1
|
Fu FS, Chen HH, Chen Y, Yuan Y, Zhao Y, Yu A, Zhang XZ. Engineered bacillus subtilis enhances bone regeneration via immunoregulation and anti-Infection. Bioact Mater 2025; 46:503-515. [PMID: 39868074 PMCID: PMC11760808 DOI: 10.1016/j.bioactmat.2025.01.003] [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: 10/26/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Chronic osteomyelitis caused by implant infections is a common complication following orthopedic surgery. Preventing bacterial infection and simultaneously improving bone regeneration are the key for osteomyelitis. Current treatments include systemic antibiotics and multiple surgical interventions, but the strategies available for treatment are limited. In this study, a multifunctional engineered Bacillus subtilis (B. sub) hydrogel with sulfasalazine (SSZ) is developed to treat methicillin-resistant Staphylococcus aureus (MRSA) infection and anti-inflammatory and promote bone regeneration. B. sub in alginate hydrogels protects B. sub from being cleared by the host immune system while allowing the release of its bioactive substances, including antibacterial peptides and anti-inflammatory agents such as SSZ. The results show that the engineered probiotic hydrogels exhibit excellent antibacterial efficacy against MRSA (97 %) and prevent the development of bacterial resistance. The antibacterial effect is primarily mediated through the secretion of bioactive peptides by B. sub, which not only inhibit MRSA growth but also reduce the likelihood of resistance development. Meanwhile, the probiotic hydrogel has a greater ability to induce M2 polarization of macrophages and promote angiogenesis, resulting in enhanced osteogenic differentiation in bone marrow mesenchymal stem cells (BMSCs) and thus enhancing bone regeneration. This engineered probiotic hydrogel offers a promising strategy by simultaneously combating bacterial infection and enhancing osteogenic differentiation for chronic osteomyelitis.
Collapse
Affiliation(s)
- Fang-Sheng Fu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Huan-Huan Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, PR China
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, PR China
| | - Yu Chen
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Ying Yuan
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Yong Zhao
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, PR China
| |
Collapse
|
2
|
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
|
3
|
Yu Z, Wang Z, Chen Y, Wang Y, Tang L, Xi Y, Lai K, Zhang Q, Li S, Xu D, Tian A, Wu M, Wang Y, Yang G, Gao C, Huang T. Programmed surface platform orchestrates anti-bacterial ability and time-sequential bone healing for implant-associated infection. Biomaterials 2025; 313:122772. [PMID: 39190942 DOI: 10.1016/j.biomaterials.2024.122772] [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: 01/15/2024] [Revised: 08/01/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024]
Abstract
Implant-associated infection (IAI) has become an intractable challenge in clinic. The healing of IAI is a complex physiological process involving a series of spatiotemporal connected events. However, existing titanium-based implants in clinic suffer from poor antibacterial effect and single function. Herein, a versatile surface platform based on the presentation of sequential function is developed. Fabrication of titania nanotubes and poly-γ-glutamic acid (γ-PGA) achieves the efficient incorporation of silver ions (Ag+) and the pH-sensitive release in response to acidic bone infection microenvironment. The optimized PGA/Ag platform exhibits satisfactory biocompatibility and converts macrophages from pro-inflammatory M1 to pro-healing M2 phenotype during the subsequent healing stage, which creates a beneficial osteoimmune microenvironment and promotes angio/osteogenesis. Furthermore, the PGA/Ag platform mediates osteoblast/osteoclast coupling through inhibiting CCL3/CCR1 signaling. These biological effects synergistically improve osseointegration under bacterial infection in vivo, matching the healing process of IAI. Overall, the novel integrated PGA/Ag surface platform proposed in this study fulfills function cascades under pathological state and shows great potential in IAI therapy.
Collapse
Affiliation(s)
- Zhou Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Zhaolong Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Yitong Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Yuchen Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Like Tang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Yue Xi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Kaichen Lai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Qi Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Shuangyang Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Danyu Xu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Anrong Tian
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Mengjie Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Tingben Huang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
4
|
Su J, Wu Y, Wang Z, Zhang D, Yang X, Zhao Y, Yu A. Probiotic biofilm modified scaffolds for facilitating osteomyelitis treatment through sustained release of bacteriophage and regulated macrophage polarization. Mater Today Bio 2025; 30:101444. [PMID: 39866782 PMCID: PMC11764121 DOI: 10.1016/j.mtbio.2025.101444] [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: 05/23/2024] [Revised: 12/09/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Osteomyelitis has gradually become a catastrophic complication in orthopedic surgery due to the formation of bacterial biofilms on the implant surface and surrounding tissue. The therapeutic challenges of antibiotic resistance and poor postoperative osseointegration provide inspiration for the development of bioactive implants. We have strategically designed bioceramic scaffolds modified with Lactobacillus reuteri (LR) and bacteriophages (phages) to achieve both antibacterial and osteogenic effects. Leveraging the tendency of bacteria to adhere to the surface of implants, bioceramics have been modified with LR biofilm to promote bone repair. The LR biofilm, sterilized by pasteurization, prevents sepsis caused by live bacteria and is biocompatible with phages. Phages, being natural enemies of bacteria, not only effectively kill bacteria and inhibit biofilm formation but also readily adsorb onto the surface of bioceramics. Hence, this scaffold, loaded with a phage cocktail, lysates specific bacterial populations, namely Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus). More importantly, the inactivated LR biofilm stimulates macrophages RAW264.7 to polarize towards an anti-inflammatory M2 phenotype, creating an immune microenvironment favorable for inducing osteogenic differentiation of rat mesenchymal stem cells in vitro. In a rat model of infectious cranial defects, the scaffold not only effectively eliminated S. aureus and alleviated associated inflammation but also mediated macrophage-mediated immunoregulation, thus resulting in effective osteogenesis. Collectively, these multifunctional modified scaffolds offer an integrated approach to both bacterium elimination and bone repair, presenting a new strategy for bioactive implants in the clinical management of osteomyelitis.
Collapse
Affiliation(s)
- Junwei Su
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Yifan Wu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Zheng Wang
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Dong Zhang
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Xianquan Yang
- Department of Orthopaedics, Gucheng County People's Hospital, Xiangyang, 441799, Hubei, China
| | - Yong Zhao
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| |
Collapse
|
5
|
Sheehy EJ, von Diemling C, Ryan E, Widaa A, O' Donnell P, Ryan A, Chen G, Brady RT, López-Noriega A, Zeiter S, Moriarty TF, O' Brien FJ. Antibiotic-eluting scaffolds with responsive dual-release kinetics facilitate bone healing and eliminate S. aureus infection. Biomaterials 2025; 313:122774. [PMID: 39208699 DOI: 10.1016/j.biomaterials.2024.122774] [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: 05/09/2024] [Revised: 08/08/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Osteomyelitis (OM) is a progressive, inflammatory infection of bone caused predominately by Staphylococcus aureus. Herein, we engineered an antibiotic-eluting collagen-hydroxyapatite scaffold capable of eliminating infection and facilitating bone healing. An iterative freeze-drying and chemical crosslinking approach was leveraged to modify antibiotic release kinetics, resulting in a layered dual-release system whereby an initial rapid release of antibiotic to clear infection was followed by a sustained controlled release to prevent reoccurrence of infection. We observed that the presence of microbial collagenase accelerated antibiotic release from the crosslinked layer of the scaffold, indicating that the material is responsive to microbial activity. As exemplar drugs, vancomycin and gentamicin-eluting scaffolds were demonstrated to be bactericidal, and supported osteogenesis in vitro. In a pilot murine model of OM, vancomycin-eluting scaffolds were observed to reduce S. aureus infection within the tibia. Finally, in a rabbit model of chronic OM, gentamicin-eluting scaffolds both facilitated radial bone defect healing and eliminated S. aureus infection. These results show that antibiotic-eluting collagen-hydroxyapatite scaffolds are a one-stage therapy for OM, which when implanted into infected bone defects simultaneously eradicate infection and facilitate bone tissue healing.
Collapse
Affiliation(s)
- Eamon J Sheehy
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland & Trinity College Dublin, Dublin, Ireland; AO Research Institute Davos, Davos, Switzerland
| | | | - Emily Ryan
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Amro Widaa
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Peter O' Donnell
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Alan Ryan
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland & Trinity College Dublin, Dublin, Ireland
| | - Gang Chen
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Robert T Brady
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Adolfo López-Noriega
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | - Fergal J O' Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland & Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
6
|
Xie E, Yuan Z, Chen Q, Hu J, Li J, Li K, Wang H, Ma J, Meng B, Zhang R, Mao H, Liang T, Wang L, Liu C, Li B, Han F. Programmed Transformation of Osteogenesis Microenvironment by a Multifunctional Hydrogel to Enhance Repair of Infectious Bone Defects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2409683. [PMID: 39840502 DOI: 10.1002/advs.202409683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/01/2025] [Indexed: 01/23/2025]
Abstract
Repair of infectious bone defects remains a serious problem in clinical practice owing to the high risk of infection and excessive reactive oxygen species (ROS) during the early stage, and the residual bacteria and delayed Osseo integrated interface in the later stage, which jointly creates a complex and dynamic microenvironment and leads to bone non-union. The melatonin carbon dots (MCDs) possess antibacterial and osteogenesis abilities, greatly simplifying the composition of a multifunctional material. Therefore, a multifunctional hydrogel containing MCDs (GH-MCD) is developed to meet the multi-stage and complex repair needs of infectious bone injury in this study. The GH-MCD can intelligently release MCDs responding to the acidic microenvironment to scavenge intracellular ROS and exhibit good antibacterial activity by inducing the production of ROS in bacteria and inhibiting the expression of secA2. Moreover, it has high osteogenesis and long-lasting antimicrobial activity during bone repair. RNA-seq results reveal that the hydrogels promote the repair of infected bone healing by enhancing cellular resistance to bacteria, balancing osteogenesis and osteoclastogenesis, and regulating the immune microenvironment. In conclusion, the GH-MCD can promote the repair of infectious bone defects through the programmed transformation of the microenvironment, providing a novel strategy for infectious bone defects.
Collapse
Affiliation(s)
- En Xie
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Zhangqin Yuan
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Qianglong Chen
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Jie Hu
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Jiaying Li
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Kexin Li
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Huan Wang
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Jinjin Ma
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Bin Meng
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Ruoxi Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, P. R. China
| | - Ting Liang
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Lijie Wang
- Sanitation & Environment Technology Institute of Soochow University Ltd., Suzhou, Jiangsu, 215000, P. R. China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Fengxuan Han
- Orthopedic Institute, Department of Orthopedic Surgery, Medical 3D Printing Center, The First Affiliated Hospital, Changzhou Geriatric hospital, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| |
Collapse
|
7
|
Al Amri A, Muzaffar R, Al Samsami I. MRSA septic arthritis of the pubic symphysis post vaginal delivery: A case report and literature review. Int J Surg Case Rep 2025; 127:110905. [PMID: 39848083 DOI: 10.1016/j.ijscr.2025.110905] [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: 12/12/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025] Open
Abstract
INTRODUCTION AND IMPORTANCE Septic arthritis of the pubic symphysis is a rare postpartum infection characterized by severe pelvic pain, fever, and elevated inflammatory markers. It is often underdiagnosed due to its rarity and nonspecific symptoms. It is commonly caused by Staphylococcus aureus, with methicillin-resistant Staphylococcus aureus (MRSA) septic being a rare but concerning pathogen. CASE PRESENTATION We report the case of a 36-year-old woman who developed septic arthritis of the pubic symphysis caused by MRSA after spontaneous vaginal delivery. CLINICAL DISCUSSION Differentiating between diastasis and septic arthritis of the pubic symphysis is crucial for treatment. Noninfective pubic osteitis can mimic septic arthritis, presenting a diagnostic challenge. Imaging, laboratory data, and cultures are essential for accurate diagnosis. MRSA colonisation during pregnancy and postpartum trauma may facilitate infection. MRI is the most sensitive imaging modality for early detection and monitoring. CONCLUSION Septic arthritis of the pubic symphysis secondary to MRSA is a rare cause of postpartum pelvic pain. Prompt diagnosis and treatment are essential for favourable outcomes.
Collapse
Affiliation(s)
- Ayman Al Amri
- Department of Surgery, Orthopaedic Division, Sultan Qaboos University Hospital, Muscat, Oman.
| | - Rahil Muzaffar
- Department of Surgery, Orthopaedic Division, Sultan Qaboos University Hospital, Muscat, Oman
| | - Ibtisam Al Samsami
- Department of Surgery, Orthopaedic Division, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
8
|
Xu T, Cheng H, Pei H, Wang J, Shi Y, Zhang X, Huang D. Emodin Enhanced Microwave-Responsive Heterojunction with Powerful Bactericidal Capacity and Immunoregulation for Curing Bacteria-Infected Osteomyelitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409979. [PMID: 39604818 PMCID: PMC11744657 DOI: 10.1002/advs.202409979] [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: 08/20/2024] [Revised: 11/13/2024] [Indexed: 11/29/2024]
Abstract
Eradication of osteomyelitis caused by bacterial infections is still a major challenge. Microwave therapy has the inherent advantage of deep penetration in curing deep tissue infections. However, the antibacterial efficiency of sensitizers is limited by the weak energy of microwaves. Here, a hybrid heterojunction system (Fe3O4/CuS/Emo) is designed for curing bacterially infected osteomyelitis. As an enhanced microwave sensitizer, it shows supernormal microwave response ability. Specifically, Fe3O4 acts as a matrix to mediate magnetic loss. After CuS loading, the heterogeneous interface forms induce significant interfacial polarization, which increasing dielectric loss. On the basis of the heterojunction formed by the two semiconductors, emodin is innovatively introduced to modify it. This integration not only accelerates the movement of charge carriers but also enhances polarization loss due to the numerous functional groups present on the surface. This further optimizes the microwave thermal and catalytic response. In addition, the unique anti-inflammatory properties of emodin confer the ability of hybrid heterojunction to regulate the immune microenvironment. In vivo studies reveal that heterojunction modified by emodin programmed elimination of bacteria and regulation of the immune microenvironment. It offers a revolutionary approach to the treatment of bacterial osteomyelitis.
