1
|
Qi Y, Ren S, Ou X, Li P, Wu H, Che Y, Wang X. Ultrasound-activated sonothermal-catalytic synergistic therapy via asymmetric electron distribution for bacterial wound infections. Biomaterials 2025; 321:123338. [PMID: 40239594 DOI: 10.1016/j.biomaterials.2025.123338] [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: 01/15/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
Antibiotic-resistant bacterial infections present a growing global health challenge, requiring innovative therapeutic solutions to overcome current limitations. We introduce boron-integrated bismuth oxide (B-BiO2) nanosheets with an asymmetrically distributed electronic structure for ultrasound-activated synergistic sonothermal and catalytic therapy. Boron incorporation enhances local electron density distribution, optimizing charge separation and significantly improving sonothermal and catalytic efficiency, as validated by density functional theory calculations. These nanosheets exhibit dual functionality, effectively generating localized heat and reactive oxygen species (ROS) under ultrasound, leading to 99.999 % antibacterial efficacy against multidrug-resistant pathogens by disrupting bacterial membranes, as demonstrated through all-atom simulations and in vitro experiments. The simulations further reveal that sonothermal conversion effects enhance bacterial membrane fluidity and induce structural defects, amplifying ROS-induced oxidative damage and membrane destabilization. In vivo, B-BiO2 nanosheets accelerate wound healing in methicillin-resistant Staphylococcus aureus (MRSA)-infected murine models, achieving 99.8 % closure by day 14 by reducing inflammation and promoting angiogenesis and tissue regeneration. Transcriptomic analysis highlights the activation of extracellular matrix remodeling, angiogenesis, and autophagy pathways, underscoring the nanosheets' therapeutic potential. This study establishes ultrasound-activated B-BiO2 nanosheets as a novel nanotherapeutic platform, leveraging asymmetric electron distribution to synergistically combat drug-resistant infections and promote effective wound healing.
Collapse
Affiliation(s)
- Ye Qi
- Research Institute of Biomedical and Advanced Materials, College of Life and Health, Dalian University, 10 Xuefu Street, Dalian, Liaoning, 116622, China.
| | - Shuangsong Ren
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, 193 Lianhe Road, Dalian, Liaoning, 116011, China
| | - Xiaolong Ou
- Research Institute of Biomedical and Advanced Materials, College of Life and Health, Dalian University, 10 Xuefu Street, Dalian, Liaoning, 116622, China
| | - Pisong Li
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, 6 Jiefang Street, Dalian, Liaoning, 116001, China
| | - Han Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, 193 Lianhe Road, Dalian, Liaoning, 116011, China
| | - Ying Che
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, 193 Lianhe Road, Dalian, Liaoning, 116011, China.
| | - Xinyi Wang
- Research Institute of Biomedical and Advanced Materials, College of Life and Health, Dalian University, 10 Xuefu Street, Dalian, Liaoning, 116622, China.
| |
Collapse
|
2
|
Huang Z, Ma Y, Yang X, Yang X, Cheng Y, Zhang A. Ultrasound-switchable piezoelectric BiVO 4/fullerene heterostructure for on-demand ROS modulation in MRSA-infected diabetic wound healing. BIOMATERIALS ADVANCES 2025; 174:214307. [PMID: 40233477 DOI: 10.1016/j.bioadv.2025.214307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Persistent microbial infections and excessive reactive oxygen species (ROS) accumulation severely impede diabetic wound healing. Herein, we developed an ultrasound-switchable BiVO4/fullerene piezoelectric heterostructure via a one-pot solvothermal method, enabling on-demand transition between bactericidal action and ROS scavenging for treating infected diabetic wounds. Under 8-min ultrasound (US) irradiation, the heterojunction sonosensitizer leveraged piezoelectric polarization to generate substantial ROS in real-time through a narrowed energy band gap and enhanced charge carrier separation and migration efficiency, resulting in the disruption of bacterial membrane integrity and 99.9 % eradication of methicillin-resistant Staphylococcus aureus (MRSA). Upon US withdrawal, the sonosensitizer spontaneously transitioned to an antioxidative state through fullerene-mediated ROS scavenging, effectively neutralizing excess ROS and restoring cellular redox balance. In an MRSA-infected diabetic wound model, this ultrasound-responsive duality effectively suppressed bacterial proliferation, reduced inflammation, enhanced angiogenesis, and ultimately accelerated wound healing within 14 days. This ultrasound-switchable therapeutic strategy offers promising insights for managing drug-resistant infections and other ROS-mediated biomedical challenges.
Collapse
Affiliation(s)
- Zini Huang
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Engineering Technology Research Centre of Energy Polymer Materials, South-Central Minzu University, Wuhan 430074, PR China
| | - Yihan Ma
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Engineering Technology Research Centre of Energy Polymer Materials, South-Central Minzu University, Wuhan 430074, PR China.
| | - Xinyi Yang
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Engineering Technology Research Centre of Energy Polymer Materials, South-Central Minzu University, Wuhan 430074, PR China
| | - Xiaoping Yang
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Engineering Technology Research Centre of Energy Polymer Materials, South-Central Minzu University, Wuhan 430074, PR China
| | - Yinjia Cheng
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Engineering Technology Research Centre of Energy Polymer Materials, South-Central Minzu University, Wuhan 430074, PR China
| | - Aiqing Zhang
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Engineering Technology Research Centre of Energy Polymer Materials, South-Central Minzu University, Wuhan 430074, PR China.
| |
Collapse
|
3
|
Yuwen L, Liu Y, Xu F, Zhang C, Chen X, Yin Z, Liang B, Wang L. Fe 3O 4/MnCO 3 microbubbles for efficient elimination of bacterial biofilms by mechanical/sonodynamic effects under ultrasound irradiation and magnetic field targeting. Biomater Sci 2025; 13:3367-3379. [PMID: 40354102 DOI: 10.1039/d5bm00227c] [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: 05/14/2025]
Abstract
Bacterial biofilms present significant challenges in treatment with traditional antibiotics. Ultrasound (US)-responsive antibacterial agents have emerged as promising alternatives for treating bacterial biofilm infections. However, these agents are often limited by antibiotic dependence, inadequate targeting, and low antibacterial efficacy. Herein, we develop Fe3O4/MnCO3 microbubbles (FMMB) by self-assembly of Fe3O4 nanoparticles (Fe3O4 NPs) and MnCO3 nanoparticles (MnCO3 NPs). Under the direction of the magnetic field (MF), FMMB can be directed toward the methicillin-resistant Staphylococcus aureus (MRSA) biofilm. Under US irradiation, FMMB can disrupt the structure of MRSA biofilms by cavitation-induced mechanical effects and kill bacteria with reactive oxygen species (ROS) generated by MnCO3 NPs through the sonodynamic effect. In a mouse model with catheter-associated MRSA biofilm infection, FMMB removed 58.8% of the biofilm with MF and US, and the bacterial inactivation efficiency reached as high as 4.1 log (99.992%). This work develops multifunctional microbubbles with both US-responsive mechanical and sonodynamic effects for biofilm disruption and MF-responsive properties for biofilm targeting, offering a promising strategy for designing antibiofilm agents to effectively treat bacterial biofilm infections.
Collapse
Affiliation(s)
- Lihui Yuwen
- State Key Laboratory of Flexible Electronics (LoFE) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Yuan Liu
- State Key Laboratory of Flexible Electronics (LoFE) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Fengjiao Xu
- State Key Laboratory of Flexible Electronics (LoFE) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Chi Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Xiaolong Chen
- State Key Laboratory of Flexible Electronics (LoFE) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Zhaowei Yin
- Department of Orthopaedic, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Bin Liang
- Department of Orthopaedic, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Lianhui Wang
- State Key Laboratory of Flexible Electronics (LoFE) & Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| |
Collapse
|
4
|
Zhang T, Luo Z. Stimulus-responsive nanomaterials for ocular antimicrobial therapy. NANOSCALE 2025. [PMID: 40390637 DOI: 10.1039/d4nr05462h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Nanomaterials exhibit a promising new avenue for treating infectious keratitis, having garnered considerable interest in the ophthalmic medical community due to their unique properties including higher target specificity, enhanced bioactivity of loaded agents, reduced drug dosage, and stimulus-responsive drug release. These stimulus-responsive nanomaterial-mediated therapeutic strategies offer innovative approaches for managing ocular antimicrobial diseases. In this review, we aim to summarize current applications of stimulus-responsive nanotherapeutics for ocular antimicrobial therapy. We briefly introduce the basic ocular structure, ocular barrier, infectious keratitis classification, and its microenvironment. Following this, we summarize the nanotherapeutic antimicrobial strategies employed in treating ocular infections including endogenous stimulus-responsive ocular nanodrugs, sonodynamic therapy, and wearable device-based therapy, focusing on their design principles, developmental progress, and advantages and limitations. Finally, we critically evaluate the biosafety profiles of responsive nanomaterials, specifically addressing cytotoxicity and immune interactions. To conclude, we discuss key challenges in this research field and future opportunities with explicit emphasis on clinical translation and practical medical applications.
