1
|
Yan X, Lin Z, Shen H, Chen Y, Chen L. Photo-responsive antibacterial metal organic frameworks. J Mater Chem B 2025. [PMID: 40370037 DOI: 10.1039/d5tb00105f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
The misuse and overuse of antibiotics have caused the emergence of antibiotic-resistant bacteria, making bacterial infections more challenging. The increasing prevalence of multidrug-resistant pathogens has driven researchers to explore novel therapeutic strategies. Phototherapy strategies that utilize photo-responsive biomaterials for their antibacterial properties have gained widespread attention due to their capability of precisely controlling bacterial inactivation with minimal side effects. Despite their potential, photodynamic therapies suffer from phototoxicity and low efficiency of photosensitizers, while photothermal therapy risks overheating, which may harm healthy tissues, thus restricting its broader application. Metal organic frameworks (MOFs) have unique physicochemical properties, which provide a promising way to deal with these challenges. MOFs can function as reservoirs, loading and releasing antibacterial agents, such as antibiotics or metal ions, upon light illumination by virtue of their metastable coordination bonds. Their porous structures enable controlled drug release and encapsulation of photosensitizers. Furthermore, MOFs' tunable composition and pore structure allow for the light-triggered generation of heat and reactive oxygen species, enhancing their antibacterial effectiveness. By doping MOFs with functional materials, it is possible to achieve multi-mode antibacterial effects. In this review, we will outline recent advancements of photo-responsive antibacterial MOFs, categorize their underlying mechanisms of action and highlight their prospects in addressing bacterial resistance.
Collapse
Affiliation(s)
- Xiaojie Yan
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Zhengzheng Lin
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - He Shen
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Yu Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Liang Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, P. R. China
| |
Collapse
|
2
|
Chen M, Liu T, Wang X, Gao L, Cheng Y, Jiang J, Zhang J. Comprehensive wound healing using ETN@Fe 7S 8 complex by positively regulating multiple programmed phases. J Nanobiotechnology 2025; 23:342. [PMID: 40355866 PMCID: PMC12070563 DOI: 10.1186/s12951-025-03396-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Wound healing requires coordinated progression through multiple programmed phases including hemostasis, infection control, inflammatory resolution, proliferation, and tissue remodeling. Many nanomaterials have shown great potential to promote wound healing, however, most of them only address partial aspects of these processes, making a recovery hard with adequate effects. In this study, we prepared a complex of nano-iron sulfide integrated with erythrocyte-templated nanozyme (ETN) (ETN@Fe7S8) for comprehensive treatment of wounds. Firstly, ETN served as a mediator to confine iron sulfide to form Fe7S8 nanocomposite in a solvothermal reaction. Secondly, the ETN@Fe7S8 demonstrated bactericidal effects against methicillin-resistant Staphylococcus aureus (MRSA) by releasing ferrous iron and polysulfide to induce ferroptosis-like cell death. Thirdly, ferrous iron along with polysulfide exerted anti-inflammatory effects by inhibiting the activation of the NF-κB signaling pathway, while the polysulfide also contributed to angiogenesis by promoting the activation of vascular endothelial growth factor A (VEGFA), initiated phosphorylation-mediated activation of the PI3K/AKT signaling pathway, a master regulatory cascade governing endothelial cell survival, migration, and angiogenesis. When employed for wound, ETN@Fe7S8 showed the ability to prevent infection, reduce inflammation, promote angiogenesis, enhance cell proliferation, and remodel keratinocytes. Along with the hemostatic effect, ETN@Fe7S8 thus performed comprehensive effects for wound healing in the whole recovery stages. Therefore, our findings provide a multifunctional candidate of ETN and nano-iron sulfide complex which is capable of regulating and promoting wound healing.
