safety than the recommended 16 g daily dose in patients with preserved renal function. Targeting fosfomycin AUC at 2131-8326 mg × h/L or steady-state concentration at 88.8-347 mg/L may be adequate for optimizing drug pharmacodynamics up to an MIC of 32 mg/L and minimizing the risk of hypernatremia and hypokalemia.
Collapse
Affiliation(s)
- Pier Giorgio Cojutti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (S.T.); (P.V.); (F.P.)
- Clinical Pharmacology Unit, Department of Integrated Infectious Risk Management, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Sara Tedeschi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (S.T.); (P.V.); (F.P.)
- Infectious Diseases Unit, Department of Integrated Infectious Risk Management, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologn, Italy; (E.Z.); (G.F.); (M.Z.)
| | - Eleonora Zamparini
- Infectious Diseases Unit, Department of Integrated Infectious Risk Management, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologn, Italy; (E.Z.); (G.F.); (M.Z.)
| | - Giacomo Fornaro
- Infectious Diseases Unit, Department of Integrated Infectious Risk Management, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologn, Italy; (E.Z.); (G.F.); (M.Z.)
| | - Manuel Zagarrigo
- Infectious Diseases Unit, Department of Integrated Infectious Risk Management, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologn, Italy; (E.Z.); (G.F.); (M.Z.)
| | - Massimiliano De Paolis
- Orthopaedics and Traumatology Unit, Department of Integrated Infectious Risk Management, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (S.T.); (P.V.); (F.P.)
- Infectious Diseases Unit, Department of Integrated Infectious Risk Management, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologn, Italy; (E.Z.); (G.F.); (M.Z.)
| | - Federico Pea
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (S.T.); (P.V.); (F.P.)
- Clinical Pharmacology Unit, Department of Integrated Infectious Risk Management, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
46
|
Tian JH, Huang S, Wang ZH, Li JJ, Song X, Jiang ZT, Shi BS, Zhao YY, Zhang HY, Wang KR, Hu XY, Zhang X, Guo DS. Supramolecular discrimination and diagnosis-guided treatment of intracellular bacteria. Nat Commun 2025; 16:1016. [PMID: 39863571 PMCID: PMC11762306 DOI: 10.1038/s41467-025-56308-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Pathogenic intracellular bacteria pose a significant threat to global public health due to the barriers presented by host cells hindering the timely detection of hidden bacteria and the effective delivery of therapeutic agents. To address these challenges, we propose a tandem diagnosis-guided treatment paradigm. A supramolecular sensor array is developed for simple, rapid, accurate, and high-throughput identification of intracellular bacteria. This diagnostic approach executes the significant guiding missions of screening a customized host-guest drug delivery system by disclosing the rationale behind the discrimination. We design eight azocalix[4]arenes with differential active targeting, cellular internalization, and hypoxia responsiveness to penetrate cells and interact with bacteria. Loaded with fluorescent indicators, these azocalix[4]arenes form a sensor array capable of discriminating eight intracellular bacterial species without cell lysis or separation. By fingerprinting specimens collected from bacteria-infected mice, the facilitated accurate diagnosis offers valuable guidance for selecting appropriate antibiotics. Moreover, mannose-modified azocalix[4]arene (ManAC4A) is screened as a drug carrier efficiently taken up by macrophages. Doxycycline loaded with ManAC4A exhibits improved efficacy against methicillin-resistant Staphylococcus aureus-infected peritonitis. This study introduces an emerging paradigm to intracellular bacterial diagnosis and treatment, offering broad potential in combating bacterial infectious diseases.
Collapse
Affiliation(s)
- Jia-Hong Tian
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Siyuan Huang
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Han Wang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Juan-Juan Li
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Xianhui Song
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Tao Jiang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Bing-Sen Shi
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ying-Ying Zhao
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Hui-Yan Zhang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ke-Rang Wang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Xin-Yue Hu
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
| | - Xinge Zhang
- College of Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China.
| | - Dong-Sheng Guo
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, Kashi University, Kashi, China.