Collapse
Affiliation(s)
- Tao Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China.
- NHC Key Laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai 200031, China
| | - Zichao Luo
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China.
- NHC Key Laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai 200031, China
| |
Collapse
|
5
|
Gao F, Zhou R, He Y, Zhang Y, Bao C, Feng G. Bio-Mimicking Nanoparticle System Facilitates Sonodynamic-Mediated Clearance of Extensively Drug-Resistant Bacteria. ACS Biomater Sci Eng 2025; 11:2988-3002. [PMID: 40294106 DOI: 10.1021/acsbiomaterials.4c02455] [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: 04/30/2025]
Abstract
The increasing prevalence of carbapenem-resistant and extensively drug-resistant Acinetobacter baumannii (XDR-Ab) poses a critical challenge in treating hospital-acquired pulmonary infections. In this study, we developed a biomimetic neutrophil membrane-coated nanoparticle system, NM@PCN-TIG, for the targeted delivery of tigecycline (TIG). The system utilizes the porphyrin-based metal-organic framework (MOF) PCN-224 as the core of the nanoparticle, encapsulating TIG and coated with a neutrophil membrane (NM) to enhance immune evasion and targeting of infection sites. Its loading efficiency, controlled release properties, cytotoxicity, and bactericidal activity under ultrasound mediation were systematically evaluated in vitro and in vivo. Our results demonstrated that NM@PCN-TIG significantly enhanced the bactericidal efficacy of TIG, increased reactive oxygen species (ROS) production, and promoted macrophage polarization toward an anti-inflammatory phenotype. This innovative biomimetic TIG nanosystem shows great potential as a platform for addressing XDR-Ab-induced pneumonia.
Collapse
Affiliation(s)
- Fenglin Gao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Runlu Zhou
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yucong He
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yuanyuan Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Cui Bao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Ganzhu Feng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| |
Collapse
|
6
|
Lan J, Zou J, Xin H, Sun J, Han T, Sun M, Niu M. Nanomedicines as disruptors or inhibitors of biofilms: Opportunities in addressing antimicrobial resistance. J Control Release 2025; 381:113589. [PMID: 40032007 DOI: 10.1016/j.jconrel.2025.113589] [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: 12/18/2024] [Revised: 02/02/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
The problem of antimicrobial resistance (AMR) has caused global concern due to its great threat to human health. Evidences are emerging for a critical role of biofilms, one of the natural protective mechanisms developed by bacteria during growth, in resisting commonly used clinical antibiotics. Advances in nanomedicines with tunable physicochemical properties and unique anti-biofilm mechanisms provide opportunities for solving AMR risks more effectively. In this review, we summarize the five "A" stages (adhesion, amplification, alienation, aging and allocation) of biofilm formation and mechanisms through which they protect the internal bacteria. Aimed at the characteristics of biofilms, we emphasize the design "THAT" principles (targeting, hacking, adhering and transport) of nanomedicines in their interactions with biofilms and internal bacteria. Furthermore, recent progresses in multimodal antibacterial nanomedicines, including biofilms disruption and bactericidal activity, and the types of currently available antibiofilm nanomedicines contained organic and inorganic nanomedicines are outlined and highlighted their potential applications in the development of preclinical research. Last but not least, we offer a perspective for the effectiveness of nanomedicines designed to address AMR and challenges associated with their clinical translation.
Collapse
Affiliation(s)
- Jiaming Lan
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jingyu Zou
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - He Xin
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Tao Han
- Department of Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Mengchi Sun
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| | - Meng Niu
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China.
| |
Collapse
|
7
|
Liu H, Nan Z, Zhao C, Bai L, Shi L, He C, Wu D, Wan M, Feng Y. Emerging synergistic strategies for enhanced antibacterial sonodynamic therapy: Advances and prospects. ULTRASONICS SONOCHEMISTRY 2025; 116:107288. [PMID: 40038013 PMCID: PMC11986242 DOI: 10.1016/j.ultsonch.2025.107288] [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: 10/23/2024] [Revised: 12/29/2024] [Accepted: 02/22/2025] [Indexed: 03/06/2025]
Abstract
Antibacterial therapy has been extensively applied in medical field to alleviate the severity and mortality of infection. However, it still exists some issues such as drug side effects, limited efficacy and bacterial resistance. Among the alternative therapies, antibacterial sonodynamic therapy (aSDT) has been explored as a promising approach to tackle those crises. It is meaningful to investigate superior strategy to augment the therapeutic efficacy of aSDT. This review summarizes the potential aSDT-based antibacterial mechanisms and comprehensively discusses the prevailing synergistic strategies, such as nanomaterials-based aSDT antibacterial strategy, aSDT + strategy with physical, chemical and biological methods. Moreover, we also reviewed the medical applications of aSDT strategies. Finally, the perspectives on the current challenges that need be resolved in aSDT are proposed. We expect that this review could provide robust support to expedite the clinical applications of aSDT.
Collapse
Affiliation(s)
- Hengyu Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Zhezhu Nan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Chen Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Liang Bai
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Linrong Shi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Chenhui He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yi Feng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China.
| |
Collapse
|
8
|
Wang L, Chen F, Wu N, Hu L, Xiao H, Zhang H, Zhou D. Inhalable Polymeric Nanoparticle Containing Triphenylphosphanium Bromide-modified Sonosensitizer for Enhanced Therapy of Acute Bacterial Pneumonia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417469. [PMID: 40171966 DOI: 10.1002/advs.202417469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/24/2025] [Indexed: 04/04/2025]
Abstract
Sonodynamic therapy (SDT) has good feasibility to deeply seated infections, but SDT alone is insufficient being highly effective against multidrug-resistant (MDR) bacteria. SDT combined with triphenylphosphanium bromide (P+Ph3Br-) is expected to solve this problem. This work develops a pseudo-conjugated polymer PFCPS-P containing cationic P+Ph3Br--modified sonosensitizer FCPS (FCPS-P) and ROS-sensitive thioketal bonds. PFCPS-P is assembled with DSPE-mPEG2000 to generate nanoparticle NPFCPS-P. FCPS has SDT effect and generates ROS under ultrasound (US) stimulation. ROS triggers the degradation of NPFCPS-P and release of FCPS-P, endowing highly favored biosafety. FCPS-P targets to bacterial surface through electrostatic interaction and achieves bacterial killing under a synergistic action of SDT and P+Ph3Br-. In vitro, NPFCPS-P+US gives >90% inhibition rates against MDR ESKAPE pathogens, moreover, it causes bacterial metabolic disorders including inhibited nucleic acid synthesis, disordered energy metabolism, excessive oxidative stress, and suppressed biofilm formation and virulence. In mice, NPFCPS-P+US exhibits a 99.3% bactericidal rate in Pseudomonas aeruginosa-induced sublethal pneumonia and renders a 90% animal survival rate in lethal pneumonia, and additionally immunological staining and transcriptomics analyses reveal that NPFCPS-P+US induces inhibited inflammatory response and accelerated lung injury repair. Taken together, NPFCPS-P+US is a promising antibiotics-alternative strategy for treating deeply seated bacterial infections.
Collapse
Affiliation(s)
- Lin Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
- Northern Medical Branch of PLA General Hospital, Beijing, 100094, China
| | - Fangzhou Chen
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Nier Wu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Lingfei Hu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| |
Collapse
|
9
|
Wang C, Yang Y, Wang N, Luan A, Wang H, Hu C. Design and application of antimicrobial nanomaterials in the treatment of periodontitis. Nanomedicine (Lond) 2025; 20:707-723. [PMID: 40042364 PMCID: PMC11970792 DOI: 10.1080/17435889.2025.2469492] [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: 11/28/2024] [Accepted: 02/17/2025] [Indexed: 04/02/2025] Open
Abstract
Periodontitis is a chronic inflammatory disease induced by the microbiome, leading to the destruction of periodontal structures and potentially resulting in tooth loss. Using local drug delivery systems as an adjunctive therapy to scaling and root planning in periodontitis is a promising strategy. However, this administration method's effectiveness is constrained by the complexity of the periodontal environment. Nanomaterials have demonstrated significant potential in the antibacterial treatment of periodontitis, attributed to their controllable size, shape, and surface charge, high design flexibility, high reactivity, and high specific surface area. In this review, we summarize the complex periodontal microenvironment and the difficulties of local drug delivery in periodontitis, explicitly reviewing the application and design strategies of nanomaterials with unique properties in the distinct microenvironment of periodontitis. Furthermore, the review discusses the limitations of current research, proposes feasible solutions, and explores prospects for using nanomaterials in this context.