Collapse
Affiliation(s)
- Mengxia Chen
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
- School of Life Sciences, Jilin Normal University, Jilin, 136000, China
| | - Ting Liu
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
- School of Life Science and Technology, Jinan University, Guangdong, 510632, China
| | - Xiaonan Wang
- Key Laboratory of Biomacromolecules, Institute of Biophysics, CAS Engineering Laboratory for Nanozyme, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Lizeng Gao
- Key Laboratory of Biomacromolecules, Institute of Biophysics, CAS Engineering Laboratory for Nanozyme, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yunqing Cheng
- School of Life Sciences, Jilin Normal University, Jilin, 136000, China.
| | - Jing Jiang
- Key Laboratory of Biomacromolecules, Institute of Biophysics, CAS Engineering Laboratory for Nanozyme, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China.
| |
Collapse
|
3
|
Wu C, You Y, Yu D, Zhu YX, Lin H, Shi J. Selenide-Driven Reactive Oxygen Species Activation and Fe(II) Regeneration for Enhanced Nanocatalytic Antibacterial Therapeutics. Adv Healthc Mater 2025:e2501021. [PMID: 40344503 DOI: 10.1002/adhm.202501021] [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: 03/05/2025] [Revised: 04/13/2025] [Indexed: 05/11/2025]
Abstract
Fenton-based nanocatalytic therapy has attracted widespread attention for its high efficiency and safety. Nevertheless, Fe2+ regeneration, as the rate-limiting step of Fenton reaction, hinders the ROS-induced oxidative killing. Herein, a Fe2+ auto-regeneration strategy is exemplified by 2D FeSe2 nanosheets to break the rate limitation of Fenton reaction and subsequently enhances the antibacterial oxidative damage via dual ROS generation pathways. To be specific, the Se species accelerate the Fe3+ reduction to maintain high ·OH productivity of Fe2+-mediated Fenton reaction, which is accompanied by the production of H2Se in the presence of H+. The H2Se further converts O2 into O2 ·- and synergistically breaks the oxidative threshold of bacteria, leading to irreversible bacterial death with glutathione depletion, lipid peroxidation, and membrane destruction. In summary, the FeSe2-mediated Fe2+ auto-regeneration and ROS self-production pathways largely elevate its oxidative killing capability, providing a potential ROS enhancement strategy for broad-spectrum nonantibiotic bacterial disinfection.
Collapse
Affiliation(s)
- Chenyao Wu
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, P. R. China
| | - Yanling You
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, P. R. China
| | - Dehong Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Ya-Xuan Zhu
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Han Lin
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, P. R. China
| | - Jianlin Shi
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
4
|
Wang X, Zhou N, Gao XJ, Zhu Z, Sun M, Wang Q, Cao H, Wu X, Zhou C, Zheng Q, Yuan Y, Liu Y, Chen L, Jiang J, Bu P, Gao L. Selective G6PDH inactivation for Helicobacter pylori eradication with transformed polysulfide. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1158-1173. [PMID: 39821832 DOI: 10.1007/s11427-024-2775-3] [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: 08/01/2024] [Accepted: 11/07/2024] [Indexed: 01/19/2025]
Abstract
Alternative treatment for the highly prevalent Helicobacter pylori infection is imperative due to rising antibiotic resistance. We unexpectedly discovered that the anti-H. pylori component in garlic is hydrogen polysulfide (H2Sn, n⩾2), not organic polysulfides. Studies on the mechanism of action (MoA) show that H2Sn specifically inactivates H. pylori glucose-6-phosphate dehydrogenase (G6PDH) by interfering with electron transfer from glucose-6-phosphate (G6P) to nicotinamide adenine dinucleotide phosphate (NADP+). However, low H2Sn yield makes garlic derivatives hard to be a reliable donor of H2Sn to treat H. pylori infection. To address this challenge, we established a polysulfide transformation process from garlic organosulfur compounds into Fe3S4 that generates H2Sn with a 25-58 times increase in yield. Through chitosan encapsulation, we designed a gastric-adaptive H2Sn microreactor (GAPSR) that eradicates H. pylori with 250 times higher efficiency under gastric conditions. A single GAPSR achieves more rapid H. pylori eradication than combined antibiotics therapy without disturbing the gut microbiota. These findings indicate a distinct MoA transformation mediated by polysulfide as an alternative candidate to treat H. pylori infection.