Collapse
Affiliation(s)
- Tao Xu
- Department of Biomedical EngineeringResearch Center for Nano‐biomaterials & Regenerative MedicineCollege of Artificial IntelligenceTaiyuan University of TechnologyTaiyuan030024China
| | - Hao Cheng
- Department of Biomedical EngineeringResearch Center for Nano‐biomaterials & Regenerative MedicineCollege of Artificial IntelligenceTaiyuan University of TechnologyTaiyuan030024China
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Hailiang Pei
- Department of Biomedical EngineeringResearch Center for Nano‐biomaterials & Regenerative MedicineCollege of Artificial IntelligenceTaiyuan University of TechnologyTaiyuan030024China
| | - Jiameng Wang
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Yiwei Shi
- NHC Key Laboratory of PneumoconiosisDepartment of Pulmonary and Critical Care MedicineFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Xiangyu Zhang
- Department of Biomedical EngineeringResearch Center for Nano‐biomaterials & Regenerative MedicineCollege of Artificial IntelligenceTaiyuan University of TechnologyTaiyuan030024China
- Shanxi Key Laboratory of Biomedical Metal MaterialsCollege of Materials Science and EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Di Huang
- Department of Biomedical EngineeringResearch Center for Nano‐biomaterials & Regenerative MedicineCollege of Artificial IntelligenceTaiyuan University of TechnologyTaiyuan030024China
| |
Collapse
|
9
|
Zhou H, Ren Y, Zou K, Jin Y, Liu H, Jiang H, Shi L, Sheng X, Weeks J, Wang H, Xue T, Schwarz EM, Xie C, Deng Z, Wang L, Chu L. Efficacy of pH-Responsive Surface Functionalized Titanium Screws in Treating Implant-associated S. aureus Osteomyelitis with Biofilms Formation. Adv Healthc Mater 2025; 14:e2403261. [PMID: 39604325 PMCID: PMC11773098 DOI: 10.1002/adhm.202403261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Implant-associated Staphylococcus aureus (S. aureus) osteomyelitis (IASO) leads to high orthopedic implant failure rates due to the formation of Staphylococcal abscess community within the bone marrow and bacterial colonization in the osteocyte lacuno-canalicular network (OLCN). To address this, antimicrobial peptides (HHC36)-loaded titania nanotubes (NTs) are developed on titanium screws (Ti-NTs-P-A), which integrate pH-responsive polymethacrylic acid to control HHC36 release for eradicating bacteria in IASO. Colony-forming unit assay confirmed that Ti-NTs-P-A screws maintained sustainable antibacterial effectiveness, killing over 65% of S. aureus even after multiple bacterial solution replacements. Notably, Ti-NTs-P-A screws exhibit significant pH-responsive HHC36 release behavior and bactericidal activity, consistent with the phenotype of peptides-killed bacteria from scanning electron microscopy. Transcriptome sequencing results reveal that Ti-NTs-P-A screws interfered with ribosome formation and disrupted the arginine biosynthesis, which is crucial for bacterial survival in acidic environments. In the non-infected implant model, the bone-implant contact ratio of the Ti-NTs-P-A screw is 2.3 times that of the clinically used titanium screw. In an IASO model, Ti-NTs-P-A screws effectively eradicated bacteria within the OLCN, achieving an 80% infection control rate and desirable osteointegration. Collectively, Ti-NTs-P-A screws with pH-responsive antibacterial properties exhibit great potential for eradicating bacteria and achieving osseointegration in IASO.
Collapse
Affiliation(s)
- Hang Zhou
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Youliang Ren
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Kaixiong Zou
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006P. R. China
| | - Ying Jin
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Hang Liu
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Haitao Jiang
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Lei Shi
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xiaomin Sheng
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Jason Weeks
- Department of OrthopaedicsNew York Medical CollegeNew YorkNY10595USA
| | - Hannah Wang
- Department of Orthopaedics, Center for Musculoskeletal ResearchUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Thomas Xue
- Department of Orthopaedics, Center for Musculoskeletal ResearchUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Edward M. Schwarz
- Department of Orthopaedics, Center for Musculoskeletal ResearchUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Chao Xie
- Department of Orthopaedics, Center for Musculoskeletal ResearchUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Zhongliang Deng
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Lin Wang
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006P. R. China
| | - Lei Chu
- Department of OrthopaedicsThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| |
Collapse
|
10
|
Pengfei S, Yifan Y, Linhui L, Yimin L, Dan X, Shaowei G, Guanqing H, Yong W. Novel antibiotics against Staphylococcus aureus without detectable resistance by targeting proton motive force and FtsH. MedComm (Beijing) 2025; 6:e70046. [PMID: 39781293 PMCID: PMC11707430 DOI: 10.1002/mco2.70046] [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: 07/16/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 01/12/2025] Open
Abstract
The increased prevalence of methicillin-resistant Staphylococcus aureus (MRSA) and its biofilms poses a great threat to human health. Especially, S. aureus-related osteomyelitis was hardly cured even by conventional antibiotics combined with surgical treatment. The development of novel structural antibiotics is urgently needed. By high-throughput screening and rational design, we identified a small molecule C218-0546 and its optimized analog STK848198 with great antimicrobial potential against MRSA avoiding resistance occurrence. And significant synergistical antimicrobial effects were found between the molecules and conventional antibiotics. Mechanisms studies by transcriptomics, fluorescent probes, molecule dynamics, and plasma surface resonance indicated that the proton motive force as well as FtsH are the main potential targets of these molecules. The compounds exhibited excellent in vivo pharmacokinetics, toxicity profiles, and antimicrobial activities in the abscess model as well as the peritonitis-sepsis model. In addition, STK848198 was found to be effective against MRSA biofilms by interacting with the quorum sensing system. STK848198 also showed in vivo efficacy in the periprosthetic joint infection model. In all, our study identified a class of antimicrobials with novel scaffolds that could be potential alternatives for the treatment of MRSA and its biofilm-related infections.
Collapse
Affiliation(s)
- She Pengfei
- Department of Laboratory MedicineThe Third Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Yang Yifan
- Department of Laboratory MedicineThe Third Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Li Linhui
- Department of Laboratory MedicineThe Third Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Li Yimin
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| | - Xiao Dan
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| | - Guo Shaowei
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| | - Huang Guanqing
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| | - Wu Yong
- Department of Laboratory MedicineThe Affiliated Changsha Hospital of Xiangya School of Medicine (The First Hospital of Changsha)Central South UniversityChangshaHunanChina
| |
Collapse
|
11
|
Yan N, Zhou H, Jin P, Li T, Liu Q, Ning H, Ma Z, Feng L, Jin T, Deng Y, Wu Z. A Multifunctional Cobalt-Containing Implant for Treating Biofilm Infections and Promoting Osteointegration in Infected Bone Defects Through Macrophage-Mediated Immunomodulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409200. [PMID: 39587976 PMCID: PMC11744729 DOI: 10.1002/advs.202409200] [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: 08/06/2024] [Revised: 10/26/2024] [Indexed: 11/27/2024]
Abstract
Treating bone infections and ensuring bone recovery is one of the major global problems facing modern orthopedics. Prolonged antibiotic use may increase the risk of antimicrobial resistance, and inflammation caused by biofilms can obstruct tissue healing, making bone infection treatment even more challenging. The optimal treatment strategy combines immune response modification to promote osteogenesis with effective bacterial infection removal that does not require long-term antibiotic use. A one-step plasma immersion ion implantation approach is used to create titanium alloy implants incorporating cobalt. According to experimental findings, cobalt-containing titanium implants exhibit improved antibacterial activity by efficiently disrupting biofilm formations and reducing Methicillin-resistant Staphylococcus aureus adherence by over 80%. Additionally, the implants exhibit superior anti-inflammatory and osseointegration properties. RNA sequencing analysis reveals the potential mechanism of Co2+ in regulating the polarization of macrophages toward the anti-inflammatory M2 phenotype, which is crucial for creating an immune environment conducive to bone healing. Concurrently, these implants promote osteogenic differentiation while suppressing osteoclast activity, further supporting bone repair. Overall, without exogenous recombinant proteins or antibiotics, the implants effectively eradicate infections and expedite bone repair, offering a novel therapeutic strategy for complex skeletal diseases with clinical promise.
Collapse
Affiliation(s)
- Nongyang Yan
- Institute of Advanced TechnologyUniversity of Science and Technology of ChinaNo. 5089 Wangjiang West RoadHefeiAnhui230031China
| | - Hao Zhou
- Department of Spine SurgeryThe Third Xiangya HospitalCentral South UniversityNo. 138 Tongzi RoadChangshaHunan410013China
| | - Penghe Jin
- Department of Spine SurgeryThe Third Xiangya HospitalCentral South UniversityNo. 138 Tongzi RoadChangshaHunan410013China
| | - Tengfei Li
- Institute of Advanced TechnologyUniversity of Science and Technology of ChinaNo. 5089 Wangjiang West RoadHefeiAnhui230031China
| | - Qi Liu
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaNo. 96 Jinzhai RoadHefeiAnhui230026China
| | - Hao Ning
- Department of Spine SurgeryThe Third Xiangya HospitalCentral South UniversityNo. 138 Tongzi RoadChangshaHunan410013China
| | - Zhixin Ma
- Comprehensive supervision officeAnhui provincial Health Commission435 Tunbrook RoadHefei230032China
| | - Linfei Feng
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaNo. 96 Jinzhai RoadHefeiAnhui230026China
- Department of Oral and Maxillofacial SurgeryThe First Affiliated Hospital of Anhui Medical UniversityNo. 218 Jixi AvenueHeifeiAnhui230032China
| | - Tao Jin
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaNo. 96 Jinzhai RoadHefeiAnhui230026China
| | - Youwen Deng
- Department of Spine SurgeryThe Third Xiangya HospitalCentral South UniversityNo. 138 Tongzi RoadChangshaHunan410013China
| | - Zhengwei Wu
- Institute of Advanced TechnologyUniversity of Science and Technology of ChinaNo. 5089 Wangjiang West RoadHefeiAnhui230031China
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaNo. 96 Jinzhai RoadHefeiAnhui230026China
| |
Collapse
|
12
|
Huo S, Lyu Z, Wang X, Liu S, Chen X, Yang M, Liu Z, Yin X. Engineering mesoporous polydopamine-based potentiate STING pathway activation for advanced anti-biofilm therapy. Biomaterials 2025; 312:122739. [PMID: 39096840 DOI: 10.1016/j.biomaterials.2024.122739] [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: 03/13/2024] [Revised: 07/07/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
The biofilm-induced "relatively immune-compromised zone" creates an immunosuppressive microenvironment that is a significant contributor to refractory infections in orthopedic endophytes. Consequently, the manipulation of immune cells to co-inhibit or co-activate signaling represents a crucial strategy for the management of biofilm. This study reports the incorporation of Mn2+ into mesoporous dopamine nanoparticles (Mnp) containing the stimulator of interferon genes (STING) pathway activator cGAMP (Mncp), and outer wrapping by M1-like macrophage cell membrane (m-Mncp). The cell membrane enhances the material's targeting ability for biofilm, allowing it to accumulate locally at the infectious focus. Furthermore, m-Mncp mechanically disrupts the biofilm through photothermal therapy and induces antigen exposure through photodynamic therapy-generated reactive oxygen species (ROS). Importantly, the modulation of immunosuppression and immune activation results in the augmentation of antigen-presenting cells (APCs) and the commencement of antigen presentation, thereby inducing biofilm-specific humoral immunity and memory responses. Additionally, this approach effectively suppresses the activation of myeloid-derived suppressor cells (MDSCs) while simultaneously boosting the activity of T cells. Our study showcases the efficacy of utilizing m-Mncp immunotherapy in conjunction with photothermal and photodynamic therapy to effectively mitigate residual and recurrent infections following the extraction of infected implants. As such, this research presents a viable alternative to traditional antibiotic treatments for biofilm that are challenging to manage.
Collapse
Affiliation(s)
- Shicheng Huo
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Zhuocheng Lyu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaoyuan Wang
- Physical Examination Center, Xi'an International Medical Center Hospital, Xi'an, China
| | - Shichang Liu
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xuxu Chen
- Department of Sports Medicine, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Ming Yang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhongkai Liu
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Xinhua Yin
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
13
|
Senthilkumar D, Don TM, Liao YJ, Kuo CY. The art of biodegradable polymer design for the treatments against osteomyelitis. Int J Biol Macromol 2025; 285:138347. [PMID: 39638180 DOI: 10.1016/j.ijbiomac.2024.138347] [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: 04/25/2024] [Revised: 11/15/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Osteomyelitis arises from the incomplete treatment of the external wounds in the healing process, while bacterial infections persist within the bone marrow, leading to abscess formation. Osteomyelitis treatments generally involve three main aspects: rapid bactericidal action, sustained bacteriostasis, and induction of bone regeneration. However, current treatment methods, which often combine surgical debridement with long-term high-dose intravenous antibiotic administration or poly(methyl methacrylate) (PMMA) beads antibiotic therapy, suffer from significant drawbacks and limitations. In an era focused on environmental protection and sustainability, there is potential for biodegradable polymers to replace non-degradable plastic materials to reduce environmental pollution and achieve sustainable development in resources, society, and the economy. With this in mind, this review aims to explore the concept and design of applying various natural biodegradable polymers like gelatin, chitosan, cellulose, etc., and synthetic biodegradable polymers like polylactic acid, poly(lactic-co-glycolic) acid, polycaprolactone, etc. for osteomyelitis treatment, including (1) replacing PMMA with biodegradable polymer beads, (2) biodegradable polymer coatings on medical implants, (3) injectable biodegradable polymer hydrogels, and (4) biodegradable polymers as scaffolds for osteogenic cell growth. This article contributes to understanding and advancing biodegradable polymer applications in biomedicine through a comprehensive review and discussion of these four aspects.