Collapse
Affiliation(s)
- Chunlin Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yujun Yang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Ning Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Aohan Luan
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Huilin Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Chen Hu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Andavar A, Bhagavathi VR, Cousin J, Parekh N, Razavi ZS, Tan B. Current Research in Drug-Free Cancer Therapies. Bioengineering (Basel) 2025; 12:341. [PMID: 40281701 PMCID: PMC12024433 DOI: 10.3390/bioengineering12040341] [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: 12/27/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/29/2025] Open
Abstract
Cancer treatment has historically depended on conventional methods like chemotherapy, radiation, and surgery; however, these strategies frequently present considerable limitations, including toxicity, resistance, and negative impacts on healthy tissues. In addressing these challenges, drug-free cancer therapies have developed as viable alternatives, utilizing advanced physical and biological methods to specifically target tumor cells while reducing damage to normal tissues. This review examines several drug-free cancer treatment strategies, such as high-intensity focused energy beams, nanosecond pulsed electric fields, and photothermal therapy as well as the use of inorganic nanoparticles to promote selective apoptosis. We also investigate the significance of targeting the tumor microenvironment, precision medicine, and immunotherapy in the progression of personalized cancer therapies. Although these approaches demonstrate significant promise, challenges including scalability, safety, and regulatory obstacles must be resolved for clinical application. This paper presents an overview of current research in drug-free cancer therapies, emphasizing recent advancements, underlying scientific principles, and the steps required for clinical implementation.
Collapse
Affiliation(s)
- Akshaya Andavar
- Karpagam Academy of Higher Education, Coimbatore 641021, India;
| | | | - Justine Cousin
- École Publique d’Ingénieurs de la Santé et du Numérique (EPISEN), Université Paris-Est Créteil (UPEC), 94000 Créteil, France;
| | - Nirvi Parekh
- Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai 400019, India;
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran 1416634793, Iran;
| | - Bo Tan
- Institute of Biomedical Engineering Science and Technology (iBEST), Faculty of Engineering and Architectural Science, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| |
Collapse
|
11
|
Yang Y, Wang X, Hu Y, Liu Z, Ma X, Feng F, Zheng F, Guo X, Liu W, Liao W, Han L. Rapid enrichment and SERS differentiation of various bacteria in skin interstitial fluid by 4-MPBA-AuNPs-functionalized hydrogel microneedles. J Pharm Anal 2025; 15:101152. [PMID: 40115811 PMCID: PMC11925168 DOI: 10.1016/j.jpha.2024.101152] [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: 07/15/2024] [Revised: 11/01/2024] [Accepted: 11/18/2024] [Indexed: 03/23/2025] Open
Abstract
Bacterial infection is a major threat to global public health, and can cause serious diseases such as bacterial skin infection and foodborne diseases. It is essential to develop a new method to rapidly diagnose clinical multiple bacterial infections and monitor food microbial contamination in production sites in real-time. In this work, we developed a 4-mercaptophenylboronic acid gold nanoparticles (4-MPBA-AuNPs)-functionalized hydrogel microneedle (MPBA-H-MN) for bacteria detection in skin interstitial fluid. MPBA-H-MN could conveniently capture and enrich a variety of bacteria within 5 min. Surface enhanced Raman spectroscopy (SERS) detection was then performed and combined with machine learning technology to distinguish and identify a variety of bacteria. Overall, the capture efficiency of this method exceeded 50%. In the concentration range of 1 × 107 to 1 × 1010 colony-forming units/mL (CFU/mL), the corresponding SERS intensity showed a certain linear relationship with the bacterial concentration. Using random forest (RF)-based machine learning, bacteria were effectively distinguished with an accuracy of 97.87%. In addition, the harmless disposal of used MNs by photothermal ablation was convenient, environmentally friendly, and inexpensive. This technique provided a potential method for rapid and real-time diagnosis of multiple clinical bacterial infections and for monitoring microbial contamination of food in production sites.
Collapse
Affiliation(s)
- Ying Yang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Xingyu Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Yexin Hu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhongyao Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiao Ma
- Gansu Institute for Drug Control, Lanzhou, Gansu, 730000, China
| | - Feng Feng
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Feng Zheng
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Xinlin Guo
- School of Chemistry, University of Manchester, Manchester, M13 9PL, UK
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
- Zhejiang Center for Safety Study of Drug Substances (Industrial Technology Innovation Platform), Hangzhou, 310018, China
| | - Wenting Liao
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Lingfei Han
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
12
|
Sun X, Chen X, Wang S, Gu H, Bao H, Ning Z, Feng X, Chen Y. Oxygenous and biofilm-targeted nanosonosensitizer anchored with Pt nanozyme and antimicrobial peptide in the gelatin/sodium alginate hydrogel for infected diabetic wound healing. Int J Biol Macromol 2025; 293:139356. [PMID: 39743100 DOI: 10.1016/j.ijbiomac.2024.139356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/08/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Sonodynamic therapy is an emerging therapeutic approach for combating bacterial infections. However, the characteristics of hypoxia, high H2O2 microenvironment, and the formation of persistent biofilms in diabetic wound sites limit its efficacy in this field. To address these issues, we developed a multifunctional antibacterial hydrogel dressing PPCN@Pt-AMPs/HGel with the cross-linked gelatin and sodium alginate as the matrix, where the nanosonosensitizer PCN-224 was decorated with the oxygen-generating Pt nanoenzyme and further coupled with a biofilm-targeting antimicrobial peptide via an interacting polydopamine layer. This nano-composite hydrogel displayed improved mechanical properties as well as good biocompatibility and biodegradability. The catalase-like activity of the nanoparticles facilitated the ultrasound-induced generation of the singlet oxygen due to the catalytic decomposition of the H2O2 into O2. In vitro results showed that the hydrogel dressing exhibited excellent antimicrobial ability under low-intensity ultrasound stimulation, which could effectively inhibit the newly formed biofilm and eliminate the full-grown biofilms. In the infected diabetic wound of rats, PPCN@Pt-AMPs/HGel significantly enhanced the wound healing rate under low-intensity ultrasound stimulation and improved the regeneration outcomes by promoting granulation tissue formation, angiogenesis, and type III collagen deposition. In conclusion, our study provides a novel and effective antibacterial hydrogel dressing for sonodynamic treatment of diabetic wounds.
Collapse
Affiliation(s)
- Xinxing Sun
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Xinzhao Chen
- Department of Histology and Embryology, College of Basic Medical Science, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Sihui Wang
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Hongchun Gu
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Hongyang Bao
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Zixun Ning
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Xun Feng
- Department of Sanitary Chemistry, School of Public Health, Shenyang Medical College, No.146 Yellow River North Street, Shenyang 110034, China.
| | - Yang Chen
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China.
| |
Collapse
|
13
|
Xu J, Liu Y. Nanomaterials for liver cancer targeting: research progress and future prospects. Front Immunol 2025; 16:1496498. [PMID: 40092984 PMCID: PMC11906451 DOI: 10.3389/fimmu.2025.1496498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/07/2025] [Indexed: 03/19/2025] Open
Abstract
The incidence and mortality rates of liver cancer in China remain elevated. Although early-stage liver cancer is amenable to surgical resection, a significant proportion of patients are diagnosed at advanced stages. Currently, in addition to surgical resection for hepatocellular carcinoma, the primary treatment modalities predominantly include chemotherapy. The widespread use of chemotherapy, which non-selectively targets both malignant and healthy cells, often results in substantial immunosuppression. Simultaneously, the accumulation of chemotherapeutic agents can readily induce drug resistance upon reaching the physiological threshold, thereby diminishing the efficacy of these treatments. Besides chemotherapy, there exist targeted therapy, immunotherapy and other therapeutic approaches. Nevertheless, the development of drug resistance remains an inevitable challenge. To address these challenges, we turn to nanomedicine, an emerging and widely utilized discipline that significantly influences medical imaging, antimicrobial strategies, drug delivery systems, and other related areas. Stable and safe nanomaterials serve as effective carriers for delivering anticancer drugs. They enhance the precision of drug targeting, improve bioavailability, and minimize damage to healthy cells. This review focuses on common nanomaterial carriers used in hepatocellular carcinoma (HCC) treatment over the past five years. The following is a summary of the three drugs: Sorafenib, Gefitinib, and lenvatinib. Each drug employs distinct nanomaterial delivery systems, which result in varying levels of bioavailability, drug release rates, and therapeutic efficacy.