Collapse
Affiliation(s)
- Xiaonan Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ning Zhou
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuejiao J Gao
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Zijing Zhu
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Minmin Sun
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qian Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Haolin Cao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuetong Wu
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Caiyu Zhou
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Qingkang Zheng
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ye Yuan
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuan Liu
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lei Chen
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Pengcheng Bu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China.
| |
Collapse
|
5
|
Corona-España AM, García-Ramírez MA, Rodríguez-Buenfil IM, Delgado-Saucedo JI, González-Reynoso O. Synthesis Mechanism and Therapeutic Effects of Thiosulfinates and Polysulfides of Different Species of Garlic from the Allium Genus. Pharmaceutics 2025; 17:437. [PMID: 40284432 PMCID: PMC12030139 DOI: 10.3390/pharmaceutics17040437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
The genus Allium contains more than 300 species where garlic, onion and leek can be found. Recent studies highlight the potential of phytochemicals present in the genus Allium as therapeutic agents, such as antimicrobial, antihypertensive, antioxidant and antiinflammatory, which makes it a widely studied genus and an attractive option for both the pharmaceutical and food industries. This review aims to explore the current knowledge in this field. It highlights key findings regarding the pharmacological approach on thiosulfinates and polysulfides for Allium sativum, Allium ampeloprasum and Allium sphaerocephalon species. Further, by integrating what has been reported in previous research, this review proposes an action mechanism for the formation of thiosulfinates and polysulfides, which will help harness the therapeutic potential of nature-inspired solutions in combating illness.
Collapse
Affiliation(s)
- Ana Montserrat Corona-España
- Department of Chemistry, University Center of Applied Sciences and Engineering, University of Guadalajara, Guadalajara 44430, Mexico;
| | - Mario Alberto García-Ramírez
- Department of Electro-Photonic Engineering, University Center of Applied Sciences and Engineering, University of Guadalajara, Guadalajara 44430, Mexico;
| | - Ingrid Mayanin Rodríguez-Buenfil
- Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C Southeast Sub-Headquarters, Mérida 97070, Mexico;
| | - Jorge Iván Delgado-Saucedo
- Department of Pharmacobiology, University Center of Applied Sciences and Engineering, University of Guadalajara, Guadalajara 44430, Mexico;
| | - Orfil González-Reynoso
- Metabolic Engineering and Bioinformatics Laboratory, Department of Chemical Engineering, University Center of Applied Sciences and Engineering, University of Guadalajara, Guadalajara 44430, Mexico
| |
Collapse
|
6
|
He S, Chen Y, Lian H, Cao X, Liu B, Wei X. Self-Assembled DNA/SG-I Nanoflower: Versatile Photocatalytic Biosensors for Disease-Related Markers. Anal Chem 2025; 97:4350-4358. [PMID: 39973756 DOI: 10.1021/acs.analchem.4c04772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
DNA nanostructures have recently attracted more attention with functionalities, programmability, and biocompatibility. Herein, a novel self-assembled photocatalytic DNA/SYBR Green I (SG-I) nanoflower (DSNF) was successfully synthesized by rolling circle amplification. DSNF was self-assembled through liquid crystallization of a high concentration of DNA in the RCA products, without relying on the Watson-Crick base-pairing principle. Interestingly, DSNF not only possessed a larger specific surface area and good stability but also exhibited excellent photocatalytic activity that generates singlet oxygen and superoxide anion to oxidate 3,3',5,5'-tetramethylbenzidine. Meanwhile, the photocatalytic DSNF combined with an enzyme-linked immunosorbent assay to develop a new colorimetric sensor for highly specific, sensitive, and visual detection of carcinoembryonic antigens (CEAs). The colorimetric sensor achieved sensitive and low-cost quantitative detection of CEA in the linear range of 0.5-80.0 ng/mL, and the LOD was 0.5 ng/mL. In addition, three negative and seven positive clinical serum samples of CEA were obtained with 100% accuracy using the proposed colorimetric sensor, showing great potential in the clinical application of cancer diagnosis. We envision that this photocatalytic DSNF is expected to provide important perspectives in fluorescence imaging, photosensitizing cancer therapy, and clinical diagnosis fields.