- College of Chemistry and Environmental Sciences, Kashi University, Kashi, China.
| |
Collapse
|
47
|
Xia Y, Hu Z, Jin Q, Chen Q, Zhao C, Qiang R, Xie Z, Li L, Zhang H. Structural characteristics, functions, and counteracting strategies of biofilms in Staphylococcus aureus. Comput Struct Biotechnol J 2025; 27:488-500. [PMID: 39916696 PMCID: PMC11799891 DOI: 10.1016/j.csbj.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
Background Staphylococcus aureus (S. aureus) is a prevalent pathogen associated with a wide range of infections, exhibiting significant antibiotic resistance and posing therapeutic challenges in clinical settings. The formation of biofilms contributes to the emergence of resistant strains, further diminishing the efficacy of antibiotics. This, in turn, leads to chronic and recurrent infections, ultimately increasing the healthcare burden. Consequently, preventing and eliminating biofilms has become a critical focus in clinical management and research. Aim of review This review systematically examines the mechanisms underlying biofilm formation in S. aureus and its contribution to antibiotic resistance, emphasizing the essential roles biofilms play in maintaining structural integrity and enhancing resistance. It also analyses the protective mechanisms that fortify S. aureus biofilms against antimicrobial treatments. Furthermore, the review provides a comprehensive overview of recent therapeutic innovations, including enzymatic therapy, nanotechnology, gene editing, and phage therapy. Key scientific concepts of review Emerging therapeutic strategies present novel approaches to combat S. aureus biofilm-associated infections through various mechanisms. This review discusses recent advancements in these therapies, their practical challenges in clinical application, and provides an in-depth analysis of each strategy's mechanisms and therapeutic potential. By mapping future research directions, this review aims to refine anti-biofilm strategies to control infection progression and effectively mitigate recurrence.
Collapse
Affiliation(s)
- Yanze Xia
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhenghui Hu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chenhao Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Qiang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liubing Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| |
Collapse
|
48
|
Li W, Ding Q, Li M, Zhang T, Li C, Qi M, Dong B, Fang J, Wang L, Kim JS. Stimuli-responsive and targeted nanomaterials: Revolutionizing the treatment of bacterial infections. J Control Release 2025; 377:495-523. [PMID: 39580080 DOI: 10.1016/j.jconrel.2024.11.044] [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: 08/15/2024] [Revised: 10/13/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Bacterial infections have emerged as a major threat to global public health. The effectiveness of traditional antibiotic treatments is waning due to the increasing prevalence of antimicrobial resistance, leading to an urgent demand for alternative antibacterial technologies. In this context, antibacterial nanomaterials have proven to be powerful tools for treating antibiotic-resistant and recurring infections. Targeting nanomaterials not only enable the precise delivery of bactericidal agents but also ensure controlled release at the infection site, thereby reducing potential systemic side effects. This review collates and categorizes nanomaterial-based responsive and precision-targeted antibacterial strategies into three key types: exogenous stimuli-responsive (including light, ultrasound, magnetism), bacterial microenvironment-responsive (such as pH, enzymes, hypoxia), and targeted antibacterial action (involving electrostatic interaction, covalent bonding, receptor-ligand mechanisms). Furthermore, we discuss recent advances, potential mechanisms, and future prospects in responsive and targeted antimicrobial nanomaterials, aiming to provide a comprehensive overview of the field's development and inspire the formulation of novel, precision-targeted antimicrobial strategies.
Collapse
Affiliation(s)
- Wen Li
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Qihang Ding
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China; Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Meiqi Li
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Tianshou Zhang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chunyan Li
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Manlin Qi
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130021, China.