Collapse
Affiliation(s)
- Dhayanithi Senthilkumar
- International Graduate Program of Energy and Optoelectronic Materials Program (EOMP), National Taipei University of Technology, Taipei City 10608, Taiwan; Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei City 10608, Taiwan
| | - Trong-Ming Don
- Department of Chemical and Materials Engineering, Tamkang University, New Taipei City 25137, Taiwan.
| | - Yu-Jie Liao
- International Graduate Program of Energy and Optoelectronic Materials Program (EOMP), National Taipei University of Technology, Taipei City 10608, Taiwan
| | - Chih-Yu Kuo
- International Graduate Program of Energy and Optoelectronic Materials Program (EOMP), National Taipei University of Technology, Taipei City 10608, Taiwan; Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei City 10608, Taiwan; High-value Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei, Taiwan.
| |
Collapse
|
14
|
Ni J, Huang S, Liang Z, Chen Z, Zhang S, Li G, An T. Concentration, pathogenic composition, and exposure risks of bioaerosol in large indoor public environments: A comparative study of urban and suburban areas. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177790. [PMID: 39615183 DOI: 10.1016/j.scitotenv.2024.177790] [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: 09/22/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024]
Abstract
Biological contamination in larger indoor environments can lead to the outbreak of various infectious diseases. This study aimed to compare the pollution profiles and associated health risks of airborne microorganisms in different indoor settings between urban and suburban areas by culturing, sequencing, and toxicological evaluation. The results indicated that the average level of culturable bacteria was higher in urban areas (955 ± 259 CFU/m3) compared to suburban areas (850 ± 85 CFU/m3), with the highest concentrations found in the market (2170 ± 798 CFU/m3) and gymnasium (2010 ± 300 CFU/m3). Conversely, the total number of airborne bacteria was higher in classroom (2.09 × 105) and laboratory (1.95 × 105 copies/m3), likely due to the presence of viable but non-culturable cells. Additionally, the concentrations of 0.5-2.0 μm total particles were higher in the market and cafeteria. Dominant airborne genera included Acinetobacter and Pseudomonas for bacteria, Cladosporium and Aspergillus for fungi, as well as Geneviridae and Herpesviridae for viruses. Bacterial and viral diversity and richness were significantly higher in suburban areas compared to urban areas, with distinct viral communities observed in hospital. Cytotoxicity assays revealed lower viability of cells in response to bioaerosols from the library (52.3 %) and laboratory (54.5 %); while lower proliferation rates were found for the cells exposed to bioaerosol from gymnasium (5.4 %) and market (6.0 %), suggesting higher toxicity of these environments. Additionally, bioaerosol exposure may impair cellular innate immunity by increasing the expression of IL-6, IL-8, TNF-α, IFN-γ. Our findings provide valuable information for assessing and controlling bioaerosol-related health risks in indoor environments.
Collapse
Affiliation(s)
- Jiasheng Ni
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China
| | - Simin Huang
- Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Basic Research Center of Excellence for Ecological Security and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Zhishu Liang
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Basic Research Center of Excellence for Ecological Security and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China.
| | - Zhen Chen
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China
| | - Simeng Zhang
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China
| | - Guiying Li
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Basic Research Center of Excellence for Ecological Security and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Taicheng An
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China; Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Basic Research Center of Excellence for Ecological Security and Green Development, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
15
|
Zhang B, Zhang H, Sun Y, Chen L, Sun K, Zhang Y, Hu X, Zhao K, Wu Z, Tang Y. Functionalized Bone Implant Inspired by Lattice Defense Strategy: Grid Management, Precise and Effective Multiple-Prevention of Osteomyelitis Recurrence and Promote Bone Regeneration. Adv Healthc Mater 2024:e2403058. [PMID: 39690830 DOI: 10.1002/adhm.202403058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/22/2024] [Indexed: 12/19/2024]
Abstract
Osteomyelitis with a high recurrence rate. Timely-prevention can avoid severe consequence and death. However, conventional drug response-release has the disadvantages of unnecessary release and waste, causing ineffective prevention. Inspired by "Lattice-defense technology", gridding lesion areas and constructing a "Triggered-precise response-release system" may be an effective multiple-prevention method. Here, a new strict pH-triggered response drug controlled-release mechanism was proposed innovatively to construct a "Triggered-precise response-release system" and achieve multiple-effective prevention. PO4 3--Ce3+ strict pH responsive release system is prepared through simultaneous hydration reaction of solution-polymerization and compounded in bone-implant. The dispersed system only targets micro-interface contact areas, achieving gridded management of the lesion site. In a normal environment, Ce3+ is captured by PO4 3- and kept electrostatic-attraction balance, ensuring the zero-concentration Ce3+ release continuously. Once osteomyelitis recurs and pH decrease, H+ at the interface will combine with PO4 3- under electrostatic drive and disrupt potential balance, achieving the release of Ce3+ only when the infection recurs. In vivo experiments was confirmed effective prevention and excellent promote bone regeneration. The adoption of "Lattice defense technology" has achieved accuracy spatiotemporal of drug delivery. Even if other lesion sites unfortunately recur again, effective-prevention can be guaranteed. Bone-implant show great potential in preventing osteomyelitis.
Collapse
Affiliation(s)
- Bo Zhang
- School of Materials Science and Engineering, Xi'an University of Technology, Xi'an, 710048, P. R. China
| | - Hao Zhang
- School of Materials Science and Engineering, Xi'an University of Technology, Xi'an, 710048, P. R. China
| | - Yani Sun
- School of Materials Science and Engineering, Xi'an University of Technology, Xi'an, 710048, P. R. China
| | - Lei Chen
- School of Materials Science and Engineering, Xi'an University of Technology, Xi'an, 710048, P. R. China
- Shaanxi Province Key Laboratory of Corrosion and Protection, Xi'an University of Technology, Xi'an, 710048, P. R. China
| | - Kaixin Sun
- School of Materials Science and Engineering, Xi'an University of Technology, Xi'an, 710048, P. R. China
| | - Yuming Zhang
- Institute of Orthopaedics, Xi'jing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Xiaofan Hu
- Institute of Orthopaedics, Xi'jing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Kang Zhao
- School of Materials Science and Engineering, Xi'an University of Technology, Xi'an, 710048, P. R. China
- Shaanxi Province Key Laboratory of Corrosion and Protection, Xi'an University of Technology, Xi'an, 710048, P. R. China
| | - Zixiang Wu
- Institute of Orthopaedics, Xi'jing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Yufei Tang
- School of Materials Science and Engineering, Xi'an University of Technology, Xi'an, 710048, P. R. China
- Shaanxi Province Key Laboratory of Corrosion and Protection, Xi'an University of Technology, Xi'an, 710048, P. R. China
| |
Collapse
|
16
|
Jin L, Liu H, Wang C, Liu X, Mao C, Zhang Y, Li Z, Zhu S, Jiang H, Cui Z, Zheng Y, Wu S. A Bacterial Capturing, Neural Network-Like Carbon Nanotubes/Prussian Blue/Puerarin Nanocomposite for Microwave Treatment of Staphylococcus Aureus-Infected Osteomyelitis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2407113. [PMID: 39420683 DOI: 10.1002/smll.202407113] [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: 08/15/2024] [Revised: 09/28/2024] [Indexed: 10/19/2024]
Abstract
Staphylococcus aureus (S. aureus)-infected osteomyelitis is a deep tissue infection that cannot be effectively treated with antibiotics. Microwave (MW) thermal therapy (MTT) and MW dynamic therapy (MDT) based on MW-responsive materials are promising for the therapy of bacteria-infected osteomyelitis occurring in deep tissues that cannot be effectively treated with antibiotics. In this work, the MW-responsive system of carbon nanotubes (CNTs)/Prussian blue (PB)/puerarin (Pue) with stable network-like structures is constructed. The PB is grown in situ on the CNTs, and its introduction not only reduces the aggregation of the network-like structures of the CNTs, but the large specific surface area and mesoporous structure can also provide many active sites for the adsorption of oxygen and polar molecules. Pue is a natural anti-inflammatory material that reduces inflammation at the infection site. The composite of the CNTs and PB avoids the skin effect and thus can improve dielectric and reflection losses. The MW thermal response of CNTs/PB/Pue is mainly due to the occurrence of reflection loss, dielectric loss, multiple reflections and scattering, interface polarization, and dipole polarization. In addition, under MW irradiation, the CNTs/PB/Pue can produce reactive oxygen species (ROS), such as singlet oxygen (1O2), hydroxyl radical (·OH).
Collapse
Affiliation(s)
- Liguo Jin
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Hanpeng Liu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Chaofeng Wang
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Xiangmei Liu
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
| | - Congyang Mao
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhaoyang Li
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Shengli Zhu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Hui Jiang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Beijing, 100871, China
| | - Shuilin Wu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Materials Science & Engineering, Peking University, Beijing, 100871, China
| |
Collapse
|
17
|
Fang X, Ding H, Chen Y, Wang Q, Yuan X, Zhang C, Huang J, Huang J, Lv J, Hu H, Huang C, Hu X, Lin Y, Zhang N, Zhou W, Huang Y, Li W, Niu S, Wu Z, Lin J, Yang B, Yuan T, Zhang W. Wireless Optogenetic Targeting Nociceptors Helps Host Cells Win the Competitive Colonization in Implant-Associated Infections. SMALL METHODS 2024; 8:e2400216. [PMID: 39087367 DOI: 10.1002/smtd.202400216] [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: 03/01/2024] [Revised: 06/20/2024] [Indexed: 08/02/2024]
Abstract
The role of nociceptive nerves in modulating immune responses to harmful stimuli via pain or itch induction remains controversial. Compared to conventional surgery, various implant surgeries are more prone to infections even with low bacterial loads. In this study, an optogenetic technique is introduced for selectively activating peripheral nociceptive nerves using a fully implantable, wirelessly rechargeable optogenetic device. By targeting nociceptors in the limbs of awake, freely moving mice, it is found that activation induces anticipatory immunity in the innervated territory and enhances the adhesion of various host cells to the implant surface. This effect mediates acute immune cell-mediated killing of Staphylococcus aureus on implants and enables the host to win "implant surface competition" against Staphylococcus aureus. This finding provides new strategies for preventing and treating implant-associated infections.
Collapse
Affiliation(s)
- Xinyu Fang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Haiqi Ding
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Yang Chen
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Qijin Wang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedics, Affiliated Mindong Hospital of Fujian Medical University, Fu'an, 355000, China
| | - Xuhui Yuan
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Chaofan Zhang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Jiagu Huang
- Department of Orthopedic Surgery, Ningde municipal Hospital, Ningde, 352000, China
| | - Jiexin Huang
- Department of Orthopedic Surgery, Nanping First Hospital, Nanping, 353000, China
| | - Jianhua Lv
- Department of Orthopedic Surgery, Affiliated Hospital of Putian University, Putian, 351100, China
| | - Hongxin Hu
- Department of Orthopedic Surgery, Affiliated Hospital of Putian University, Putian, 351100, China
| | - Changyu Huang
- Department of Orthopedic Surgery, Quanzhou Orthopedic-traumatological Hospital, Quanzhou, 362000, China
| | - Xueni Hu
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Yiming Lin
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Nanxin Zhang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Wei Zhou
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Ying Huang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Wenbo Li
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Susheng Niu
- Key Laboratory of Orthopedics & Traumatology of Traditional Chinese Medicine and Rehabilitation Ministry of Education, Fujian university of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Zhaoyang Wu
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Jianhua Lin
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Bin Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200000, China
| | - Wenming Zhang
- Department of Orthopedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350000, China
| |
Collapse
|
18
|
Alunni Cardinali M, Govoni M, Stefani S, Maso A, Storni E, Valenti F, Maglio M, Morresi A, Fioretto D, Dallari D, Sassi P. Combining Multiple Spectroscopic Techniques to Reveal the Effects of Staphylococcus aureus Infection on Human Bone Tissues. APPLIED SPECTROSCOPY 2024; 78:1295-1306. [PMID: 39344289 DOI: 10.1177/00037028241278903] [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: 10/01/2024]
Abstract
Osteomyelitis (OM) and periprosthetic joint infections (PJIs) are major public health concerns in Western countries due to increased life expectancy. Infections usually occur due to bacterial spread through fractures, implants, or blood-borne transmission. The pathogens trigger an inflammatory response that hinders bone tissue regeneration. Treatment requires surgical intervention, which involves the precise removal of infected tissue, wound cleansing, and local and systemic antibiotic administration. Staphylococcus aureus (SA) is one of the most common pathogens causing infection-induced OM and PJIs. It forms antimicrobial-resistant biofilms and is frequently found in healthcare settings. In this proof-of-concept, we present an approach based on multiple spectroscopic techniques aimed at investigating the effects of SA infection on bone tissue, as well as identifying specific markers useful to detect early bacterial colonization on the tissue surface. A cross-section of a human femoral diaphysis, with negative-culture results, was divided into three parts, and the cortical and trabecular regions were separated from each other. Two portions of each bone tissue type were infected with SA for one and seven days, respectively. Multiple techniques were used to investigate the impact of the infection on bone tissue, Brillouin-Raman microspectroscopy and attenuated total reflection Fourier transform infrared spectroscopy were employed to assess and develop a new noninvasive diagnostic method to detect SA by targeting the bone of the host. The results indicate that exposure to SA infection significantly alters the bone structure, especially in the case of the trabecular type, even after just one day. Moreover, Raman spectral markers of the tissue damage were identified, indicating that this technique can detect the effect of the pathogens' presence in bone biopsies and pave the way for potential application during surgery, due to its nondestructive and contactless nature.
Collapse
Affiliation(s)
| | - Marco Govoni
- Reconstructive Orthopaedic Surgery and Innovative Techniques - Musculoskeletal Tissue Bank, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sara Stefani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Alessandra Maso
- Laboratory of Microbiology and GMP Quality Control, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Storni
- Laboratory of Microbiology and GMP Quality Control, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Valenti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Melania Maglio
- Surgical Science and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Assunta Morresi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Daniele Fioretto
- Department of Physics and Geology, University of Perugia, Perugia, Italy
- CEMIN-Center of Excellence for Innovative Nanostructured Material, Perugia, Italy
| | - Dante Dallari
- Reconstructive Orthopaedic Surgery and Innovative Techniques - Musculoskeletal Tissue Bank, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Paola Sassi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| |
Collapse
|
19
|
Zhang Y, Xu T, Li T, Chen H, Xu G, Hu W, Li Y, Dong Y, Liu Z, Han B. A three-phase strategy of bionic drug reservoir scaffold by 3D printing and layer-by-layer modification for chronic relapse management in traumatic osteomyelitis. Mater Today Bio 2024; 29:101356. [PMID: 39687799 PMCID: PMC11648807 DOI: 10.1016/j.mtbio.2024.101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
We have developed a novel three-phase strategy for osteomyelitis treatment, structured into three distinct phases: the "strong antimicrobial" phase, the "monitoring and osteogenesis" phase and the "bone repair" phase. To implement this staged therapeutic strategy, we engineered a bionic drug reservoir scaffold carrying a dual-drug combination of antimicrobial peptides (AMPs) and simvastatin (SV). The scaffold integrated a bilayer gel drug-carrying structure, based on an induced membrane and combined with a 3D-printed rigid bone graft using a layer-by-layer modification strategy. The mechanical strength of the composite scaffold (73.40 ± 22.44 MPa) is comparable to that of cancellous bone. This scaffold enables controlled, sequential drug release through a spatial structure design and nanoparticle drug-carrying strategy. AMPs are released rapidly, with the release efficiency of 74.90 ± 8.19 % at 14 days (pH = 7.2), thus enabling rapid antimicrobial therapy. Meanwhile, SV is released over a prolonged period, with a release efficiency of 98.98 ± 0.05 % over 40 days in vitro simulations, promoting sustained osteogenesis and facilitating the treatment of intracellular infections by activating macrophage extracellular traps (METs). The antimicrobial, osteogenic and immunomodulatory effects of the scaffolds were verified through in vitro and in vivo experiments. It was demonstrated that composite scaffolds were able to combat the chronic recurrence of osteomyelitis after debridement, by providing rapid sterilization, stimulating METs formation, and supporting osteogenic repair.