Collapse
Affiliation(s)
- Shan He
- College of Materials Science and Engineering, Huaqiao University, Xiamen 361021, China
- School of Resources and Civil Engineering, Gannan University of Science and Technology, Ganzhou 341000, China
| | - Yiyu Chen
- College of Materials Science and Engineering, Huaqiao University, Xiamen 361021, China
| | - Huiting Lian
- College of Materials Science and Engineering, Huaqiao University, Xiamen 361021, China
- Key Laboratory of Molecular Designing and Green Conversions, Huaqiao University, Xiamen 361021, China
| | - Xuegong Cao
- College of Materials Science and Engineering, Huaqiao University, Xiamen 361021, China
- Key Laboratory of Molecular Designing and Green Conversions, Huaqiao University, Xiamen 361021, China
| | - Bin Liu
- College of Materials Science and Engineering, Huaqiao University, Xiamen 361021, China
- Key Laboratory of Molecular Designing and Green Conversions, Huaqiao University, Xiamen 361021, China
- Xiamen Key Laboratory of Optoelectronic Materials and Advanced Manufacturing, Huaqiao University, Xiamen 361021, China
| | - Xiaofeng Wei
- College of Materials Science and Engineering, Huaqiao University, Xiamen 361021, China
- Key Laboratory of Molecular Designing and Green Conversions, Huaqiao University, Xiamen 361021, China
- Xiamen Key Laboratory of Optoelectronic Materials and Advanced Manufacturing, Huaqiao University, Xiamen 361021, China
| |
Collapse
|
7
|
Fan L, Shen Y, Lou D, Gu N. Progress in the Computer-Aided Analysis in Multiple Aspects of Nanocatalysis Research. Adv Healthc Mater 2025; 14:e2401576. [PMID: 38936401 DOI: 10.1002/adhm.202401576] [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: 04/29/2024] [Revised: 06/08/2024] [Indexed: 06/29/2024]
Abstract
Making the utmost of the differences and advantages of multiple disciplines, interdisciplinary integration breaks the science boundaries and accelerates the progress in mutual quests. As an organic connection of material science, enzymology, and biomedicine, nanozyme-related research is further supported by computer technology, which injects in new vitality, and contributes to in-depth understanding, unprecedented insights, and broadened application possibilities. Utilizing computer-aided first-principles method, high-speed and high-throughput mathematic, physic, and chemic models are introduced to perform atomic-level kinetic analysis for nanocatalytic reaction process, and theoretically illustrate the underlying nanozymetic mechanism and structure-function relationship. On this basis, nanozymes with desirable properties can be designed and demand-oriented synthesized without repeated trial-and-error experiments. Besides that, computational analysis and device also play an indispensable role in nanozyme-based detecting methods to realize automatic readouts with improved accuracy and reproducibility. Here, this work focuses on the crossing of nanocatalysis research and computational technology, to inspire the research in computer-aided analysis in nanozyme field to a greater extent.
Collapse
Affiliation(s)
- Lin Fan
- Medical School of Nanjing University, Nanjing, 210093, P. R. China
- School of Integrated Circuit Science and Engineering (Industry-Education Integration School), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| | - Yilei Shen
- School of Integrated Circuit Science and Engineering (Industry-Education Integration School), Nanjing University of Posts and Telecommunications, Nanjing, 210023, P. R. China
| | - Doudou Lou
- Nanjing Institute for Food and Drug Control, Nanjing, 211198, P. R. China
| | - Ning Gu
- Medical School of Nanjing University, Nanjing, 210093, P. R. China
| |
Collapse
|
8
|
Liang Z, Liang Z, Hu H, Howell K, Fang Z, Zhang P. Food substances alter gut resistome: Mechanisms, health impacts, and food components. Compr Rev Food Sci Food Saf 2025; 24:e70143. [PMID: 40047321 PMCID: PMC11884230 DOI: 10.1111/1541-4337.70143] [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: 09/28/2024] [Revised: 01/13/2025] [Accepted: 02/02/2025] [Indexed: 03/09/2025]
Abstract
Antibiotics are effective in treating bacterial infections, but their widespread use has spurred antibiotic resistance, which is linked closely with human disease. While dietary components are known to influence the gut microbiome, specific effects on the gut resistome-the collection of antibiotic-resistant genes in the gut-remain underexplored. This review outlines the mechanisms of antibiotic action and the development of resistance, emphasizing the connection between the gut resistome and human diseases such as metabolic disorders, cardiovascular disease, liver disease, and nervous system disorders. It also discusses the effects of diet habits and dietary components, including bioactive macronutrients, phytochemicals, and probiotics, on the composition of the gut resistome by enhancing antibiotic efficacy and potentially reducing resistance. This review highlights the emerging trend of increasing interest in functional foods aimed at targeting the gut resistome and a growing focus on bioactive plant compounds with the potential to modulate antibiotic resistance.