Collapse
Affiliation(s)
| | - Yefu Liu
- Department of Hepatopancreatobiliary Surgery, Cancer Hospital of Dalian University of
Technology, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
14
|
Yang X, Fang R, Li X, Kong W, Jin Y, Jiao R, Liu Z, Zhang M, Peng Q, Zhang Y, Song N. Engineered Nanovesicles for the Precise and Noninvasive Treatment of Deep Osteomyelitis Caused by MRSA Infection with Enhanced Immune Response. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11795-11810. [PMID: 39945439 DOI: 10.1021/acsami.4c20893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The clinical treatment of hospital-acquired persistent osteomyelitis caused by methicillin-resistant Staphylococcus aureus (MRSA) presents two major challenges: ineffective drug delivery into deep tissues and counteracting the rapid establishment of an immunosuppressive microenvironment. Indeed, MRSA can evade immunosurveillance and undermine both innate and adaptive immune responses. Herein, the engineered nanovesicles, functioning by combining sonodynamic therapy (SDT) with immune modulation, were constructed for the precise and noninvasive removal of MRSA in deep tissue and activation of the antimicrobial immune response using a newly engineered nanovesicle. Macrophage-derived M1 phenotypic microvesicles (M1-MW) internalized vancomycin-cross-linked micelles with the acoustic sensitizer indocyanine green (ICG) (VCG micelles). The vesicles of M1-MW were grafted with PEGylated mannose, allowing for targeted accumulation at the infection site. The VCG micelles were responsive to the highly reducing environment and released ICG to generate ROS after exposure to ultrasounds. This effect was combined with the presence of vancomycin to kill MRSA. In an osteomyelitis infection model, we observed an improved survival rate and reprogramming of macrophages to a pro-inflammatory M1 phenotype. The latter promoted T-cell activation and immune defense against MRSA-camouflaged homologous cell-transferred infections. Thus, our study presents a noninvasive and efficient treatment (VCG@MMW) for deep osteomyelitis with improved bacterial clearance and reduced risk of recurrence with enhanced immune response.
Collapse
Affiliation(s)
- Xingyue Yang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ren Fang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Xiaotian Li
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Weihao Kong
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yubao Jin
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ruohan Jiao
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Zhenggong Liu
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Meiqi Zhang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Qixian Peng
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300350, P. R. China
| | - Ningning Song
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| |
Collapse
|
15
|
谢 梦, 杨 敏, 李 欣, 杜 永. [Low-intensity pulsed ultrasound combined with nystatin treatment synergistically inhibits vaginal Candida albicans biofilm infection in rabbits]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:296-303. [PMID: 40031973 PMCID: PMC11875854 DOI: 10.12122/j.issn.1673-4254.2025.02.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Indexed: 03/05/2025]
Abstract
OBJECTIVES To explore the efficacy of low-intensity pulsed ultrasound (LIPUS) combined with nystatin for treatment of vaginal Candida albicans biofilm infection. METHODS In vitro cultured Candida albicans biofilm were treated with LIPUS, nystatin, or both, and the minimum inhibitory concentration (MIC) of nystatin was determined. Crystal violet staining, confocal laser microscopy (CLSM) and scanning electron microscopy were used to quantify the biofilm and observe the activity and morphological changes of the biofilms; DCFH-DA was used to detect the changes in reactive oxygen species (ROS). Twenty female New Zealand White rabbits with vaginal inoculation of Candida albicans biofilm were randomized into 4 groups for treatment with normal saline, LIPUS, nystatin, or both LIPUS and nystatin. The changes in vulvar symptoms of the rabbits were observed, and the histopathological and ultrastructural changes of the vagina before and after treatment were observed using HE staining and transmission electron microscopy. RESULTS In the combined treatment group, the MIC50 and MIC80 of nystatin in Candida albicans biofilms were both reduced by 50% compared with those in nystatin group, and the biofilm clearance rate increased by 26% and 68% compared with nystatin and LIPUS groups, respectively. Compared with nystatin and LIPUS treatment alone, the combined treatment produced stronger effects for inhibiting biofilm activity, causing structural disruption and promoting ROS production. In the rabbit models, the combined treatment more effectively improved vulvar symptoms and inflammatory infiltration, reduced residual vaginal hyphae/strains, and improved ultrastructure of the vaginal epithelium than LIPUS and nystatin treatment alone. CONCLUSIONS LIPUS combined with nystatin produces a significant synergistic antifungal effect against Candida albicans biofilm both in vitro and in vivo.
Collapse
|
16
|
Javanmard Z, Pourhajibagher M, Bahador A. New strategies to enhance antimicrobial photo-sonodynamic therapy based on nanosensitizers against bacterial infections. Folia Microbiol (Praha) 2025; 70:55-70. [PMID: 39367131 DOI: 10.1007/s12223-024-01206-8] [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: 04/13/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
The rapid evolution and spread of multidrug resistance among bacterial pathogens has significantly outpaced the development of new antibiotics, underscoring the urgent need for alternative therapies. Antimicrobial photodynamic therapy and antimicrobial sonodynamic therapy have emerged as promising treatments. Antimicrobial photodynamic therapy relies on the interaction between light and a photosensitizer to produce reactive oxygen species, which are highly cytotoxic to microorganisms, leading to their destruction without fostering resistance. Antimicrobial sonodynamic therapy, a novel variation, substitutes ultrasound for light to activate the sonosensitizers, expanding the therapeutic reach. To increase the efficiency of antimicrobial photodynamic therapy and antimicrobial sonodynamic therapy, the combination of these two methods, known as antimicrobial photo-sonodynamic therapy, is currently being explored and considered a promising approach. Recent advances, particularly in the application of nanomaterials, have further enhanced the efficacy of these therapies. Nanosensitizers, due to their improved reactive oxygen species generation and targeted delivery, offer significant advantages in overcoming the limitations of conventional sensitizers. These breakthroughs provide new avenues for treating bacterial infections, especially multidrug-resistant strains and biofilm-associated infections. Continued research, including comprehensive clinical studies, is crucial to optimizing nanomaterial-based antimicrobial photo-sonodynamic therapy for clinical use, ensuring their effectiveness in real-world applications.
Collapse
Affiliation(s)
- Zahra Javanmard
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Abbas Bahador
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Fellowship in Clinical Laboratory Sciences, BioHealth Lab, Tehran, Iran.
| |
Collapse
|
17
|
Zhao W, Zhang Y, Chen J, Hu D. Revolutionizing oral care: Reactive oxygen species (ROS)-Regulating biomaterials for combating infection and inflammation. Redox Biol 2025; 79:103451. [PMID: 39631247 PMCID: PMC11664010 DOI: 10.1016/j.redox.2024.103451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024] Open
Abstract
The human oral cavity is home to a delicate symbiosis between its indigenous microbiota and the host, the balance of which is easily perturbed by local or systemic factors, leading to a spectrum of oral diseases such as dental caries, periodontitis, and pulp infections. Reactive oxygen species (ROS) play crucial roles in the host's innate immune defenses. However, in chronic inflammatory oral conditions, dysregulated immune responses can result in excessive ROS production, which in turn exacerbates inflammation and causes tissue damage. Conversely, the potent antimicrobial properties of ROS have inspired the development of various anti-infective therapies. Therefore, the strategic modulation of ROS by innovative biomaterials is emerging as a promising therapeutic approach for oral infection and inflammation. This review begins by highlighting the state-of-the-art of ROS-regulating biomaterials, which are designed to generate, scavenge, or modulate ROS in a bidirectional manner. We then delve into the latest innovations in these biomaterials and their applications in treating a range of oral diseases, including dental caries, endodontic and periapical conditions, periodontitis, peri-implantitis, and oral candidiasis. The review concludes with an overview of the current challenges and future potential of these biomaterials in clinical settings. This review provides novel insights for the ongoing development of ROS-based therapeutic strategies for oral diseases.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Rehabilitation Medicine, Rehabilitation Medical Center, Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, PR China; State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Jing Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China; Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology Sichuan University, Chengdu, 610041, PR China.