Collapse
Affiliation(s)
- Yutong Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Tongtong Xu
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Tieshu Li
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
- Changchun University of Chinese Medicine, Changchun, 130117, China
- Affiliated Hospital of Yangzhou University, Yangzhou, 225009, China
| | - Hening Chen
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Guangzhe Xu
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Wenxin Hu
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yongting Li
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yue Dong
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Zhihui Liu
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Bing Han
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| |
Collapse
|
20
|
Geng S, Hu B, Guan Y, Jiang Y, Shu Z, Li C, Huang G. Advances of the multifaceted functions of PSTPIP2 in inflammatory diseases. Front Immunol 2024; 15:1492878. [PMID: 39660128 PMCID: PMC11628490 DOI: 10.3389/fimmu.2024.1492878] [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: 09/08/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
The complex interaction between the immune system and autoinflammatory disorders highlights the centrality of autoimmune mechanisms in the pathogenesis of autoinflammatory diseases. With the exploration of PSTPIP2, it has been discovered to play an inhibitory role in immune diseases, suggesting its potential utility in the research and treatment of rheumatic diseases. This review outlines the mechanisms of PSTPIP2 in chronic multifocal osteomyelitis (CMO), rheumatoid arthritis (RA), synovitis-acne-pustulosis-hyperostosis-osteitis (SAPHO) syndrome, liver diseases, renal diseases, pressure ulcer sepsis and diabetic obesity. The mechanisms include inhibiting the IL-1β inflammatory responses, NF-κB, ERK phosphorylation etc., promoting Erβ, and modulating the polarization of macrophage to prevent the inflammatory diseases. This review summarized current findings and offered perspectives on future research directions, laying a foundation for applying of PSTPIP2 in inflammatory diseases.
Collapse
Affiliation(s)
- Shaohui Geng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Bohan Hu
- School of Chinese Materia Medica , Beijing University of Chinese Medicine, Beijing, China
| | - Yiwei Guan
- School of Chinese Materia Medica , Beijing University of Chinese Medicine, Beijing, China
| | - Yijin Jiang
- School of Chinese Materia Medica , Beijing University of Chinese Medicine, Beijing, China
| | - Zixuan Shu
- School of Chinese Materia Medica , Beijing University of Chinese Medicine, Beijing, China
| | - Chen Li
- Department of Rheumatology, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
21
|
Zhang Y, Zhang M, Li M, Miao M, Shou D, Tong P. Icariin-Enhanced Osteoclast-Derived Exosomes Promote Repair of Infected Bone Defects by Regulating Osteoclast and Osteoblast Communication. Int J Nanomedicine 2024; 19:12389-12407. [PMID: 39588260 PMCID: PMC11587802 DOI: 10.2147/ijn.s483621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/08/2024] [Indexed: 11/27/2024] Open
Abstract
Background Infected bone defects pose a challenging clinical issue due to an imbalance of osteoclasts (OC) and osteoblasts (OB). Exosomes are crucial for intercellular signaling of OC and OB in bone repair. Icariin, has been shown to regulate the balance between OC and OB. However, the specific mechanisms by which icariin influences exosomes derived from osteoclasts, and subsequently impacts osteoblast activity, remain unclear. This study aims to investigate the effects of icariin-treated osteoclast-derived exosomes (ICA-OC-Exo) on osteoblast function and bone repair in cases of infected bone defects. Methods We investigated the exosome profile and localization of multivesicular bodies (MVB) and quantification of intraluminal vesicles (ILVs) in osteoclasts by using transmission electron microscopy. Additionally, the expressions of Rab27A and MITF, which are associated with exosome release, were determined through immunofluorescence staining and Western blot. The profiling of exosomal miRNA expression was conducted via miRNA-sequencing. The effects of ICA-OC-Exo on osteoblast differentiation were determined using RT-qPCR, Western blot, alkaline phosphatase staining. Additionally, ICA-OC-Exo was administered into the localized bone defect of the infected bone rat models, and bone formation was assessed using Micro-CT. Results Icariin increased the presence of MVBs in the cytoplasm through modulation of the MITF/Rab27A signaling pathway, resulting in higher number of ICA-OC-Exo compared to OC-Exo. Additionally, miR-331-3p expression in ICA-OC-Exo was found to be elevated compared to OC-Exo. ICA-OC-Exo was observed to stimulate osteoblast function by targeting FGF23, reducing DKK1, and subsequently upregulating ALP. In the in vivo study, ICA-OC-Exo exhibited the capacity to enhance bone healing at the site of a local bone defect following anti-infection treatment. Conclusion Icariin enhanced the quantification of OC-Exo and the expression of miRNA-331-3p in OC-Exo, leading to the regulation of osteoblast function via activation of the miRNA-331-3p/FGF23/DKK1 pathway. ICA-OC-Exo demonstrated potential clinical applicability in bone repair of infected bone defects.
Collapse
Affiliation(s)
- Yang Zhang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People’s Republic of China
| | - Minjie Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Mengying Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Maomao Miao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Dan Shou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People’s Republic of China
| |
Collapse
|
22
|
Liu Y, Li Y, Jiao L, Kang Y, Du B, Cai W, Cui H, Zhang R. Hypoxia-Activated Biodegradable Porphyrin-Based Covalent Organic Frameworks for Photodynamic and Photothermal Therapy of Wound Infection. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39557630 DOI: 10.1021/acsami.4c14909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Wound infections have gradually become a major threat to human health. Recently, covalent organic frameworks (COFs) have shown great potential in antibacterial and wound healing; however, difficult biodegradability and long-time in vivo retention limit their further application. Herein, biodegradable COFs containing porphyrin backbones and hypoxia-sensitive azobenzene group, namely, HRCOFs, are fabricated for photodynamic therapy (PDT) and photothermal therapy (PTT) of wound infection. Due to the introduction of a porphyrin molecule, HRCOFs can produce singlet oxygen (1O2) under 660 nm laser irradiation. The prepared HRCOFs can also generate thermal energy under 808 nm NIR laser irradiation. HRCOFs show excellent synergetic antibacterial ability against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) in vitro. The in vivo experiments also demonstrate synergistic PDT and PTT effects of HRCOFs against wound infection. Importantly, HRCOFs are response to wound microenvironment, can be degraded for clearance, and avoid some adverse effects caused by long-time retention in vivo, exhibiting good biocompatibility. In general, the obtained biodegradable HRCOFs with both photodynamic and photothermal effects can be used for antibacterial infections and provide great value for promoting wound healing.
Collapse
Affiliation(s)
- Yulong Liu
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Ya Li
- Department of School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Liqin Jiao
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Yefang Kang
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Baojie Du
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Wenwen Cai
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Hong Cui
- Department of School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People' Hospital, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
23
|
Yekani M, Dizaj SM, Sharifi S, Sedaghat H, Saffari M, Memar MY. Nano-scaffold-based delivery systems of antimicrobial agents in the treatment of osteomyelitis ; a narrative review. Heliyon 2024; 10:e38392. [PMID: 39559197 PMCID: PMC11570522 DOI: 10.1016/j.heliyon.2024.e38392] [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: 02/02/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 11/20/2024] Open
Abstract
Osteomyelitis caused by drug-resistant pathogens is one of the most important medical challenges due to high rates of mortality and morbidity, and limited therapeutical options. The application of novel nano-scaffolds loaded with antibiotics has widely been studied and extensively evaluated for in vitro and in vivo inhibition of pathogens, regenerating damaged bone tissue, and increasing bone cell proliferation. The treatment of bone infections using the local osteogenic scaffolds loaded with antimicrobial agents may efficiently overcome the problems of the systemic use of antimicrobial agents and provide a controlled release and sufficient local levels of antibiotics in the infected sites. The present study reviewed various nano-scaffolds delivery systems of antimicrobial drugs evaluated to treat osteomyelitis. Nano-scaffolds offer promising approaches because they simulate natural tissue regeneration in terms of their mechanical, structural, and sometimes chemical properties. The potential of several nano-scaffolds prepared by natural polymers such as silk, collagen, gelatin, fibrinogen, chitosan, cellulose, hyaluronic, alginate, and synthetic compounds such as polylactic acid, polyglycolic acid, poly (lactic acid-co-glycolic acid), poly-ɛ-caprolactone have been studied for usage as drug delivery systems of antimicrobial agents to treat osteomyelitis. In addition to incorporated antimicrobial agents and the content of scaffolds, the physical and chemical characteristics of the prepared delivery systems are a determining factor in their effectiveness in treating osteomyelitis.
Collapse
Affiliation(s)
- Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Sedaghat
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahmood Saffari
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Machado A, Gama M, Martins JA. Development of a dextrin-vitamin D3 micelle nanocarrier for the antimicrobial peptide LLKKK18 as a potential therapeutic agent for bone infections. J Mater Chem B 2024; 12:11464-11476. [PMID: 39392350 DOI: 10.1039/d4tb00903g] [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: 10/12/2024]
Abstract
In this work, an expedite synthesis was developed for a self-assembled micelle carrier for the antimicrobial peptide LL18. Covalent one-pot functionalization of dextrin with succinylated vitamin D3 and succinic anhydride produced an amphiphilic material that undergoes self-assembly into micelles in aqueous medium. Succinylated dextrin-vitamin D3 micelles were efficiently loaded with LL18 by electrostatic and hydrophobic interactions. Remarkably, the LL18-loaded micelle formulation dramatically improves the antibacterial activity of free LL18 against S. aureus, completely abrogates its severe hemolytic activity, redirects the internalization of LL18 from the perinuclear region of osteoblasts to the lysosomes and reduces cellular toxicity towards osteoblasts and macrophages. Overall, this work demonstrates that self-assembled micelle formulations based on dextrin, vitamin D3 and antimicrobial peptides, are promising platforms to develop multifunctional antibiotic-independent antimicrobial agents, not prone to the development of bacterial resistance, to treat bone infections.
Collapse
Affiliation(s)
- Alexandra Machado
- Centre of Biological Engineering, University of Minho (CEB), Campus de Gualtar, 4710-057 Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Miguel Gama
- Centre of Biological Engineering, University of Minho (CEB), Campus de Gualtar, 4710-057 Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - José Alberto Martins
- Center of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| |
Collapse
|
25
|
Gupta S, Qayoom I, Mairal A, Singh S, Kumar A. Local Delivery of Exosomes and Antibiotics in Hydroxyapatite-Based Nanocement for Osteomyelitis Management. ACS Infect Dis 2024; 10:3994-4008. [PMID: 39469832 DOI: 10.1021/acsinfecdis.4c00721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
The management of bone and joint infections is a formidable challenge in orthopedics and poses a global health concern. While clinical management emphasizes infection prevention and complete eradication, an effective strategy for stabilizing skeletal tissue with adequate soft tissue coverage remains limited. In this study, we have employed a novel approach of using the local delivery of exosomes and antibiotics (rifampicin) using a hydroxyapatite-based nanocement carrier to manage the residual space created during debridement effectively. Additionally, we synthesized a periosteum-guiding antioxidant herbal membrane to leverage the inherent periosteum regeneration capability of the bone, facilitating bone callus repair and natural healing. The synthesized scaffolds were biocompatible and demonstrated potent antibacterial activity in vitro. When evaluated in vivo in the Staphylococcus aureus-induced rat tibial osteomyelitis model, the released drugs successfully cleared the residual bacteria and the released exosome promoted bone healing, resulting in 3-fold increase in bone volume as analyzed via micro-CT analysis. Immunofluorescence staining of periosteum-specific markers also indicated the complete formation of periosteal layers, thus highlighting the complete bone repair.
Collapse
Affiliation(s)
- Sneha Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| | - Irfan Qayoom
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| | - Ayushi Mairal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| | - Sneha Singh
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Centre of Excellence in Orthopaedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
| |
Collapse
|
26
|
Yuan Y, Li H, Song Y, Zhang D, Wang Z, Yi X, Qi B, Zhang X, Jiang P, Yu A. Drug-Free "Triboelectric Immunotherapy" Activating Immunity for Osteomyelitis Treatment and Recurrence Prevention. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408473. [PMID: 39212208 DOI: 10.1002/adma.202408473] [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: 06/14/2024] [Revised: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Treatment of osteomyelitis is clinically challenging with low therapeutic efficacy and high risk of recurrence owing to the immunosuppressive microenvironment. Existing therapies are limited by drug concentration and single regulatory effect on the immune network, and emphasize the role of anti-inflammatory effects in reducing osteoclast rather than the role of proinflammatory effects in accelerating infection clearance, which is not conducive to complete bacteria elimination and recurrence prevention. Herein, a direct-current triboelectric nanogenerator (DC-TENG) is established to perform antibacterial effects and modulate immunological properties of infectious microenvironments of osteomyelitis through electrical stimulation, namely triboelectric immunotherapy. Seeing from the results, the triboelectric immunotherapy successfully activates polarization to proinflammatory (M1) macrophages in vitro, accompanied by satisfying direct antibacterial effects. The antibacterial and osteogenic abilities of triboelectric immunotherapy are verified in rat cranial osteomyelitis models. The effects on the polarization and differentiation of immune-related cells in vivo are investigated by establishing in situ tibial osteomyelitis models and immunosurveillance models in C57 mice respectively, indicating the ability of activating immunity and producing immunological memory for in situ infection and secondary recurrence, thus accelerating healing and preventing relapse. This study provides an efficient, long-acting, multifunctional, and wearable triboelectric immunotherapy strategy for drug-free osteomyelitis treatment systems.