Collapse
Affiliation(s)
- Ze Liang
- School of Agriculture, Food and Ecosystem Sciences, Faculty of ScienceThe University of MelbourneParkvilleVictoriaAustralia
| | - Zijian Liang
- School of Agriculture, Food and Ecosystem Sciences, Faculty of ScienceThe University of MelbourneParkvilleVictoriaAustralia
| | - Hang‐Wei Hu
- School of Agriculture, Food and Ecosystem Sciences, Faculty of ScienceThe University of MelbourneParkvilleVictoriaAustralia
| | - Kate Howell
- School of Agriculture, Food and Ecosystem Sciences, Faculty of ScienceThe University of MelbourneParkvilleVictoriaAustralia
| | - Zhongxiang Fang
- School of Agriculture, Food and Ecosystem Sciences, Faculty of ScienceThe University of MelbourneParkvilleVictoriaAustralia
| | - Pangzhen Zhang
- School of Agriculture, Food and Ecosystem Sciences, Faculty of ScienceThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
9
|
Sead FF, Jain V, Kumar A, M M R, Kundlas M, Gupta S, Kumari M, Kazemi M, Javahershenas R. Magnetically recoverable catalysts for efficient multicomponent synthesis of organosulfur compounds. RSC Adv 2025; 15:3928-3953. [PMID: 39917045 PMCID: PMC11799890 DOI: 10.1039/d4ra08769k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/22/2025] [Indexed: 02/09/2025] Open
Abstract
This manuscript introduces a groundbreaking study on the development and application of magnetically recoverable catalysts for the efficient multicomponent synthesis of organosulfur compounds. Capitalizing on the unique advantages of magnetic recovery, these catalysts streamline the synthesis process, offering an innovative solution that marries efficiency with environmental sustainability. By facilitating the multicomponent reaction of key precursors in the presence of sulfur sources, the catalysts enable the straightforward synthesis of various valuable organosulfur compounds, crucial in numerous pharmaceutical, agricultural, and material science applications. Key findings demonstrate a significant enhancement in reaction yields and selectivity and the remarkable ease with which the catalysts can be recovered and reused, thereby reducing both waste and operational costs. Magnetic catalysts, often based on magnetic iron nanoparticles, facilitate rapid and efficient reactions under mild conditions, offering superior atom economy, reduced solvent use, and the potential for scalable processes. Additionally, magnetically separating the catalysts from the reaction mixture enables multiple recycling cycles, reducing waste and operational costs. The review also discusses the mechanistic insights, challenges, and recent advancements in this field alongside future directions for developing more robust and versatile magnetic catalytic systems. This research embodies a significant step forward in the field of catalysis, highlighting the potential of magnetically recoverable catalysts to revolutionize the synthesis of complex molecules. Future perspectives discussed in the manuscript focus on expanding the scope of these catalysts to broader applications, optimizing catalyst design for enhanced performance, and further aligning chemical synthesis processes with the principles of green chemistry. This review covers the literature from 2010 to the end of 2024, and it encompasses the different one-pot protocols for synthesizing various heterocyclic organosulfur compounds based on magnetically recoverable catalysts.