| | - Danrong Hu
- Department of Rehabilitation Medicine, Rehabilitation Medical Center, Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
18
|
Xu Z, Zhang X, Shan Q, Zhu W, Jiang S, Li R, Wu X, Huo M, Ying B, Chen C, Chen X, Zhang K, Chen W, Chen J. Fluorocarbon-Functionalized Polymerization-Induced Self-Assembly Nanoparticles Alleviate Hypoxia to Enhance Sonodynamic Cancer Therapy. Adv Healthc Mater 2025; 14:e2403251. [PMID: 39487634 DOI: 10.1002/adhm.202403251] [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: 08/29/2024] [Revised: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Sonodynamic therapy (SDT) is an ultrasound-based, noninvasive cancer treatment that targets tumor cells by triggering reactive oxygen species production. However, the limited accumulation of sonosensitizers and the insufficient supply of O2 to the hypoxic environment at the tumor site greatly limit the effectiveness of SDT. To address these issues, positively charged porphyrin-containing nanoparticles (NPs) from self-assembling of fluorocarbon/polyethylene glycol amphiphilic block copolymer, which is synthesized through reversible addition-fragmentation chain transfer polymerization, are constructed. The NPs with fluorocarbon core and positively charged hydrophilic shells not only stabilize the sonosensitizer and improve its cellular uptake, but also act as an O2 carrier alleviating the hypoxic tumor environment. In vitro and in vivo experiments demonstrate that the NPs effectively deliver O2 to the tumor and supply sufficient O2 to Renca cells after ultrasound treatment. Consequently, the NPs inhibit hypoxia-induced resistance to SDT and significantly produce reactive oxygen species by activated porphyrin moieties, inducing apoptosis in cancer cells. These oxygen-enhanced sonosensitizer NPs hold promise for cancer therapies such as photodynamic therapy, radiotherapy, and chemotherapy by overcoming hypoxia-induced resistance.
Collapse
Affiliation(s)
- Zhikang Xu
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xuanxuan Zhang
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Qianyun Shan
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wei Zhu
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Shangxu Jiang
- Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, School of Chemistry and Chemical Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Rumei Li
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xiaojin Wu
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Meng Huo
- Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, School of Chemistry and Chemical Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Bin Ying
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chen Chen
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xiaoting Chen
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Kai Zhang
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, School of Chemistry and Chemical Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Weiyu Chen
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Jian Chen
- Department of Ultrasound in Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| |
Collapse
|
19
|
Wang G, Zhang C, Huang Z, Chen J, Chen H, Lin T, Zhou Z, Gu N, Huang P. Transcytosable and Ultrasound-Activated Liposome Enables Deep Penetration of Biofilm for Surgical Site Infection Management. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411092. [PMID: 39463041 DOI: 10.1002/adma.202411092] [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: 07/29/2024] [Revised: 09/22/2024] [Indexed: 10/29/2024]
Abstract
Biofilm-associated surgical site infection (BSSI) is a common and grievous postoperative complication lacking effective remedies, mainly due to the poor drug accumulation and penetration in the biofilms featured by dense extracellular polymeric substances (EPSs). Here, it is found that the vascular cell adhesion molecule-1 (VCAM1) is highly overexpressed in the vascular cells of BSSI. It is proposed that the combination of VCAM1-mediated transcytosis and ultrasonic cavitation can consecutively overcome the biological barriers of vascular endothelial cells and EPS for biofilm eradication. To demonstrate the feasibility, a VCAM1-targeted and ultrasound (US)-activated liposome (LPCOTML) loaded with a reactive-oxygen-species (ROS)-responsive lipoid prodrug of oleoyl meropenem, sonosensitizer of lipoid Ce6, and perfluoropentane is developed. LPCOTML can recognize the receptors on vascular cells, and initiate receptor-mediated transcytosis for transendothelial transport into the BSSI periphery. LPCOTML subsequently transforms from nanoparticle into microbubble via liquid-gas phase transition under US irradiation, triggering strong ultrasonic cavitation to blow up the EPS and deeply penetrate the biofilms. The sonosensitizer Ce6 induces ROS production under US irradiation and triggers the release of meropenem to induce potent antibacterial effect in a BSSI model. This study presents an effective strategy to tackle the biological barriers in BSSI via combining receptor-mediated transcytosis and ultrasonic cavitation.
Collapse
Affiliation(s)
- Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Chengyue Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zixuan Huang
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310030, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hongjian Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310009, China
| | - Tao Lin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zhuxian Zhou
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ning Gu
- Medical School of Nanjing University, Nanjing University, Nanjing, 210093, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
20
|
Caliskan M, Ilikci‐Sagkan R, Bayrak G, Ozlem‐Caliskan S. Monitoring Apoptosis and Myeloid Differentiation of Acridine Orange-Mediated Sonodynamic Therapy-Induced Human Promyelocytic Leukemia HL60 Cells. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2025; 44:15-34. [PMID: 39257135 PMCID: PMC11632649 DOI: 10.1002/jum.16575] [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: 04/01/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVES In the treatment of acute myeloid leukemia (AML), conventional therapies can lead to severe side effects and drug resistance. There is a need for alternative treatments that do not cause treatment resistance and have minimal or no side effects. Sonodynamic therapy (SDT), due to its noninvasive, multiple repeatability, localized treatment feature and do not cause treatment resistance, emerges as an alternative treatment option. However, it has not received sufficient attention in the treatment of AML especially acute promyelocytic leukemia (APL). The aim of the study was to investigate the potential differentiation and antileukemic effects of acridine orange (AO)-mediated SDT on HL60 cells. METHODS Cell viability was determined by the 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) method in the control, ultrasound, AO concentrations, and ultrasound-exposed AO concentrations groups. Transmission electron microscopy (TEM) was used to determine morphology, and flow cytometry was used to determine apoptosis, DNA cycle, cell volume, mitochondria membrane potential (Δψm), reactive oxygen species (ROS) production, and differentiation markers (CD11b and CD15) expressions. Additionally, toluidine blue staining for semithin sections was used to determine differentiation. RESULTS The cytotoxicity of AO-mediated SDT on HL60 cells was significantly higher than other groups, and TEM images showed that it caused various morphological changes typical for apoptosis. Flow cytometry results showed the presence of early apoptosis, subG1 arrest, loss of Δψm, increase of intracellular ROS production, decreased cell volume, and increased expression of CD11b (1.3-fold) antigen and CD15 (1.2-fold) antigen. CONCLUSION Data showed that AO-mediated SDT significantly induced apoptosis in HL60 cells. Increased expression of CD11b and CD15 antigens and morphological findings demonstrated that AO-mediated SDT contributes to granulocytic differentiation in HL60 cells. AO-mediated SDT has potential as an alternative treatment of APL.
Collapse
Affiliation(s)
- Metin Caliskan
- Department of Medical Biology, Faculty of MedicineUsak UniversityUsakTurkey
| | | | - Gulsen Bayrak
- Department of Histology and Embryology, Faculty of MedicineUsak UniversityUsakTurkey
| | | |
Collapse
|
21
|
Zheng R, Yu C, Yao D, Cai M, Zhang L, Ye F, Huang X. Engineering Stimuli-Responsive Materials for Precision Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406439. [PMID: 39444066 PMCID: PMC11707583 DOI: 10.1002/smll.202406439] [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: 07/28/2024] [Revised: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Over the past decade, precision medicine has garnered increasing attention, making significant strides in discovering new therapeutic drugs and mechanisms, resulting in notable achievements in symptom alleviation, pain reduction, and extended survival rates. However, the limited target specificity of primary drugs and inter-individual differences have often necessitated high-dosage strategies, leading to challenges such as restricted deep tissue penetration rates and systemic side effects. Material science advancements present a promising avenue for these issues. By leveraging the distinct internal features of diseased regions and the application of specific external stimuli, responsive materials can be tailored to achieve targeted delivery, controllable release, and specific biochemical reactions. This review aims to highlight the latest advancements in stimuli-responsive materials and their potential in precision medicine. Initially, we introduce disease-related internal stimuli and capable external stimuli, elucidating the reaction principles of responsive functional groups. Subsequently, we provide a detailed analysis of representative pre-clinical achievements of stimuli responsive materials across various clinical applications, including enhancements in the treatment of cancers, injury diseases, inflammatory diseases, infection diseases, and high-throughput microfluidic biosensors. Finally, we discuss some clinical challenges, such as off-target effects, long-term impacts of nano-materials, potential ethical concerns, and offer insights into future perspectives of stimuli-responsive materials.