Collapse
Affiliation(s)
- Ying Yuan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Haimei Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, P. R. China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Wuhan University, Wuhan, 430072, China
| | - Yuchen Song
- 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
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xinzeyu Yi
- 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
| | - Xianzheng Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, 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, P. R. 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
|
27
|
Jin L, Liu H, Wang C, Mao C, Wu S, Zhang Y, Li Z, Zhu S, Jiang H, Cui Z, Zheng Y, Liu X. Interface/Dipole Polarized Antibiotics-Loaded Fe 3O 4/PB Nanoparticles for Non-Invasive Therapy of Osteomyelitis Under Medical Microwave Irradiation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410917. [PMID: 39344940 DOI: 10.1002/adma.202410917] [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: 07/26/2024] [Revised: 09/12/2024] [Indexed: 10/01/2024]
Abstract
Due to their poor light penetration, photothermal therapy and photodynamic therapy are ineffective in treating deep tissue infections, such as osteomyelitis caused by Staphylococcus aureus (S. aureus). Here, a microwave (MW)-responsive magnetic targeting composite system consisting of ferric oxide (Fe3O4)/Prussian blue (PB) nanoparticles, gentamicin (Gent), and biodegradable poly(lactic-co-glycolic acid) (PLGA) is reported. The PLGA/Fe3O4/PB/Gent complex is used in combination with MW thermal therapy (MTT), MW dynamic therapy (MDT), and chemotherapy (CT) to treat acute osteomyelitis infected with S. aureus-infected. The powerful antibacterial effect of the PLGA/Fe3O4/PB/Gent is determined by the synergistic effects of heat and reactive oxygen species (ROS) generation by the Fe3O4/PB nanoparticles under MW irradiation and the effective release of Gent at the infection site via magnetic targeting. The antibacterial mechanism of the PLGA/Fe3O4/PB/Gent under MW irradiation is analyzed using bacterial transcriptome RNA sequencing. The MW heat and ROS reduce the activity of the protein transporters on the bacterial membrane, along with the transport of various ions and the acceleration of phosphate metabolism, which can lead to increased permeability of the bacterial membrane, damage the ribosome and DNA, and accompany the internal protein efflux of the bacteria, thus effectively killing the bacteria.
Collapse
Affiliation(s)
- Liguo Jin
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Hanpeng Liu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Chaofeng Wang
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Congyang Mao
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
| | - Shuilin Wu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Materials Science & Engineering, Peking University, Beijing, 100871, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhaoyang Li
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Shengli Zhu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Hui Jiang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Beijing, 100871, China
| | - Xiangmei Liu
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
- School of Materials Science & Engineering, Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
| |
Collapse
|
28
|
Park J, Lim Y, Park C, Kum KY, Yun CH, Park OJ, Han SH. Heat-killed Lancefieldella Rimae Induces Bone Resorption by Promoting Osteoclast Differentiation. J Endod 2024; 50:1593-1601. [PMID: 39182718 DOI: 10.1016/j.joen.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION Apical periodontitis, mainly caused by bacterial infection in the dental pulp, is often accompanied by abscess, periapical inflammation, and alveolar bone loss. Lancefieldella rimae has been detected in the root canals of patients with apical periodontitis. Here, we investigated whether L. rimae is associated with bone resorption. METHODS L. rimae was anaerobically cultured and heat-killed (HKLr). A mouse calvarial implantation model was used to determine the bone resorption in vivo. Committed osteoclasts prepared from C57BL/6 wild-type or Toll-like receptor 2 (TLR2)-deficient mice were differentiated into mature osteoclasts in the presence or absence of HKLr. The mRNA expression of tartrate-resistant acid phosphatase (TRAP), ATPase H+ transporting V0 subunit D2, cathepsin K, interleukin-6, tumor necrosis factor-α, and glyceraldehyde 3-phosphate dehydrogenase was quantified using real-time reverse transcription-polymerase chain reaction. The protein levels of c-Fos and NFATc1 were determined by Western blot analysis. RESULTS Implantation of HKLr onto the mouse calvaria induced the bone destruction with an increase of TRAP-positive areas. While HKLr enhanced the differentiation of osteoclasts, this effect was not observed in TLR2-deficient osteoclasts. HKLr dose-dependently increased the mRNA expression of genes associated with osteoclast differentiation including TRAP, ATPase H+ transporting V0 subunit D2, and cathepsin K. In addition, HKLr enhanced the expression of c-Fos and NFATc1, which are important transcription factors for osteoclast differentiation. Moreover, HKLr increased the expression of interleukin-6 and tumor necrosis factor-α. CONCLUSION L. rimae induces bone resorption by enhancing osteoclast differentiation through the TLR2 signaling pathway, implying that L. rimae is a causative agent responsible for the alveolar bone resorption accompanying apical periodontitis.
Collapse
Affiliation(s)
- Jinsung Park
- Department of Oral Microbiology and Immunology, and DRI, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Yeonjin Lim
- Department of Oral Microbiology and Immunology, and DRI, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Chaeyeon Park
- Department of Oral Microbiology and Immunology, and DRI, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Kee-Yeon Kum
- Department of Conservative Dentistry, DRI, Seoul National University Dental Hospital, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ok-Jin Park
- Department of Oral Microbiology and Immunology, and DRI, Seoul National University School of Dentistry, Seoul, Republic of Korea.
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and DRI, Seoul National University School of Dentistry, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Du J, Chu Y, Hu Y, Liu J, Liu H, Wang H, Yang C, Wang Z, Yu A, Ran J. A multifunctional self-reinforced injectable hydrogel for enhancing repair of infected bone defects by simultaneously targeting macrophages, bacteria, and bone marrow stromal cells. Acta Biomater 2024; 189:232-253. [PMID: 39396629 DOI: 10.1016/j.actbio.2024.10.014] [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: 07/02/2024] [Revised: 09/21/2024] [Accepted: 10/09/2024] [Indexed: 10/15/2024]
Abstract
Injectable hydrogels (IHs) have demonstrated huge potential in promoting repair of infected bone defects (IBDs), but how to endow them with desired anti-bacterial, immunoregulatory, and osteo-inductive properties as well as avoid mechanical failure during their manipulation are challenging. In this regard, we developed a multifunctional AOHA-RA/Lap nanocomposite IH for IBDs repair, which was constructed mainly through two kinds of reversible cross-links: (i) the laponite (Lap) crystals mediated electrostatic interactions; (ii) the phenylboronic acid easter bonds between the 4-aminobenzeneboronic acid grafted oxidized hyaluronic acid (AOHA) and rosmarinic acid (RA). Due to the specific structural composition, the AOHA-RA/Lap IH demonstrated superior injectability, self-recoverability, spatial adaptation, and self-reinforced mechanical properties after being injected to the bone defect site. In addition, the RA molecules could be locally released from the hydrogel following a Weibull model for over 10 days. Systematic in vitro/vivo assays proved the strong anti-bacterial activity of the hydrogel against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli). Moreover, its capability of inducing M2 polarization of macrophages (Mφ) and osteogenic differentiation of bone marrow stromal cells (BMSCs) was verified either, and the mechanism of the former was identified to be related to the JAK1-STAT1 and PI3K-AKT signaling pathways and that of the latter was identified to be related to the calcium signaling pathway, extracellular matrix (ECM) receptor interaction and TGF-β signaling pathway. After being implanted to a S. aureus infected rat skull defect model, the AOHA-RA/Lap IH significantly accelerated repair of IBDs without causing significant systemic toxicity. STATEMENT OF SIGNIFICANCE: Rosmarinic acid and laponite were utilized to develop an injectable hydrogel, promising for accelerating repair of infected bone defects in clinic. The gelation of the hydrogel was completely driven by two kinds of reversible cross-links, which endow the hydrogel superior spatial adaption, self-recoverability, and structural stability. The as-prepared hydrogel demonstrated superior anti-bacterial/anti-biofilm activity and could induce M2 polarization of macrophages and osteogenic differentiation of BMSCs. The mechanism behind macrophages polarization was identified to be related to the JAK1-STAT1 and PI3K-AKT signaling pathways. The mechanism behind osteogenic differentiation of BMSCs was identified to be related to the ECM receptor interaction and calcium signaling/TGF-β signaling pathways.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Yan Hu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan, 430000, China
| | - Jin Liu
- Hubei Engineering & Technology Research Center for Functional Materials from Biomass, School of Chemistry and Material Science, Hubei Engineering University, Xiaogan, Hubei 432000, 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
| | - 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
| | - 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
| | - Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan, 430000, China.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan, 430000, 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.
| |
Collapse
|
30
|
Ge M, Ruan Z, Zhu YX, Wu W, Yang C, Lin H, Shi J. A natural killer cell mimic against intracellular pathogen infections. SCIENCE ADVANCES 2024; 10:eadp3976. [PMID: 39475620 PMCID: PMC11524181 DOI: 10.1126/sciadv.adp3976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024]
Abstract
In the competition between the pathogen infection and the host defense, infectious microorganisms may enter the host cells by evading host defense mechanisms and use the intracellular biomolecules as replication nutrient. Among them, intracellular Staphylococcus aureus relies on the host cells to protect itself from the attacks by antibiotics or immune system to achieve long-term colonization in the host, and the consequent clinical therapeutic failures and relapses after antibiotic treatment. Here, we demonstrate that intracellular S. aureus surviving well even in the presence of vancomycin can be effectively eliminated using an emerging cell-mimicking therapeutic strategy. These cell mimics with natural killer cell-like activity (NKMs) are composed of a redox-responsive degradable carrier, and perforin and granzyme B within the carrier. NKMs perform far more effectivly than clinical antibiotics in treating intracellular bacterial infections, providing a direct evidence of the NK cell-mimicking immune mechanism in the treatment of intracellular S. aureus.
Collapse
Affiliation(s)
- Min Ge
- 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, P. R. China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Zesong Ruan
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P. R. China
| | - Ya-Xuan Zhu
- 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, P. R. China
| | - Wencheng Wu
- 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, P. R. China
| | - Chuang Yang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P. R. 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, P. R. China
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. 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, P. R. China
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
| |
Collapse
|
31
|
Adouane M, Kadri N, Benzitoune N, Lakhdari C, Djellal S, Ousmer L, Tahraoui H, Amrane A, Remini H, Dahmoune F, Madani K. Understanding bacterial diversity, infection dynamics, prevention of antibiotic resistance: an integrated study in an Algerian hospital context. Eur J Clin Microbiol Infect Dis 2024; 43:2093-2105. [PMID: 39136832 DOI: 10.1007/s10096-024-04919-3] [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: 05/26/2024] [Accepted: 07/29/2024] [Indexed: 11/05/2024]
Abstract
PURPOSE Bacterial infections, particularly bacteremia, urinary tract infections (UTIs), and pus infections, remain among hospitals' most worrying medical problems. This study aimed to explore bacterial diversity, infection dynamics, and antibiotic resistance profiles of bacterial isolates. METHODS We analyzed data from 1750 outpatients and 920 inpatients, of whom 1.6% and 8.47% respectively had various bacterial infections. RESULTS The analysis revealed that UTIs were the most prevalent at 41.01%, particularly affecting women. UTIs also showed a distinct distribution across admission departments, notably in emergency (23.07%) and pediatric (14.10%) units. The most frequently isolated microorganisms were Escherichia coli (E. coli), followed by Klebsiella ornithinolytica. Skin infections followed UTIs, accounting for 35.88% of cases, more prevalent in men, with Staphylococcus aureus (S. aureus) being the primary pathogen (57%). Gram-negative bacteria (GNB) like E. coli and Pseudomonas aeruginosa contributed significantly to skin infections (43%). Bacteremia cases constituted 11.52% of bacterial infections, predominantly affecting women (67%) and linked to GNB (78%). A comparative study of antibiotic susceptibility profiles revealed more pronounced resistance in GNB strains isolated from inpatients, particularly to antibiotics such as Amoxicillin/clavulanic acid, Tetracyclin, Gentamicin, Chloramphenicol, and Ampicillin. In contrast, strains from ambulatory patients showed greater resistance to Colistin. Gram-positive bacteria from hospitalized patients showed higher resistance to quinolones and cephalosporins, while ambulatory strains showed high resistance to aminoglycosides, macrolides, fluoroquinolones, and penicillin. Furthermore, these analyses identified the most effective antibiotics for the empirical treatment of both community-acquired and nosocomial infections. Ciprofloxacin, aztreonam, and amikacin exhibited low resistance rates among GNB, with gentamicin and chloramphenicol being particularly effective for community-acquired strains. For S. aureus, ciprofloxacin, rifampicin, and cefoxitin were especially effective, with vancomycin showing high efficacy against community-acquired isolates and fosfomycin and chloramphenicol being effective for hospital-acquired strains. CONCLUSION These results are essential for guiding antibiotic therapy and improving clinical outcomes, thus contributing to precision medicine and antimicrobial stewardship efforts.
Collapse
Affiliation(s)
- Meriem Adouane
- Laboratoire de Gestion et Valorisation des Ressources Naturelles et Assurance Qualité (LGVRNAQ), Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
- Département de Biologie, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
| | - Nabil Kadri
- Département de Biologie, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
- Laboratoire de Biomathématiques, Biophysique, Biochimie, et Scientométrie (L3BS), Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, 06000, Algeria
| | - Nourelimane Benzitoune
- Laboratoire de Gestion et Valorisation des Ressources Naturelles et Assurance Qualité (LGVRNAQ), Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
- Département de Biologie, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
| | - Chafika Lakhdari
- Laboratoire de Gestion et Valorisation des Ressources Naturelles et Assurance Qualité (LGVRNAQ), Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
- Département de Biologie, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
| | - Samia Djellal
- Laboratoire de Gestion et Valorisation des Ressources Naturelles et Assurance Qualité (LGVRNAQ), Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
- Département de Biologie, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
| | - Lilla Ousmer
- Laboratoire de Gestion et Valorisation des Ressources Naturelles et Assurance Qualité (LGVRNAQ), Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
- Département de Biologie, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
| | - Hichem Tahraoui
- Laboratoire de Génie des Procédés Chimiques, Department of process engineering, University of Ferhat Abbas, Setif, Algeria
- Univ Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR-UMR6226, Rennes, 35000, France
| | - Abdeltif Amrane
- Univ Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR-UMR6226, Rennes, 35000, France.
| | - Hocine Remini
- Département de Biologie, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
- Laboratoire de Biomathématiques, Biophysique, Biochimie, et Scientométrie (L3BS), Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, 06000, Algeria
- Centre de Recherche en Technologies Agroalimentaires, Route de Targa Ouzemmour, Campus Universitaire, Bejaia, 06000, Algeria
| | - Farid Dahmoune
- Département de Biologie, Faculté des Sciences de la Nature et de la Vie et des Sciences de la Terre, Université de Bouira, Bouira, 10000, Algeria
- Laboratoire de Biomathématiques, Biophysique, Biochimie, et Scientométrie (L3BS), Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, 06000, Algeria
| | - Khodir Madani
- Laboratoire de Biomathématiques, Biophysique, Biochimie, et Scientométrie (L3BS), Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, 06000, Algeria
- Centre de Recherche en Technologies Agroalimentaires, Route de Targa Ouzemmour, Campus Universitaire, Bejaia, 06000, Algeria
| |
Collapse
|
32
|
Li S, Xu G, Guo Z, Liu Y, Ouyang Z, Li Y, Huang Y, Sun Q, Giri BR, Fu Q. Deficiency of hasB accelerated the clearance of Streptococcus equi subsp. Zooepidemicus through gasdermin d-dependent neutrophil extracellular traps. Int Immunopharmacol 2024; 140:112829. [PMID: 39083933 DOI: 10.1016/j.intimp.2024.112829] [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: 04/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Streptococcus equi subsp. zooepidemicus (S. zooepidemicus, SEZ) is an essential zoonotic bacterial pathogen that can cause various inflammation, such as meningitis, endocarditis, and pneumonia. UDP-glucose dehydrogenase (hasB) is indispensable in synthesizing SEZ virulence factor hyaluronan capsules. Our study investigated the infection of hasB on mice response to SEZ by employing a constructed capsule-deficient mutant strain designated as the ΔhasB strain. This deficiency was associated with a reduced SEZ bacterial load in the mice's blood and peritoneal lavage fluid (PLF) post-infection. Besides, the ΔhasB SEZ strain exhibited a higher propensity for neutrophil infiltration and release of cell-free DNA (cfDNA) in vivo compared to the wild-type (WT) SEZ strain. In vitro experiments further revealed that ΔhasB SEZ more effectively induced the formation of neutrophil extracellular traps (NETs) containing histone 3 (H3), neutrophil elastase (NE), and DNA, than its WT counterpart. Moreover, the release of NETs was determined to be gasdermin D (GSDMD)-dependent during the infection process. Taken together, these findings underscore that the deficiency of the hasB gene in SEZ leads to enhanced GSDMD-dependent NET release from neutrophils, thereby reducing SEZ's capacity to resist NETs-mediated eradication during infection. Our finding paves the way for the development of innovative therapeutic strategies against SEZ.