Collapse
Affiliation(s)
- Fadhil Faez Sead
- Department of Dentistry, College of Dentistry, The Islamic University Najaf Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah Al Diwaniyah Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon Babylon Iraq
| | - Vicky Jain
- Marwadi University Research Center, Department of Chemistry, Faculty of Science, Marwadi University Rajkot-360003 Gujarat India
| | - Anjan Kumar
- Department of Electronics and Communication Engineering, GLA University Mathura-281406 India
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University) Bangalore Karnataka India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University Rajpura 140401 Punjab India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri Mohali 140307 Punjab India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan Jaipur India
| | - Mosstafa Kazemi
- Young Researchers and Elite Club, Tehran Branch, Islamic Azad University Tehran Iran
| | | |
Collapse
|
10
|
Yuan X, He X, Fan J, Tai Y, Yao Y, Luo Y, Chen J, Luo H, Zhou X, Luo F, Niu Q, Hu WW, Sun X, Ying B. Advances in nanozymes with peroxidase-like activity for biosensing and disease therapy applications. J Mater Chem B 2025; 13:1599-1618. [PMID: 39751853 DOI: 10.1039/d4tb02315c] [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: 01/04/2025]
Abstract
Natural enzymes are crucial in biological systems and widely used in biomedicine, but their disadvantages, such as insufficient stability and high cost, have limited their widespread application. Since discovering the enzyme-like activity of Fe3O4 nanoparticles, extensive research progress in diverse nanozymes has been made with their in-depth investigation, resulting in rapid development of related nanotechnologies. Nanozymes can compensate for the defects of natural enzymes and show higher stability with lower costs. Among them, peroxidase (POD)-like nanozymes have attracted extensive attention in biomedical applications owing to their efficient catalytic performance and diverse structures. This review explores different types of nanozymes with POD-like activity and discusses their activity regulation, particularly emphasizing their latest development trends and advances in biosensing and disease treatment. Finally, the challenges and prospects for the development of POD-like nanozymes and their potential future applications in the biomedical field are also provided.
Collapse
Affiliation(s)
- Xiaohua Yuan
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xun He
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jiwen Fan
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yunze Tai
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yongchao Yao
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Luo
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jie Chen
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Han Luo
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xingli Zhou
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Fengming Luo
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qian Niu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenchuang Walter Hu
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xuping Sun
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, Shandong, China
| | - Binwu Ying
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
11
|
Wang H, Wang X, Mao M, Chen X, Han Z, Xun Z, Wang Q, Qi Y, Zhao W, Li T, Yan X, Liu J, Gao L, Xue X. Oral iron sulfide prevents acute alcohol intoxication by initiating the endogenous multienzymatic antioxidant defense system. SCIENCE ADVANCES 2025; 11:eadr4231. [PMID: 39823343 PMCID: PMC11740971 DOI: 10.1126/sciadv.adr4231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Acute alcohol intoxication could cause multiorgan damage, including nervous, digestive, and cardiovascular systems, and in particular, irreversible damage to the brain and liver. Emerging studies have revealed that the endogenous multienzymatic antioxidant defense system (MEAODS) plays a central role in preventing oxidative stress and other toxicological compounds produced by alcohol. However, few available drugs could quickly regulate MEAODS. Herein, we report a nanosized iron sulfide (nFeS) that can rapidly release polysulfide species in gastric juice. The released hydrogen polysulfide activates the Keap1/Nrf2 pathway via S-persulfidation of cysteine residues in Keap1, which promotes the expression of antioxidant enzymes and glutathione synthesis-related enzymes, thus potentiating MEAODS. Results indicate that the activated MEAODS not only alleviates oxidative stress and inflammation in the brain and liver but also mitigates movement dysfunction after only 2.5 hours of oral nFeS treatment. Collectively, this study provides a MEAODS-regulated strategy with nFeS and may aid the prevention of acute alcoholic injury.