Collapse
Affiliation(s)
- Ruixuan Zheng
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Chang Yu
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
- Intervention DepartmentThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Dan Yao
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Mengsi Cai
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Lexiang Zhang
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000China
| | - Fangfu Ye
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000China
- Beijing National Laboratory for Condensed Matter PhysicsInstitute of PhysicsChinese Academy of SciencesBeijing100190China
| | - Xiaoying Huang
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| |
Collapse
|
22
|
Alagha HZ, Gülsoy M. An In vitro Study on the Antibacterial Effect of a Combined Photodynamic and Sonodynamic Therapy Using IR780 Iodide-loaded Mesoporous Silica Nanoparticles Against P. aeruginosa and Multi-Drug Resistant P. aeruginosa. RECENT ADVANCES IN ANTI-INFECTIVE DRUG DISCOVERY 2025; 20:64-76. [PMID: 40302549 DOI: 10.2174/0127724344309438240529064221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/07/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2025]
Abstract
BACKGROUND The aim of this study was to investigate the antimicrobial photodynamic, sonodynamic, and combined photodynamic and sonodynamic potentials of IR780 iodide loaded mesoporous silica nanoparticles against gram-negative Pseudomonas aeruginosa (P. aeruginosa) and multi drug resistant Pseudomonas aeruginosa (MDR P. aeruginosa). METHODS IR780 iodide loaded mesoporous silica nanoparticles were synthesized, and their antimicrobial photodynamic and sonodynamic potentials against one P. aeruginosa strain, and one MDR P. aeruginosa strain were investigated. Laser irradiation was achieved via a 785 nm diode laser (500 mW/cm2, 5 min). Ultrasound irradiation was achieved via a 1-MHz ultrasound unit (1.5 W/cm2, 50% duty cycle, 3 min). Viable bacterial cells were counted by serial dilution method. Data were analyzed by ANOVA followed by Tukey's test (p ≤ 0.05). RESULTS The results revealed that for P. aeruginosa, the combined photodynamic therapy (PDT) and sonodynamic therapy (SDT) showed a 44% reduction in bacterial cell viability as compared to 18% and 31% when exposed to SDT alone and PDT alone, respectively. For MDR. P. aeruginosa, the combined treatment resulted in a 45% reduction in bacterial cell viability, as compared to 14% and 30% when exposed to SDT alone and PDT alone, respectively. The killing effect was mainly due to the photodynamic and sonodynamic effects of the nanoparticles, mainly caused by singlet oxygen. No photothermal effect was involved in the killing. CONCLUSION The results of this study demonstrated that IR780 iodide-loaded mesoporous silica nanoparticles have the potential to be utilized as photo/sono therapeutic agents for the inactivation of drug-resistant bacteria.
Collapse
Affiliation(s)
- Heba Z Alagha
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Murat Gülsoy
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| |
Collapse
|
23
|
Elbehiry A, Abalkhail A, Anajirih N, Alkhamisi F, Aldamegh M, Alramzi A, AlShaqi R, Alotaibi N, Aljuaid A, Alzahrani H, Alzaben F, Rawway M, Ibrahem M, Abdelsalam MH, Rizk NI, Mostafa MEA, Alfaqir MR, Edrees HM, Alqahtani M. Helicobacter pylori: Routes of Infection, Antimicrobial Resistance, and Alternative Therapies as a Means to Develop Infection Control. Diseases 2024; 12:311. [PMID: 39727641 PMCID: PMC11727528 DOI: 10.3390/diseases12120311] [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: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
Helicobacter pylori (H. pylori) is a Gram-negative, spiral-shaped bacterium that colonizes the gastric epithelium and is associated with a range of gastrointestinal disorders, exhibiting a global prevalence of approximately 50%. Despite the availability of treatment options, H. pylori frequently reemerges and demonstrates increasing antibiotic resistance, which diminishes the efficacy of conventional therapies. Consequently, it is imperative to explore non-antibiotic treatment alternatives to mitigate the inappropriate use of antibiotics. This review examines H. pylori infection, encompassing transmission pathways, treatment modalities, antibiotic resistance, and eradication strategies. Additionally, it discusses alternative therapeutic approaches such as probiotics, anti-biofilm agents, phytotherapy, phototherapy, phage therapy, lactoferrin therapy, and vaccine development. These strategies aim to reduce antimicrobial resistance and enhance treatment outcomes for H. pylori infections. While alternative therapies can maintain low bacterial levels, they do not achieve complete eradication of H. pylori. These therapies are designed to bolster the immune response, minimize side effects, and provide gastroprotective benefits, rendering them suitable for adjunctive use alongside conventional treatments. Probiotics may serve as adjunctive therapy for H. pylori; however, their effectiveness as a monotherapy is limited. Photodynamic and phage therapies exhibit potential in targeting H. pylori infections, including those caused by drug-resistant strains, without the use of antibiotics. The development of a reliable vaccine is also critical for the eradication of H. pylori. This review identifies candidate antigens such as VacA, CagA, and HspA, along with various vaccine formulations, including vector-based and subunit vaccines. Some vaccines have demonstrated efficacy in clinical trials, while others have shown robust immune protection in preclinical studies. Nevertheless, each of the aforementioned alternative therapies requires thorough preclinical and clinical evaluation to ascertain their efficacy, side effects, cost-effectiveness, and patient compliance.
Collapse
Affiliation(s)
- Ayman Elbehiry
- Department of Public Health, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia
| | - Adil Abalkhail
- Department of Public Health, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia
| | - Nuha Anajirih
- Medical Emergency Services Department, Faculty of Health Sciences, Umm Al-Qura University, Al-Qunfudah P.O. Box 1109, Saudi Arabia
| | - Fahad Alkhamisi
- Department of Preventive Medicine, King Fahad Armed Hospital, Jeddah 23311, Saudi Arabia
| | - Mohammed Aldamegh
- Pathology and Laboratory Medicine Department, Armed Forces Hospital-Jubail, Jubail 31951, Saudi Arabia
| | - Abdullah Alramzi
- Medical Radiology Department, Armed Forces Hospital-Jubail, Jubail 31951, Saudi Arabia
| | - Riyad AlShaqi
- Biomedical Engineer, Armed Forces Medical Services, Riyadh 12426, Saudi Arabia
| | - Naif Alotaibi
- Medical Hospital Administration Department, Armed Forces Hospital-Jubail, Jubail 31951, Saudi Arabia
| | - Abdullah Aljuaid
- Medical Hospital Administration Department, Armed Forces Hospitals in Al Kharj, AL Kharj 16278, Saudi Arabia
| | - Hilal Alzahrani
- Physical Medicine and Rehabilitation Department, Armed Forces Center for Health Rehabilitation, Taif 21944, Saudi Arabia
| | - Feras Alzaben
- Department of Food Service, King Fahad Armed Forces Hospital, Jeddah 23311, Saudi Arabia
| | - Mohammed Rawway
- Biology Department, College of Science, Jouf University, Sakaka 42421, Saudi Arabia
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt
| | - Mai Ibrahem
- Department of Public Health, College of Applied Medical Science, King Khalid University, Abha 61421, Saudi Arabia
| | - Moustafa H. Abdelsalam
- Department of Physiology, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Nermin I. Rizk
- Department of Physiology, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Mohamed E. A. Mostafa
- Department of Anatomy, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Moneef Rohail Alfaqir
- Department of Anatomy, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Husam M. Edrees
- Department of Physiology, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Mubarak Alqahtani
- Department of Radiology, King Fahd Armed Forces Hospital, Jeddah 23311, Saudi Arabia
| |
Collapse
|
24
|
Javanmard Z, Pourhajibagher M, Bahador A. Advancing Anti-Biofilm Strategies: Innovations to Combat Biofilm-Related Challenges and Enhance Efficacy. J Basic Microbiol 2024; 64:e2400271. [PMID: 39392011 DOI: 10.1002/jobm.202400271] [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: 05/11/2024] [Revised: 08/20/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024]
Abstract
Biofilms are complex communities of microorganisms that can cause significant challenges in various settings, including industrial processes, environmental systems, and human health. The protective nature of biofilms makes them resistant to traditional anti-biofilm strategies, such as chemical agents, mechanical interventions, and surface modifications. To address the limitations of conventional anti-biofilm methods, researchers have explored emerging strategies that encompass the use of natural compounds, nanotechnology-based methods, quorum-sensing inhibition, enzymatic degradation, and antimicrobial photodynamic/sonodynamic therapy. There is an increasing focus on combining multiple anti-biofilm strategies to combat resistance and enhance effectiveness. Researchers are continuously investigating the mechanisms of biofilm formation and developing innovative approaches to overcome the limitations of conventional anti-biofilm methods. These efforts aim to improve the management of biofilms and prevent infections while preserving the environment. This study provides a comprehensive overview of the latest advancements in anti-biofilm strategies. Given the dynamic nature of this field, exploring new approaches is essential to stimulate further research and development initiatives. The effective management of biofilms is crucial for maintaining the health of industrial processes, environmental systems, and human populations.