Collapse
Affiliation(s)
- Shun Li
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Guobin Xu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zheng Guo
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Yuxuan Liu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zhiliang Ouyang
- Houjie Town Agricultural Technology Service Center, Dongguan, Guangdong, China
| | - Yajuan Li
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Yunfei Huang
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Qinqin Sun
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Bikash R Giri
- Department of Zoology, K.K.S. Women's College, Balasore, Odisha, India
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China.
| |
Collapse
|
33
|
Yin C, Deng M, Yu J, Chen Y, Zheng K, Huang Y, Deng X, Tian Y, Ma Y, Zeng B, Guo X, Guo B. An Andrias davidianus derived composite hydrogel with enhanced antibacterial and bone repair properties for osteomyelitis treatment. Sci Rep 2024; 14:24626. [PMID: 39426986 PMCID: PMC11490572 DOI: 10.1038/s41598-024-75997-8] [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: 05/02/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Effective antibacterial therapy while accelerating the repair of bone defects is crucial for the treatment of osteomyelitis. Inspired by the protective mechanism of Andrias davidianus, we constructed an antibacterial hydrogel scaffold with excellent rigidity and long-term slow-release activity. While retaining the toughness of the skin secretion of Andrias davidianus (SSAD), the rigidity of the hydrogel material is increased by incorporating hydroxyapatite to meet the demands of bone-defect-filling materials. It also exerted antibacterial effects via the slow-release of vancomycin from local osteomyelitis lesions. Notably, the hydrogel can also carry a high stable recombinant miR-214-3p inhibitor (MSA-anti214). By the delivery of nano vector polyvinylamine, the long-term slow-release of MSA-anti214 is achieved to promote bone repair, making this composite hydrogel a potential SSAD-based osteomyelitis alleviator (SOA). In vitro and vivo results verified that the SOA effectively eliminated Staphylococcus aureus and repaired bone defects, ultimately mitigating the progression of osteomyelitis. This composite hydrogel extends the economic application prospects of A. davidianus and has provided new insights for the treatment of osteomyelitis. The study also explored new insights for the bone filling materials of bone defection and other skeletal system diseases.
Collapse
Affiliation(s)
- Chong Yin
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Meng Deng
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Jinshu Yu
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Yonghao Chen
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Kaiyuan Zheng
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Yi Huang
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xudong Deng
- Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Ye Tian
- Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Yuwen Ma
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Beilei Zeng
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xiaolan Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
| | - Bin Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
| |
Collapse
|
34
|
Guo J, Shu X, Yu S, Guo C, Shen G, Chen L, Zhou J, Xiao J, Guo H, Chen Y, Zeng Z, Wang P. Injectable hydrogel microsphere-bomb for MRSA-infected chronic osteomyelitis. J Control Release 2024; 376:337-353. [PMID: 39413850 DOI: 10.1016/j.jconrel.2024.10.021] [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: 06/19/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Biofilm and bone tissue defect induced by the bacterial infection severely impede chronic osteomyelitis treatment. It is critical to break though the densely and obstinate biofilm so that the target drugs can deliver to the infected bone more effectively. Herein, an acoustically responsive multifunctional hydrogel microsphere-bomb (EMgel) was designed and prepared by microfluidic technology, which could be injected to the focus of bone infection, and blasted into the nidus deeply to destroy the bacterial biofilm matrix barrier under penetrating ultrasound, so the encapsulated natural polyphenolic EGCG and bioactive MoS2 released to repair the damaged bone. The results proved the hydrogel microsphere-bomb exhibited controlling drug release, favorable antibacterial (as high as 99 %), high biofilm resistance, fascinating antioxidation, good cytocompatibility, and osteogenic differentiation. The acoustically responsive microsphere-bomb further proved their fantastic ability to eradicate biofilm and promote bone regeneration in the Methicillin-resistant Staphylococcus aureus (MRSA) infected chronic osteomyelitis model due to the synergy effects of EGCG and bioactive MoS2. Especially, immunohistochemical staining showed lower inflammatory reaction and higher expression of OCN in EMgel group treated with ultrasound wave. This study presents a new design of hydrogel microsphere-based intelligence drug delivery for osteomyelitis treatment, which exhibit great promising potential for dealing with chronic orthopedic infections, drug delivery system and tissue engineering.
Collapse
Affiliation(s)
- Jiayi Guo
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Xian Shu
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Shan Yu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Cuiping Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Guangxin Shen
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Guangdong Province, Foshan 528031, China
| | - Longsheng Chen
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Jiayi Zhou
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Jiangwei Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Huilong Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Yi Chen
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Zhiwen Zeng
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China.
| | - Ping Wang
- Department of Ultrasound, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
35
|
Good CJ, Butrico CE, Colley ME, Emmerson LN, Gibson-Corley KN, Cassat JE, Spraggins JM, Caprioli RM. Uncovering lipid dynamics in Staphylococcus aureus osteomyelitis using multimodal imaging mass spectrometry. Cell Chem Biol 2024; 31:1852-1868.e5. [PMID: 39389064 DOI: 10.1016/j.chembiol.2024.09.005] [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: 01/19/2024] [Revised: 06/20/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
Osteomyelitis occurs when Staphylococcus aureus invades the bone microenvironment, resulting in a bone marrow abscess with a spatially defined architecture of cells and biomolecules. Imaging mass spectrometry and microscopy are tools that can be employed to interrogate the lipidome of S. aureus-infected murine femurs and reveal metabolic and signaling consequences of infection. Here, nearly 250 lipids were spatially mapped to healthy and infection-associated morphological features throughout the femur, establishing composition profiles for tissue types. Ether lipids and arachidonoyl lipids were altered between cells and tissue structures in abscesses, suggesting their roles in abscess formation and inflammatory signaling. Sterols, triglycerides, bis(monoacylglycero)phosphates, and gangliosides possessed ring-like distributions throughout the abscess, suggesting a hypothesized dysregulation of lipid metabolism in a population of cells that cannot be discerned with traditional microscopy. These data provide insight into the signaling function and metabolism of cells in the fibrotic border of abscesses, likely characteristic of lipid-laden macrophages.
Collapse
Affiliation(s)
- Christopher J Good
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Casey E Butrico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Madeline E Colley
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Lauren N Emmerson
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37235, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA.
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
36
|
Nasr J, Abdessamad H, Mina J, Haykal T, Jamil Y, Abboud E, Mahdi A, Asmar R, Abi Assaad R, Alameddine D, Bourji A, Mahdi M, Abdulaal R, Tomassian S, El Ahmadieh H, Azzam W, Mokhbat JE, Moghnieh R, Rodriguez-Morales AJ, Husni R. The epidemiology of gram-negative bacteremia in Lebanon: a study in four hospitals. Ann Clin Microbiol Antimicrob 2024; 23:90. [PMID: 39385237 PMCID: PMC11465513 DOI: 10.1186/s12941-024-00740-0] [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: 03/05/2024] [Accepted: 08/16/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Gram-negative bacteremia is a life-threatening infection with high morbidity and mortality. Its incidence is rising worldwide, and treatment has become more challenging due to emerging bacterial resistance. Little data is available on the burden and outcome of such infections in Lebanon. METHODS We conducted this retrospective study in four Lebanese hospitals. Data on medical conditions and demographics of 2400 patients diagnosed with a bloodstream infection based on a positive blood culture were collected between January 2014 and December 2020. RESULTS Most bacteremias were caused by Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter baumannii, with the more resistant organisms being hospital-acquired. Third-generation cephalosporin and quinolone resistance was steady throughout the study, but carbapenem resistance increased. Mortality with such infections is high, but carbapenem resistance or infection with Pseudomonas or Acinetobacter species were significant risk factors for poor outcomes. CONCLUSION This is the first multi-center study from Lebanon on gram-negative bacteremia, resistance patterns, and factors associated with a poor outcome. More surveillance is needed to provide data to guide empirical treatment for bacteremia in Lebanon.
Collapse
Affiliation(s)
- Janane Nasr
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Hilal Abdessamad
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Johnathan Mina
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Tony Haykal
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Yasser Jamil
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Emma Abboud
- Laboratory Director, Mount Lebanon Hospital University Medical Center, Beirut, 1102, Lebanon
| | - Ahmad Mahdi
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rana Asmar
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rawad Abi Assaad
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Dana Alameddine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Alaa Bourji
- Department of Surgery, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Mahmoud Mahdi
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Razan Abdulaal
- Department of Internal Medicine, University of Balamand, Balamand, Lebanon
| | - Serge Tomassian
- Department of Internal Medicine, University of Balamand, Balamand, Lebanon
| | - Hanane El Ahmadieh
- Infection Control Coordination, Mount Lebanon Hospital University Medical Center, Beirut, 1102, Lebanon
| | - Wael Azzam
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Jacques E Mokhbat
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rima Moghnieh
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Alfonso J Rodriguez-Morales
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
- Master of Clinical Epidemiology and Biostatistics, Universidad Cientifica del Sur, Lima, 15067, Peru
| | - Rola Husni
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon.
| |
Collapse
|
37
|
Zelmer AR, Yang D, Gunn NJ, Solomon LB, Nelson R, Kidd SP, Richter K, Atkins GJ. Osteomyelitis-relevant antibiotics at clinical concentrations show limited effectivity against acute and chronic intracellular S. aureus infections in osteocytes. Antimicrob Agents Chemother 2024; 68:e0080824. [PMID: 39194210 PMCID: PMC11459924 DOI: 10.1128/aac.00808-24] [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: 06/02/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
Osteomyelitis caused by Staphylococcus aureus can involve the persistent infection of osteocytes. We sought to determine if current clinically utilized antibiotics were capable of clearing an intracellular osteocyte S. aureus infection. Rifampicin, vancomycin, levofloxacin, ofloxacin, amoxicillin, oxacillin, doxycycline, linezolid, gentamicin, and tigecycline were assessed for their minimum inhibitory concentration (MIC) and minimum bactericidal concentrations against 12 S. aureus strains, at pH 5.0 and 7.2 to mimic lysosomal and cytoplasmic environments, respectively. Those antibiotics whose bone estimated achievable concentration was commonly above their respective MIC for the strains tested were further assayed in a human osteocyte infection model under acute and chronic conditions. Osteocyte-like cells were treated at 1×, 4×, and 10× the MIC for 1 and 7 days following infection (acute model), or at 15 and 21 days of infection (chronic model). The intracellular effectivity of each antibiotic was measured in terms of CFU reduction, small colony variant formation, and bacterial mRNA expression change. Only rifampicin, levofloxacin, and linezolid reduced intracellular CFU numbers significantly in the acute model. Consistent with the transition to a non-culturable state, few if any CFU could be recovered from the chronic model. However, no treatment in either model reduced the quantity of bacterial mRNA or prevented non-culturable bacteria from returning to a culturable state. These findings indicate that S. aureus adapts phenotypically during intracellular infection of osteocytes, adopting a reversible quiescent state that is protected against antibiotics, even at 10× their MIC. Thus, new therapeutic approaches are necessary to cure S. aureus intracellular infections in osteomyelitis.
Collapse
Affiliation(s)
- Anja R. Zelmer
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Dongqing Yang
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Nicholas J. Gunn
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - L. Bogdan Solomon
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Department of Orthopedics and Trauma, Royal Adelaide Hospital, Adelaide, Australia
| | - Renjy Nelson
- Department of Infectious Diseases, Central Adelaide Local Health Network, Adelaide, Australia
| | - Stephen P. Kidd
- Australian Center for Antimicrobial Resistance Ecology, University of Adelaide, Adelaide, Australia
- Research Center for Infectious Disease, School of Biological Science, University of Adelaide, Adelaide, Australia
| | - Katharina Richter
- Department of Surgery, Richter Lab, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, Australia
| | - Gerald J. Atkins
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
38
|
Chang J, Kerr D, Zheng M, Seyler T. Chondrocyte Invasion May Be a Mechanism for Persistent Staphylococcus Aureus Infection In Vitro. Clin Orthop Relat Res 2024; 482:1839-1847. [PMID: 38662927 PMCID: PMC11419450 DOI: 10.1097/corr.0000000000003074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/11/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Recurrent bone and joint infection with Staphylococcus aureus is common. S. aureus can invade and persist in osteoblasts and fibroblasts, but little is known about this mechanism in chondrocytes. If S. aureus were able to invade and persist within chondrocytes, this could be a difficult compartment to treat. QUESTION/PURPOSE Can S. aureus infiltrate and persist intracellularly within chondrocytes in vitro? METHODS Cell lines were cultured in vitro and infected with S. aureus. Human chondrocytes (C20A4) were compared with positive controls of human osteoblasts (MG63) and mouse fibroblasts (NIH3T3), which have previously demonstrated S. aureus invasion and persistence (human fibroblasts were not available to us). Six replicates per cell type were followed for 6 days after infection. Cells were treated daily with antibiotic media for extracellular killing. To determine whether S. aureus can infiltrate chondrocytes, fluorescence microscopy was performed to qualitatively assess the presence of intracellular bacteria, and intracellular colony-forming units (CFU) were enumerated 2 hours after infection. To determine whether S. aureus can persist within chondrocytes, intracellular CFUs were enumerated from infected host cells each day postinfection. RESULTS S. aureus invaded human chondrocytes (C20A4) at a level (2.8 x 10 5 ± 5.5 x 10 4 CFUs/mL) greater than positive controls of human osteoblasts (MG63) (9.5 x 10 2 ± 2.5 x 10 2 CFUs/mL; p = 0.01) and mouse fibroblasts (NIH3T3) (9.1 x 10 4 ± 2.5 x 10 4 CFUs/mL; p = 0.02). S. aureus also persisted within human chondrocytes (C20A4) for 6 days at a level (1.4 x 10 3 ± 5.3 x 10 2 CFUs/mL) greater than that of human osteoblasts (MG63) (4.3 x 10 2 ± 3.5 x 10 1 CFUs/mL; p = 0.02) and mouse fibroblasts (NIH3T3) (0 CFUs/mL; p < 0.01). S. aureus was undetectable within mouse fibroblasts (NIH3T3) after 4 days. There were 0 CFUs yielded from cell media, confirming extracellular antibiotic treatment was effective. CONCLUSION S. aureus readily invaded human chondrocytes (C20A4) in vitro and persisted viably for 6 days after infection, evading extracellular antibiotics. Chondrocytes demonstrated a greater level of intracellular invasion and persistence by S. aureus than positive control human osteoblast (MG63) and mouse fibroblast (NIH3T3) cell lines. CLINICAL RELEVANCE Chondrocyte invasion and persistence may contribute to recurrent bone and joint infections. Additional research should assess longer periods of persistence and whether this mechanism is present in vivo.