| | - Jiao Fang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Lin Wang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
49
|
Liu Y, Zheng Y, Ding S. Development and validation of a prognostic nomogram model for severe osteomyelitis patients. Sci Rep 2025; 15:318. [PMID: 39747915 PMCID: PMC11695742 DOI: 10.1038/s41598-024-83418-z] [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: 02/03/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
After severe infection in osteomyelitis patients in the Intensive Care Unit (ICU), there's a higher risk of mortality. However, limited research exists on predicting prognosis. Develop a predictive model for 1-year mortality risk in ICU-admitted osteomyelitis patients to inform clinical diagnosis and treatment. MIMIC IV database was used to retrieve ICU data for osteomyelitis patients. The data were randomly split into training and validation sets (7:3 ratio). Univariate and multiple logistic regression identified independent predictors of one-year mortality and constructed a risk prediction nomogram in the training set. Predictive value of the nomogram was assessed using C-indexes, ROC curves, DCA, CIC and calibration curves. This study included a total of 1153 osteomyelitis patients, with 137 deaths within one year. These patients were randomly divided into training (N = 807) and validation (N = 346) sets. In the training set, multiple features were identified as key predictors of one-year mortality in osteomyelitis patients in the ICU. These factors were incorporated into the nomogram model, demonstrating good identification performance, with AUCs of 0.872 and 0.826 for the training and validation sets, respectively. The calibration curve and ROC curve indicate excellent predictive accuracy. DCA suggests strong clinical utility and robust predictive efficiency. Further analysis through CIC illustrates the clinical effectiveness of this predictive model. We have developed a nomogram model to predict the 1-year mortality rate for osteomyelitis patients admitted to the ICU, providing valuable predictive information for clinical decision-making.
Collapse
Affiliation(s)
- Yunlong Liu
- Department of Pediatric Surgery, Women and Children's Hospital Affiliated to Ningbo University, No. 339 Liuting Street, Ningbo, 315012, Zhejiang Province, China.
| | - Yan Zheng
- Department of School of Foundation, Zhejiang Pharmaceutical University, Ningbo, China
| | - Sheng Ding
- Department of Pediatric Surgery, Women and Children's Hospital Affiliated to Ningbo University, No. 339 Liuting Street, Ningbo, 315012, Zhejiang Province, China
| |
Collapse
|
50
|
Li M, Zhao P, Wang J, Zhang X, Li J. Functional antimicrobial peptide-loaded 3D scaffolds for infected bone defect treatment with AI and multidimensional printing. MATERIALS HORIZONS 2025; 12:20-36. [PMID: 39484845 DOI: 10.1039/d4mh01124d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Infection is the most prevalent complication of fractures, particularly in open fractures, and often leads to severe consequences. The emergence of bacterial resistance has significantly exacerbated the burden of infection in clinical practice, making infection control a significant treatment challenge for infectious bone defects. The implantation of a structural stent is necessary to treat large bone defects despite the increased risk of infection. Therefore, there is a need for the development of novel antibacterial therapies. The advancement in antibacterial biomaterials and new antimicrobial drugs offers fresh perspectives on antibacterial treatment. Although antimicrobial 3D scaffolds are currently under intense research focus, relying solely on material properties or antibiotic action remains insufficient. Antimicrobial peptides (AMPs) are one of the most promising new antibacterial therapy approaches. This review discusses the underlying mechanisms behind infectious bone defects and presents research findings on antimicrobial peptides, specifically emphasizing their mechanisms and optimization strategies. We also explore the potential prospects of utilizing antimicrobial peptides in treating infectious bone defects. Furthermore, we propose that artificial intelligence (AI) algorithms can be utilized for predicting the pharmacokinetic properties of AMPs, including absorption, distribution, metabolism, and excretion, and by combining information from genomics, proteomics, metabolomics, and clinical studies with computational models driven by machine learning algorithms, scientists can gain a comprehensive understanding of AMPs' mechanisms of action, therapeutic potential, and optimizing treatment strategies tailored to individual patients, and through interdisciplinary collaborations between computer scientists, biologists, and clinicians, the full potential of AI in accelerating the discovery and development of novel AMPs will be realized. Besides, with the continuous advancements in 3D/4D/5D/6D technology and its integration into bone scaffold materials, we anticipate remarkable progress in the field of regenerative medicine. This review summarizes relevant research on the optimal future for the treatment of infectious bone defects, provides guidance for future novel treatment strategies combining multi-dimensional printing with new antimicrobial agents, and provides a novel and effective solution to the current challenges in the field of bone regeneration.
Collapse
Affiliation(s)
- Mengmeng Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Peizhang Zhao
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jingwen Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xincai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Jun Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|