Collapse
Affiliation(s)
- Zahra Javanmard
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Bahador
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Zhang YS, Ke S, Hu X, Wang SY, Peng WQ, Qian XH, Tian LH, Wu HJ, Li BH, Zeng XT, Zhang LL. Enhancing wound healing through sonodynamic silver/barium titanate heterostructures-loading gelatin/PCL nanodressings. Int J Biol Macromol 2024; 283:137648. [PMID: 39547623 DOI: 10.1016/j.ijbiomac.2024.137648] [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: 05/22/2024] [Revised: 08/28/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Bacterial infections present a formidable challenge in surgical procedures, and are a major threat to wound healing. Sonodynamic therapy (SDT) is a non-invasive approach for fighting pathogens; however, it is hindered by the efficiency of sonosensitizers and effective antibacterial time. In this study, we developed a biocompatible nanodressing to improve the antibacterial efficacy and accelerate wound healing via SDT. Silver nanoparticles (NPs) were synthesized on tetragonal barium titanate (BTO) NPs to create an Ag@BTO heterostructure of sonosensitizers to improve their piezocatalytic activities, which were then incorporated into gelatin/polycaprolactone (PCL) to form Ag@BTO-gelatin/PCL nanofiber (ABT-gP NFs) dressings. The loading of Ag@BTO NPs resulted in ABT-gP NFs with better mechanical properties and excellent piezocatalysis, which produced reactive oxygen species and Ag+ to kill bacteria during ultrasound (US) irradiation. Additionally, nanodressing released moderate amounts of silver ions without US, prolonging the antibacterial time, while promoting fibroblast migration. This approach was effective in killing Gram-positive and Gram-negative bacteria (100 % and 90.8 %, respectively), promoting cell migration in vitro and accelerating wound healing without adverse effects in vivo. This study extends the potential applications of ultrasound-triggered nanodressing to the field of wound healing.
Collapse
Affiliation(s)
- Yu-Sen Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Urology, Zhongnan Hosptial of Wuhan University, Wuhan 430071, China
| | - Shuai Ke
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Urology, Zhongnan Hosptial of Wuhan University, Wuhan 430071, China
| | - Xiao Hu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Urology, Zhongnan Hosptial of Wuhan University, Wuhan 430071, China
| | - Shuang-Ying Wang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wan-Qi Peng
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xin-Hang Qian
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ling-Hui Tian
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hui-Jun Wu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Bing-Hui Li
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xian-Tao Zeng
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Urology, Zhongnan Hosptial of Wuhan University, Wuhan 430071, China.
| | - Ling-Ling Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
26
|
Yang F, Lv J, Ma W, Yang Y, Hu X, Yang Z. Engineering Sonosensitizer-Derived Nanotheranostics for Augmented Sonodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402669. [PMID: 38970544 DOI: 10.1002/smll.202402669] [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: 04/04/2024] [Revised: 06/13/2024] [Indexed: 07/08/2024]
Abstract
Sonodynamic therapy (SDT), featuring noninvasive, deeper penetration, low cost, and repeatability, is a promising therapy approach for deep-seated tumors. However, the general or only utilization of SDT shows low efficiency and unsatisfactory treatment outcomes due to the complicated tumor microenvironment (TME) and SDT process. To circumvent the issues, three feasible approaches for enhancing SDT-based therapeutic effects, including sonosensitizer optimization, strategies for conquering hypoxia TME, and combinational therapy are summarized, with a particular focus on the combination therapy of SDT with other therapy modalities, including chemodynamic therapy, photodynamic therapy, photothermal therapy, chemotherapy, starvation therapy, gas therapy, and immunotherapy. In the end, the current challenges in SDT-based therapy on tumors are discussed and feasible approaches for enhanced therapeutic effects are provided. It is envisioned that this review will provide new insight into the strategic design of high-efficiency sonosensitizer-derived nanotheranostics, thereby augmenting SDT and accelerating the potential clinical transformation.
Collapse
Affiliation(s)
- Fuhong Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Jingqi Lv
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Wen Ma
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Yanling Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Xiaoming Hu
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
- Jiangxi Key Laboratory of Nanobiomaterials, School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, China
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| |
Collapse
|
27
|
Huang J, Hong X, Chen S, He Y, Xie L, Gao F, Zhu C, Jin X, Yan H, Ye Y, Shao M, Du X, Feng G. Biomimetic Metal-Organic Framework Gated Nanoplatform for Sonodynamic Therapy against Extensively Drug Resistant Bacterial Lung Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402473. [PMID: 38962911 PMCID: PMC11434100 DOI: 10.1002/advs.202402473] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/03/2024] [Indexed: 07/05/2024]
Abstract
Novel antimicrobial strategies are urgently needed to treat extensively drug-resistant (XDR) bacterial infections due to the high mortality rate and lack of effective therapeutic agents. Herein, nanoengineered human umbilical cord mesenchymal stem cells (hUC-MSCs), named PMZMU, are designed as a sonosensitizer for synergistic sonodynamic-nano-antimicrobial therapy against gram-negative XDR bacteria. PMZMU is composed of a bacterial targeting peptide (UBI29-41) modified hUC-MSCs membrane (MSCm), a sonosensitizer meso-tetra(4-car-boxyphenyl) porphine doped mesoporous organo-silica nanoparticle and an acidity-responsive metal-organic framework ZIF-8. This innovative formulation enables efficient loading of polymyxin B, reduces off-target drug release, increases circulation and targeting efficacy, and generates reactive oxygen species upon ultrasound irradiation. PMZMU exhibits remarkable in vitro inhibitory activity against four XDR bacteria: Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa (PA), and Escherichia coli. Taking advantage of the bacterial targeting ability of UBI29-41 and the inflammatory chemotaxis of hUC-MSC, PMZMU can be precisely delivered to lung infection sites thereby augmenting polymyxin B concentration. PMZMU-mediated sonodynamic therapy significantly reduces bacterial burden, relieves inflammatory damage by promoting the polarization of macrophages toward M2 phenotype, and improves survival rates without introducing adverse events. Overall, this study offers promising strategies for treating deep-tissue XDR bacterial infections, and guides the design and optimization of biomimetic nanomedicine.
Collapse
Affiliation(s)
- Jianling Huang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xiuwen Hong
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Sixi Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Yucong He
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Lixu Xie
- Department of Pulmonary and Critical Care Medicine, Qi Lu Hospital of Shandong University, Wen hua xi Road 107#, Jinan, 250012, China
| | - Fenglin Gao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Chenghua Zhu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xiao Jin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Haihao Yan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Yongxia Ye
- Department of Radiology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, 210009, China
| | - Mingyue Shao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xingran Du
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China
| | - Ganzhu Feng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| |
Collapse
|
28
|
Pan Z, Dai C, Li W. Material-based treatment strategies against intraosseous implant biofilm infection. Biochem Biophys Rep 2024; 39:101764. [PMID: 39040541 PMCID: PMC11261528 DOI: 10.1016/j.bbrep.2024.101764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/22/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Implant-associated infections present a significant clinical obstacle for orthopedic practitioners, with bacterial biofilm formation serving as a pivotal factor in the initiation, progression, and management of such infections. Conventional approaches have proven inadequate in fully eradicating biofilm-related infections. Consequently, novel material-based therapeutic strategies have been developed, encompassing the utilization of antimicrobial agents, delivery vehicles, and synergistic antibacterial systems. In this review, we provide a succinct overview of recent advancements in anti-biofilm strategies, with the aim of offering insights that may aid in the treatment of intraosseous implant infections.