Collapse
Affiliation(s)
- Jerry Chang
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - David Kerr
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Megan Zheng
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Thorsten Seyler
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
39
|
Hu X, Chen J, Yang S, Zhang Z, Wu H, He J, Qin L, Cao J, Xiong C, Li K, Liu X, Qian Z. 3D Printed Multifunctional Biomimetic Bone Scaffold Combined with TP-Mg Nanoparticles for the Infectious Bone Defects Repair. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403681. [PMID: 38804867 DOI: 10.1002/smll.202403681] [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: 05/07/2024] [Revised: 05/18/2024] [Indexed: 05/29/2024]
Abstract
Infected bone defects are one of the most challenging problems in the treatment of bone defects due to the high antibiotic failure rate and the lack of ideal bone grafts. In this paper, inspired by clinical bone cement filling treatment, α-c phosphate (α-TCP) with self-curing properties is composited with β-tricalcium phosphate (β-TCP) and constructed a bionic cancellous bone scaffolding system α/β-tricalcium phosphate (α/β-TCP) by low-temperature 3D printing, and gelatin is preserved inside the scaffolds as an organic phase, and later loaded with a metal-polyphenol network structure of tea polyphenol-magnesium (TP-Mg) nanoparticles. The scaffolds mimic the structure and components of cancellous bone with high mechanical strength (>100 MPa) based on α-TCP self-curing properties through low-temperature 3D printing. Meanwhile, the scaffolds loaded with TP-Mg exhibit significant inhibition of Staphylococcus aureus (S.aureus) and promote the transition of macrophages from M1 pro-inflammatory to M2 anti-inflammatory phenotype. In addition, the composite scaffold also exhibits excellent bone-enhancing effects based on the synergistic effect of Mg2+ and Ca2+. In this study, a multifunctional ceramic scaffold (α/β-TCP@TP-Mg) that integrates anti-inflammatory, antibacterial, and osteoinduction is constructed, which promotes late bone regenerative healing while modulating the early microenvironment of infected bone defects, has a promising application in the treatment of infected bone defects.
Collapse
Affiliation(s)
- Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan, 610081, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiao Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shuhao Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Zhen Zhang
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Haoming Wu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan, 610081, China
| | - Jian He
- College of Medical, Henan University of Science and Technology, Luoyang, 471023, China
| | - Leilei Qin
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Jianfei Cao
- School of Materials and Environmental Engineering, Chengdu Technological University, Chengdu, Sichuan, 611730, China
| | - Chengdong Xiong
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Kainan Li
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan, 610081, China
| | - Xian Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
40
|
Liu M, Li X, Li Y, Zou Y. Insights into the airborne microorganisms in a Sichuan south-road dark tea pile fermentation plant during production. Front Microbiol 2024; 15:1439133. [PMID: 39286348 PMCID: PMC11402737 DOI: 10.3389/fmicb.2024.1439133] [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: 05/28/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Sichuan south-road dark tea (SSDT) is generally produced through a series of processes, including fixing, rolling, pile fermentation, and drying, with microbial action during pile fermentation playing a crucial role in determining tea quality. The air within the SSDT pile fermentation plant (SSDTPP) is considered an important source of these microbes, but research in this area has been limited. Methods In this study, air samples from SSDTPP were collected on the 1st (SSDT1), 12th (SSDT2), and 24th (SSDT3) days of pile fermentation and comprehensively analyzed by high-throughput sequencing. Results and discussion The results revealed the presence of 2 and 24 phyla, 9 and 49 classes, 18 and 88 orders, 28 and 153 families, 38 and 253 genera, and 47 and 90 species of fungi and bacteria, respectively, across all samples. SSDT1 and SSDT2 individually had the highest fungal and bacterial diversity, while Aspergillus was the dominant genus throughout the pile fermentation with an abundance of 34.6%, 91.17%, and 67.86% in SSDT1, SSDT2, and SSDT3, respectively. Microbial populations in SSDT1 were predominantly involved in xenobiotic biodegradation and metabolism, amino acid metabolism, the biosynthesis of other secondary metabolites, etc. However, SSDT2 exhibited a higher prevalence of human disease-related functions. SSDT3 primarily focused on the metabolism of other amino acids and carbohydrate metabolism. Additionally, 104 genera and 22 species coexisted in both SSDTPP air and piled SSDT, suggesting that frequent microbial exchange may occur between them. These findings pave the way for microbial traceability during SSDT production and provide a foundation for further functional microbial research.
Collapse
Affiliation(s)
- Miaoyi Liu
- Department of Tea Science, College of Horticulture, Sichuan Agricultural University, Chengdu, China
| | - Xian Li
- Department of Tea Science, College of Horticulture, Sichuan Agricultural University, Chengdu, China
| | - Yimiao Li
- Department of Tea Science, College of Horticulture, Sichuan Agricultural University, Chengdu, China
| | - Yao Zou
- Department of Tea Science, College of Horticulture, Sichuan Agricultural University, Chengdu, China
- Tea Refining and Innovation Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
41
|
Guo Y, Mao C, Wu S, Wang C, Zheng Y, Liu X. Ultrasound-Triggered Piezoelectric Catalysis of Zinc Oxide@Glucose Derived Carbon Spheres for the Treatment of MRSA Infected Osteomyelitis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400732. [PMID: 38764258 DOI: 10.1002/smll.202400732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/28/2024] [Indexed: 05/21/2024]
Abstract
Currently, methicillin-resistant Staphylococcus aureus (MRSA)-induced osteomyelitis is a clinically life-threatening disease, however, long-term antibiotic treatment can lead to bacterial resistance, posing a huge challenge to treatment and public health. In this study, glucose-derived carbon spheres loaded with zinc oxide (ZnO@HTCS) are successfully constructed. This composite demonstrates the robust ability to generate reactive oxygen species (ROS) under ultrasound (US) irradiation, eradicating 99.788% ± 0.087% of MRSA within 15 min and effectively treating MRSA-induced osteomyelitis infection. Piezoelectric force microscopy tests and finite element method simulations reveal that the ZnO@HTCS composite exhibits superior piezoelectric catalytic performance compared to pure ZnO, making it a unique piezoelectric sonosensitizer. Density functional theory calculations reveal that the formation of a Mott-Schottky heterojunction and an internal piezoelectric field within the interface accelerates the electron transfer and the separation of electron-hole pairs. Concurrently, surface vacancies of the composite enable the adsorption of a greater amount of oxygen, enhancing the piezoelectric catalytic effect and generating a substantial quantity of ROS. This work not only presents a promising approach for augmenting piezoelectric catalysis through construction of a Schottky heterojunction interface but also provides a novel, efficient therapeutic strategy for treating osteomyelitis.
Collapse
Affiliation(s)
- Yihao Guo
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Congyang Mao
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
| | - Shuilin Wu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Chaofeng Wang
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Xiangmei Liu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
| |
Collapse
|
42
|
Di Matteo V, Di Filippo MF, Ballarin B, Bonvicini F, Iaquinta MR, Panzavolta S, Mazzoni E, Cassani MC. Porous titanium scaffolds modified with Zeolitic Imidazolate Framework (ZIF-8) with enhanced osteogenic activity for the prevention of implant-associated infections. Front Chem 2024; 12:1452670. [PMID: 39268004 PMCID: PMC11390653 DOI: 10.3389/fchem.2024.1452670] [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: 06/21/2024] [Accepted: 07/26/2024] [Indexed: 09/15/2024] Open
Abstract
In this study, zeolitic imidazolate framework 8 (ZIF-8) was coated on porous Ti6Al4V scaffolds, either bare or previously modified using hydroxyapatite (HA) or HA and gelatin (HAgel), via a growing single-step method in aqueous media using two contact times at 6 h and 24 h. The coated scaffolds termed ZIF-8@Ti, ZIF-8@HA/Ti, and ZIF-8@HAgel/Ti were characterized via scanning electron microscopy (SEM), powder X-ray diffraction (PXRD), attenuated total reflectance-Fourier transform infrared (ATR-FTIR), and molecular plasma-atomic emission spectroscopy (MP-AES). In order to assess the cell proliferation rate, the cytocompatibility of the scaffolds was evaluated in primary osteoblasts (hOBs) using alamarBlue assay, while the osteoconductivity was analyzed in hOBs using a real-time approach, evaluating the expression of secreted phosphoprotein 1 (SPP1). Osteopontin, which is the protein encoded by this gene, represents the major non-collagenous bone protein that binds tightly to HA. The scaffolds were shown to be non-cytotoxic based on hOB proliferation at all time points of analysis (24 h and 72 h). In hOB cultures, the scaffolds induced the upregulation of SPP1 with different fold changes. Some selected scaffolds were assayed in vitro for their antibacterial potential against Staphylococcus epidermidis; the scaffolds coated with ZIF-8 crystals, regardless of the presence of HA and gelatin, strongly inhibited bacterial adhesion to the materials and reduced bacterial proliferation in the culture medium, demonstrating the suitable release of ZIF-8 in a bioactive form. These experiments suggest that the innovative scaffolds, tested herein, provide a good microenvironment for hOB adhesion, viability, and osteoconduction with effective prevention of S. epidermidis adhesion.
Collapse
Affiliation(s)
- Valentina Di Matteo
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Bologna, Italy
| | | | - Barbara Ballarin
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Bologna, Italy
| | - Francesca Bonvicini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Maria Rosa Iaquinta
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Silvia Panzavolta
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Elisa Mazzoni
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Maria Cristina Cassani
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Bologna, Italy
| |
Collapse
|
43
|
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 2024:10.1007/s00210-024-03357-4. [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] [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
|
44
|
Hu H, Ding H, Lyu J, Chen Y, Huang C, Zhang C, Li W, Fang X, Zhang W. Detection of rare microorganisms in bone and joint infections by metagenomic next-generation sequencing. Bone Joint Res 2024; 13:401-410. [PMID: 39142657 PMCID: PMC11324352 DOI: 10.1302/2046-3758.138.bjr-2023-0420.r1] [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] [Indexed: 08/16/2024] Open
Abstract
Aims This aim of this study was to analyze the detection rate of rare pathogens in bone and joint infections (BJIs) using metagenomic next-generation sequencing (mNGS), and the impact of mNGS on clinical diagnosis and treatment. Methods A retrospective analysis was conducted on 235 patients with BJIs who were treated at our hospital between January 2015 and December 2021. Patients were divided into the no-mNGS group (microbial culture only) and the mNGS group (mNGS testing and microbial culture) based on whether mNGS testing was used or not. Results A total of 147 patients were included in the no-mNGS group and 88 in the mNGS group. The mNGS group had a higher detection rate of rare pathogens than the no-mNGS group (21.6% vs 10.2%, p = 0.016). However, the mNGS group had lower rates of antibiotic-related complications, shorter hospital stays, and higher infection control rates compared with the no-mNGS group (p = 0.017, p = 0.003, and p = 0.028, respectively), while there was no significant difference in the duration of antibiotic use (p = 0.957). In culture-negative cases, the mNGS group had lower rates of antibiotic-related complications, shorter hospital stays, and a higher infection control rate than the no-mNGS group (p = 0.036, p = 0.033, p = 0.022, respectively), while there was no significant difference in the duration of antibiotic use (p = 0.748). Conclusion mNGS improves detection of rare pathogens in BJIs. mNGS testing reduces antibiotic-related complications, shortens hospital stay and antibiotic use duration, and improves treatment success rate, benefits which are particularly evident in culture-negative cases.
Collapse
Affiliation(s)
- Hongxin Hu
- Department of Orthopaedics, Affiliated Hospital of Putian University, Putian, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Haiqi Ding
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianhua Lyu
- Department of Orthopaedics, Affiliated Hospital of Putian University, Putian, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yang Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Changyu Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Chaofan Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Wenbo Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xinyu Fang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Wenming Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Orthopaedic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Institute of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
45
|
Rahman MA, Amirkhani A, Mempin M, Ahn SB, Deva AK, Baker MS, Vickery K, Hu H. The Low-Abundance Plasma Proteome Reveals Differentially Abundant Proteins Associated with Breast Implant Capsular Contracture: A Pilot Study. Proteomes 2024; 12:22. [PMID: 39189262 PMCID: PMC11348101 DOI: 10.3390/proteomes12030022] [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: 05/31/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/28/2024] Open
Abstract
Capsular contracture (CC) is one of the most common postoperative complications associated with breast implant-associated infections. The mechanisms that lead to CC remain poorly understood. Plasma is an ideal biospecimen for early proteomics biomarker discovery. However, as high-abundance proteins mask signals from low-abundance proteins, identifying novel or specific proteins as biomarkers for a particular disease has been hampered. Here, we employed depletion of high-abundance plasma proteins followed by Tandem Mass Tag (TMT)-based quantitative proteomics to compare 10 healthy control patients against 10 breast implant CC patients. A total of 450 proteins were identified from these samples. Among them, 16 proteins were significantly differentially expressed in which 5 proteins were upregulated and 11 downregulated in breast implant CC patients compared to healthy controls. Gene Ontology enrichment analysis revealed that proteins related to cell, cellular processes and catalytic activity were highest in the cellular component, biological process, and molecular function categories, respectively. Further, pathway analysis revealed that inflammatory responses, focal adhesion, platelet activation, and complement and coagulation cascades were enriched pathways. The differentially abundant proteins from TMT-based quantitative proteomics have the potential to provide important information for future mechanistic studies and in the development of breast implant CC biomarkers.
Collapse
Affiliation(s)
- Md. Arifur Rahman
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | | | - Maria Mempin
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Seong Beom Ahn
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Anand K. Deva
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Mark S. Baker
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Karen Vickery
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Honghua Hu
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
- Jinhua Institute of Zhejiang University, Jinhua 321016, China
| |
Collapse
|
46
|
Okpala OE, Rondevaldova J, Osei-Owusu H, Kudera T, Kokoskova T, Kokoska L. Susceptibility of Staphylococcus aureus to Anti-Inflammatory Drugs with a Focus on the Combinatory Effect of Celecoxib with Oxacillin In Vitro. Molecules 2024; 29:3665. [PMID: 39125072 PMCID: PMC11314137 DOI: 10.3390/molecules29153665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Musculoskeletal infections (MIs) are among the most difficult-to-treat staphylococcal diseases due to antibiotic resistance. This has encouraged the development of innovative strategies, such as combination therapy, to combat MI. The aim of this study was to investigate the in vitro antistaphylococcal activity of anti-inflammatory drugs and the combined antimicrobial effect of celecoxib and oxacillin. The minimum inhibitory concentrations (MICs) of 17 anti-inflammatory drugs against standard strains and clinical isolates of S. aureus, including methicillin-resistant strains (MRSAs), were determined using the broth microdilution method. The fractional inhibitory concentration indices (FICIs) were evaluated using checkerboard assays. Celecoxib produced the most potent antistaphylococcal effect against all tested strains (MICs ranging from 32 to 64 mg/L), followed by that of diacerein against MRSA3 and MRSA ATCC 33592 (MIC 64 mg/L). Several synergistic effects were observed against the tested S. aureus strains, including MRSA (FICI ranging from 0.087 to 0.471). The strongest synergistic interaction (FICI 0.087) was against MRSA ATCC 33592 at a celecoxib concentration of 2 mg/L, with a 19-fold oxacillin MIC reduction (from 512 to 26.888 mg/L). This is the first report on the combined antistaphylococcal effect of celecoxib and oxacillin. These findings suggest celecoxib and its combination with oxacillin as perspective agents for research focused on the development of novel therapies for MI caused by S. aureus. This study further indicates that celecoxib could resensitize certain MRSA strains, in some cases, to be susceptible to β-lactams (e.g., oxacillin) that were not previously tested. It is essential to mention that the in vitro concentrations of anti-inflammatory drugs are higher than those typically obtained in patients. Therefore, an alternative option for its administration could be the use of a drug delivery system for the controlled slow release from an implant at the infection site.
Collapse
Affiliation(s)
- Onyedika Emmanuel Okpala
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic; (O.E.O.); (J.R.); (H.O.-O.)
| | - Johana Rondevaldova
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic; (O.E.O.); (J.R.); (H.O.-O.)
| | - Hayford Osei-Owusu
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic; (O.E.O.); (J.R.); (H.O.-O.)
| | - Tomas Kudera
- Drift-Food Research Centre, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic;
| | - Tersia Kokoskova
- Department of Animal Science and Food Processing, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic;
| | - Ladislav Kokoska
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic; (O.E.O.); (J.R.); (H.O.-O.)
| |
Collapse
|
47
|
Zhou C, Wang Q, Cao H, Jiang J, Gao L. Nanozybiotics: Advancing Antimicrobial Strategies Through Biomimetic Mechanisms. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403362. [PMID: 38874860 DOI: 10.1002/adma.202403362] [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: 03/05/2024] [Revised: 06/09/2024] [Indexed: 06/15/2024]
Abstract
Infectious diseases caused by bacterial, viral, and fungal pathogens present significant global health challenges. The rapid emergence of antimicrobial resistance exacerbates this issue, leading to a scenario where effective antibiotics are increasingly scarce. Traditional antibiotic development strategies are proving inadequate against the swift evolution of microbial resistance. Therefore, there is an urgent need to develop novel antimicrobial strategies with mechanisms distinct from those of existing antibiotics. Nanozybiotics, which are nanozyme-based antimicrobials, mimic the catalytic action of lysosomal enzymes in innate immune cells to kill infectious pathogens. This review reinforces the concept of nanozymes and provides a comprehensive summary of recent research advancements on potential antimicrobial candidates. Initially, nanozybiotics are categorized based on their activities, mimicking either oxidoreductase-like or hydrolase-like functions, thereby highlighting their superior mechanisms in combating antimicrobial resistance. The review then discusses the progress of nanozybiotics in treating bacterial, viral, and fungal infections, confirming their potential as novel antimicrobial candidates. The translational potential of nanozybiotic-based products, including hydrogels, nanorobots, sprays, bandages, masks, and protective clothing, is also considered. Finally, the current challenges and future prospects of nanozybiotic-related products are explored, emphasizing the design and antimicrobial capabilities of nanozybiotics for future applications.
Collapse
Affiliation(s)
- Caiyu Zhou
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Haidian, Beijing, 100049, China
| | - Qian Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Haidian, Beijing, 100049, China
| | - Haolin Cao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Haidian, Beijing, 100049, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450052, China
| |
Collapse
|
48
|
Zorrón M, Cabrera AL, Sharma R, Radhakrishnan J, Abbaszadeh S, Shahbazi M, Tafreshi OA, Karamikamkar S, Maleki H. Emerging 2D Nanomaterials-Integrated Hydrogels: Advancements in Designing Theragenerative Materials for Bone Regeneration and Disease Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403204. [PMID: 38874422 PMCID: PMC11336986 DOI: 10.1002/advs.202403204] [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: 03/26/2024] [Revised: 05/16/2024] [Indexed: 06/15/2024]
Abstract
This review highlights recent advancements in the synthesis, processing, properties, and applications of 2D-material integrated hydrogels, with a focus on their performance in bone-related applications. Various synthesis methods and types of 2D nanomaterials, including graphene, graphene oxide, transition metal dichalcogenides, black phosphorus, and MXene are discussed, along with strategies for their incorporation into hydrogel matrices. These composite hydrogels exhibit tunable mechanical properties, high surface area, strong near-infrared (NIR) photon absorption and controlled release capabilities, making them suitable for a range of regeneration and therapeutic applications. In cancer therapy, 2D-material-based hydrogels show promise for photothermal and photodynamic therapies, and drug delivery (chemotherapy). The photothermal properties of these materials enable selective tumor ablation upon NIR irradiation, while their high drug-loading capacity facilitates targeted and controlled release of chemotherapeutic agents. Additionally, 2D-materials -infused hydrogels exhibit potent antibacterial activity, making them effective against multidrug-resistant infections and disruption of biofilm generated on implant surface. Moreover, their synergistic therapy approach combines multiple treatment modalities such as photothermal, chemo, and immunotherapy to enhance therapeutic outcomes. In bio-imaging, these materials serve as versatile contrast agents and imaging probes, enabling their real-time monitoring during tumor imaging. Furthermore, in bone regeneration, most 2D-materials incorporated hydrogels promote osteogenesis and tissue regeneration, offering potential solutions for bone defects repair. Overall, the integration of 2D materials into hydrogels presents a promising platform for developing multifunctional theragenerative biomaterials.
Collapse
Affiliation(s)
- Melanie Zorrón
- Institute of Inorganic ChemistryDepartment of ChemistryFaculty of Mathematics and Natural SciencesUniversity of CologneGreinstraße 650939CologneGermany
| | - Agustín López Cabrera
- Institute of Inorganic ChemistryDepartment of ChemistryFaculty of Mathematics and Natural SciencesUniversity of CologneGreinstraße 650939CologneGermany
| | - Riya Sharma
- Institute of Inorganic ChemistryDepartment of ChemistryFaculty of Mathematics and Natural SciencesUniversity of CologneGreinstraße 650939CologneGermany
| | - Janani Radhakrishnan
- Department of BiotechnologyNational Institute of Animal BiotechnologyHyderabad500 049India
| | - Samin Abbaszadeh
- Department of Pharmacology and ToxicologySchool of PharmacyUrmia University of Medical SciencesUrmia571478334Iran
| | - Mohammad‐Ali Shahbazi
- Department of Biomaterials and Biomedical TechnologyUniversity Medical Center GroningenUniversity of GroningenAntonius Deusinglaan 1GroningenAV, 9713The Netherlands
| | - Omid Aghababaei Tafreshi
- Microcellular Plastics Manufacturing LaboratoryDepartment of Mechanical and Industrial EngineeringUniversity of TorontoTorontoOntarioM5S 3G8Canada
- Smart Polymers & Composites LabDepartment of Mechanical and Industrial EngineeringUniversity of TorontoTorontoOntarioM5S 3G8Canada
| | - Solmaz Karamikamkar
- Terasaki Institute for Biomedical Innovation11570 W Olympic BoulevardLos AngelesCA90024USA
| | - Hajar Maleki
- Institute of Inorganic ChemistryDepartment of ChemistryFaculty of Mathematics and Natural SciencesUniversity of CologneGreinstraße 650939CologneGermany
- Center for Molecular Medicine CologneCMMC Research CenterRobert‐Koch‐Str. 2150931CologneGermany
| |
Collapse
|
49
|
Sherafati Chaleshtori A, Marzhoseyni Z, Saeedi N, Azar Bahadori R, Mollazadeh S, Pourghadamyari H, Sajadimoghadam E, Abbaszadeh‐Goudarzi K, Moradi Hasan-Abad A, Sharafati Chaleshtori R. Gelatin-based nanoparticles and antibiotics: a new therapeutic approach for osteomyelitis? Front Mol Biosci 2024; 11:1412325. [PMID: 39139812 PMCID: PMC11319135 DOI: 10.3389/fmolb.2024.1412325] [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: 04/04/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
The result of infection of bone with microorganisms is osteomyelitis and septic arthritis. Methicillin-resistant Staphylococcus aureus (MRSA) is responsible for most of its cases (more than 50%). Since MRSA is resistant to many treatments, it is accompanied by high costs and numerous complications, necessitating more effective new treatments. Recently, development of gelatin nanoparticles have attracted the attention of scientists of biomedicine to itself, and have been utilized as a delivery vehicle for antibiotics because of their biocompatibility, biodegradability, and cost-effectiveness. Promising results have been reported with gelatin modification and combinations with chemical agents. Although these findings have been suggested that gelatin has the potential to be a suitable option for continuous release of antibiotics in osteomyelitis and septic arthritis treatment, they still have not become routine in clinical practices. The most deliver antibiotic using gelatin-derived composites is vancomycin which is showed the good efficacy. To date, a number of pre-clinical studies evaluated the utility of gelatin-based composites in the management of osteomyelitis. Gelatin-based composites were found to have satisfactory performance in the control of infection, as well as the promotion of bone defect repair in chronic osteomyelitis models. This review summarized the available evidence which provides a new insight into gelatin-derived composites with controlled release of antibiotics.
Collapse
Affiliation(s)
- Ali Sherafati Chaleshtori
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Orthopedics, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zeynab Marzhoseyni
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Negin Saeedi
- Department of Microbiology, Faculty of Biological Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Rosita Azar Bahadori
- Department of Molecular Genetics, Parand Branch, Islamic Azad University, Tehran, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hossein Pourghadamyari
- Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Esmaeil Sajadimoghadam
- Department of Nursing, School of Nursing and Midwifery, Bam University of Medical Sciences, Bam, Iran
| | | | - Amin Moradi Hasan-Abad
- Autoimmune Diseases Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| | - Reza Sharafati Chaleshtori
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
50
|
Loffredo M, Casciaro B, Bellavita R, Troiano C, Brancaccio D, Cappiello F, Merlino F, Galdiero S, Fabrizi G, Grieco P, Stella L, Carotenuto A, Mangoni ML. Strategic Single-Residue Substitution in the Antimicrobial Peptide Esc(1-21) Confers Activity against Staphylococcus aureus, Including Drug-Resistant and Biofilm Phenotype. ACS Infect Dis 2024; 10:2403-2418. [PMID: 38848266 PMCID: PMC11250030 DOI: 10.1021/acsinfecdis.4c00130] [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: 02/19/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024]
Abstract
Staphylococcus aureus, a bacterium resistant to multiple drugs, is a significant cause of illness and death worldwide. Antimicrobial peptides (AMPs) provide an excellent potential strategy to cope with this threat. Recently, we characterized a derivative of the frog-skin AMP esculentin-1a, Esc(1-21) (1) that is endowed with potent activity against Gram-negative bacteria but poor efficacy against Gram-positive strains. In this study, three analogues of peptide 1 were designed by replacing Gly8 with α-aminoisobutyric acid (Aib), Pro, and dPro (2-4, respectively). The single substitution Gly8 → Aib8 in peptide 2 makes it active against the planktonic form of Gram-positive bacterial strains, especially Staphylococcus aureus, including multidrug-resistant clinical isolates, with an improved biostability without resulting in cytotoxicity to mammalian cells. Moreover, peptide 2 showed a higher antibiofilm activity than peptide 1 against both reference and clinical isolates of S. aureus. Peptide 2 was also able to induce rapid bacterial killing, suggesting a membrane-perturbing mechanism of action. Structural analysis of the most active peptide 2 evidenced that the improved biological activity of peptide 2 is the consequence of a combination of higher biostability, higher α helical content, and ability to reduce membrane fluidity and to adopt a distorted helix, bent in correspondence of Aib8. Overall, this study has shown how a strategic single amino acid substitution is sufficient to enlarge the spectrum of activity of the original peptide 1, and improve its biological properties for therapeutic purposes, thus paving the way to optimize AMPs for the development of new broad-spectrum anti-infective agents.
Collapse
Affiliation(s)
- Maria
Rosa Loffredo
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| | - Bruno Casciaro
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| | - Rosa Bellavita
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Cassandra Troiano
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133 Rome, Italy
| | - Diego Brancaccio
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Floriana Cappiello
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| | - Francesco Merlino
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Stefania Galdiero
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Giancarlo Fabrizi
- Department
of Chemistry and Technology of Drugs, “Department of Excellence
2018−2022”, Sapienza University
of Rome, 00185 Rome, Italy
| | - Paolo Grieco
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Lorenzo Stella
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133 Rome, Italy
| | - Alfonso Carotenuto
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Maria Luisa Mangoni
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| |
Collapse
|