Collapse
Affiliation(s)
- Zhuoer Pan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
| | - Chengxin Dai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
| | - Weixu Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
| |
Collapse
|
29
|
Zhou J, Ji X, Wang H, Hsu JC, Hua C, Yang X, Liu Z, Guo H, Huang Y, Li Y, Cai W, Lin X, Ni D. Design of Ultrasound-Driven Charge Interference Therapy for Wound Infection. NANO LETTERS 2024; 24:7868-7878. [PMID: 38912706 PMCID: PMC11334693 DOI: 10.1021/acs.nanolett.4c00930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Wound infections, especially those caused by pathogenic bacteria, present a considerable public health concern due to associated complications and poor therapeutic outcomes. Herein, we developed antibacterial nanoparticles, namely, PGTP, by coordinating guanidine derivatives with a porphyrin-based sonosensitizer. The synthesized PGTP nanoparticles, characterized by their strong positive charge, effectively disrupted the bacterial biosynthesis process through charge interference, demonstrating efficacy against both Gram-negative and Gram-positive bacteria. Additionally, PGTP nanoparticles generated reactive oxygen species under ultrasound stimulation, resulting in the disruption of biofilm integrity and efficient elimination of pathogens. RNA-seq analysis unveiled the detailed mechanism of wound healing, revealing that PGTP nanoparticles, when coupled with ultrasound, impair bacterial metabolism by interfering with the synthesis and transcription of amino acids. This study presents a novel approach to combatting wound infections through ultrasound-driven charge-interfering therapy, facilitated by advanced antibacterial nanomaterials.
Collapse
Affiliation(s)
- Jingwei Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiuru Ji
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Han Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Chen Hua
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xi Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zeyang Liu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haiyan Guo
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying Huang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuhan Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Xiaoxi Lin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Dalong Ni
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou 215163, China
| |
Collapse
|
30
|
Wu X, Wang H, Xiong J, Yang GX, Hu JF, Zhu Q, Chen Z. Staphylococcus aureus biofilm: Formulation, regulatory, and emerging natural products-derived therapeutics. Biofilm 2024; 7:100175. [PMID: 38298832 PMCID: PMC10827693 DOI: 10.1016/j.bioflm.2023.100175] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 02/02/2024] Open
Abstract
Staphylococcus aureus can readily form biofilm which enhances the drug-resistance, resulting in life-threatening infections involving different organs. Biofilm formation occurs due to a series of developmental events including bacterial adhesion, aggregation, biofilm maturation, and dispersion, which are controlled by multiple regulatory systems. Rapidly increasing research and development outcomes on natural products targeting S. aureus biofilm formation and/or regulation led to an emergent application of active phytochemicals and combinations. This review aimed at providing an in-depth understanding of biofilm formation and regulation mechanisms for S. aureus, outlining the most important antibiofilm strategies and potential targets of natural products, and summarizing the latest progress in combating S. aureus biofilm with plant-derived natural products. These findings provided further evidence for novel antibiofilm drugs research and clinical therapies.
Collapse
Affiliation(s)
- Xiying Wu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
- Institute of Natural Medicine and Health Products, School of Pharmaceutical Sciences, Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Taizhou University, Zhejiang, 318000, China
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Huan Wang
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Juan Xiong
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Guo-Xun Yang
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jin-Feng Hu
- Institute of Natural Medicine and Health Products, School of Pharmaceutical Sciences, Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Taizhou University, Zhejiang, 318000, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
| |
Collapse
|
31
|
Liu X, Peng S, Pei Y, Huo Y, Zong Y, Ren J, Zhao J. Facile fabrication of chitosan/hyaluronic acid hydrogel-based wound closure material Co-loaded with gold nanoparticles and fibroblast growth factor to improve anti-microbial and healing efficiency in diabetic wound healing and nursing care. Regen Ther 2024; 26:1018-1029. [PMID: 39553541 PMCID: PMC11565426 DOI: 10.1016/j.reth.2024.10.003] [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: 08/13/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024] Open
Abstract
Generally, diabetic wounds heal very slowly and inefficiently with an increasing risk of infections. Recent nanotechnology and biomaterial advances elaborate developed multi-functional hydrogels and nanoparticles offer promising solutions to accelerate wound healing for diabetic patients. This research work demonstrates to use of solvent diffusion method to develop hydrogel nanocomposites composed of chitosan (CS), hyaluronic acid (HA), gold (Au), and fibroblast growth factors (FGF). The biological analysis of nanocomposites exhibited enhanced wound healing efficiency by incorporating bioactive molecules like FGF and bioactive Au nanoparticles. In vitro, cell compatibility analysis (MTT assay) of prepared hydrogel nanocomposites was studied on fibroblast cell lines NIH-3T3-L1 and L929 and exhibited greater cell survival ability (>90 %), cell proliferation and migration ability, which demonstrated the suitability of nanocomposite for wound healing treatment. In vitro, anti-bacterial analyses established that FGF-Au@CS/HA has strong antibacterial effectiveness against gram-positive and gram-negative pathogens. The observation of the present research revealed that prepared FGF-Au@CS/HA hydrogel composites could be a suitable biomaterial for diabetic wound care, potentially improving its antibacterial and healing efficacies.
Collapse
Affiliation(s)
- Xin Liu
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Shengwei Peng
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yongju Pei
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yuanyuan Huo
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yadi Zong
- Department of Pediatric Surgery, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Jianwei Ren
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Jing Zhao
- Department of Respiratory Intensive Care Unit, Henan Provincial Key Medicine Laboratory of Nursing, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| |
Collapse
|
32
|
Tsikouras P, Oikonomou E, Nikolettos K, Andreou S, Kyriakou D, Damaskos C, Garmpis N, Monastiridou V, Nalmpanti T, Bothou A, Iatrakis G, Nikolettos N. The Impact of Periodontal Disease on Preterm Birth and Preeclampsia. J Pers Med 2024; 14:345. [PMID: 38672972 PMCID: PMC11051368 DOI: 10.3390/jpm14040345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/19/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
This review delves into the possible connection between periodontitis and negative pregnancy outcomes, such as preeclampsia and preterm birth. It highlights the potential influence of an unidentified microbial factor on preeclampsia and the effects of inflammatory responses on the rate of preterm births. Furthermore, it underscores the prevalent occurrence of oral ailments within the populace and their significant repercussions on quality of life. Hormonal fluctuations during pregnancy may exacerbate oral conditions such as pregnancy gingivitis and periodontitis, necessitating bespoke therapeutic approaches that take into account potential fetal ramifications. Periodontal disease, characterized by microbial attack and inflammatory response, results in tissue destruction and tooth loss. The oral cavity's susceptibility to bacterial colonization, which is primarily due to its role as a site for food intake, is highlighted. Furthermore, research indicates a correlation between inflammatory responses and factors such as prostaglandin E2 and IL-1β, and preterm birth. Therapeutic interventions are a focus of international research, with efforts being aimed at optimizing outcomes through larger studies involving pregnant women.
Collapse
Affiliation(s)
- Panagiotis Tsikouras
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.O.); (K.N.); (S.A.); (D.K.); (V.M.); (T.N.); (N.N.)
| | - Efthymios Oikonomou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.O.); (K.N.); (S.A.); (D.K.); (V.M.); (T.N.); (N.N.)
| | - Konstantinos Nikolettos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.O.); (K.N.); (S.A.); (D.K.); (V.M.); (T.N.); (N.N.)
| | - Sotiris Andreou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.O.); (K.N.); (S.A.); (D.K.); (V.M.); (T.N.); (N.N.)
| | - Dimitrios Kyriakou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.O.); (K.N.); (S.A.); (D.K.); (V.M.); (T.N.); (N.N.)
| | - Christos Damaskos
- Department of Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Renal Transplantation Unit, Laiko General Hospital, 11527 Athens, Greece
| | | | - Vassiliki Monastiridou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.O.); (K.N.); (S.A.); (D.K.); (V.M.); (T.N.); (N.N.)
| | - Theopi Nalmpanti
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.O.); (K.N.); (S.A.); (D.K.); (V.M.); (T.N.); (N.N.)
| | - Anastasia Bothou
- Neonatal Department, University Hospital Alexandra, 11528 Athens, Greece;
- Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - George Iatrakis
- Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 11528 Athens, Greece;
- Department of Obstetrics and Gynecology, Rea Maternity Hospital, 17564 Athens, Greece
| | - Nikolaos Nikolettos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.O.); (K.N.); (S.A.); (D.K.); (V.M.); (T.N.); (N.N.)
| |
Collapse
|