Collapse
Affiliation(s)
- Heping Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People’s Republic of China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, P. R. China
| | - Xiaonan Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Mingxing Mao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People’s Republic of China
| | - Xi Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People’s Republic of China
| | - Ziwei Han
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People’s Republic of China
| | - Zengyu Xun
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People’s Republic of China
| | - Qian Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yilin Qi
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People’s Republic of China
| | - Weitao Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People’s Republic of China
| | - Tianqi Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People’s Republic of China
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, P. R. China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xue Xue
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People’s Republic of China
| |
Collapse
|
12
|
Duan X, Xu K, Zhang M, Xia Y, Wang L, Chen B, Wang C, Wei S, Zhou L. Crystal Form-Dependent MnS for Diabetic Wound Healing: Performance and Mechanistic Insights. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402496. [PMID: 39402776 DOI: 10.1002/smll.202402496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/14/2024] [Indexed: 12/20/2024]
Abstract
In pharmaceuticals, the structural and functional alterations induced by biotransformation are well-documented. Many pharmaceuticals exist in various crystal forms, which govern their transformation and significantly impact their activity. However, in the field of inorganic nanomedicine, there is a paucity of research focusing on the influence of crystal form-dependent "metabolism" (transformation) on their activity and biomechanism. This study delves into the distinct performances of two crystal forms of manganese sulfide (MnS), namely α-MnS and γ-MnS, in bacteria-infected diabetic wound healing. In the initial stage of a wound, where the environment is neutral to slightly alkaline, MnS partially converts to MnxOy (comprising Mn2O3 and Mn3O4) and concurrently produces hydrogen sulfide (H2S); the conversion efficiency of γ-MnS significantly surpasses that of α-MnS. Additionally, γ-MnS is more soluble than α-MnS, which allows it to generate more Mn2+. These components collectively contribute to the superior bacteriostatic properties of MnS. In wound related cells, MnS stimulates the production of collagen I and vascular endothelial growth factor (VEGF), promote the M1 macrophages polarizing to the M2 phenotype, for extracellular matrix (ECM) remodeling. Notably, different transformation products have distinct functions. Consequently, the activity of MnS is dependent on its original crystal form related solubility and transformation efficiency.
Collapse
Affiliation(s)
- Xiaomeng Duan
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Bio-functional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Nanjing Normal University, Nanjing, 210023, China
| | - Kaikai Xu
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Bio-functional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Nanjing Normal University, Nanjing, 210023, China
| | - Mingzhu Zhang
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Bio-functional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Nanjing Normal University, Nanjing, 210023, China
| | - Yuanyuan Xia
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Bio-functional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Nanjing Normal University, Nanjing, 210023, China
| | - Liping Wang
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Bio-functional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Nanjing Normal University, Nanjing, 210023, China
| | - Bingbing Chen
- Department of Energy Science and Engineering, Nanjing Tech University, Nanjing, Jiangsu, 210009, China
| | - Chongchong Wang
- School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Shaohua Wei
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Bio-functional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Nanjing Normal University, Nanjing, 210023, China
| | - Lin Zhou
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Bio-functional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
13
|
Huang L, Pu H, Sun DW. Spatiotemporally Guided Single-Atom Bionanozyme for Targeted Antibiofilm Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2407747. [PMID: 39370579 DOI: 10.1002/smll.202407747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Indexed: 10/08/2024]
Abstract
The heterogeneous and dynamic microenvironment of biofilms complicates bacterial infection treatment. Nanozyme catalytic therapy has recently been promising in treating biofilm infections. However, active nanozymes designed with the required precision targeting the biofilm microenvironment are lacking. This work proposes a spatiotemporally guided single-atom bionanozyme (BioSAzyme) for targeted antibiofilm therapy based on protein engineering of copper single-atom nanozyme (Cu SAzyme). The Cu SAzyme, synthesized via a novel mechanochemistry-assisted method, features highly accessible Cu-N4 active sites exposed on 2D N-doped carbon, exhibiting excellent triple enzyme-like activities according to experimental results and density functional theory calculations. Inheriting biofunctionality from both glucose oxidase and concanavalin A, BioSAzyme can localize the biofilm glycocalyx and catalyze endogenous glucose into H₂O₂ and gluconic acid, thus triggering multiplex cascade reactions with pH self-adaption to consume glucose and glutathione and generate •OH radicals. This spatiotemporally guided bionanocatalytic agent effectively inhibits E. coli O157: H7 and methicillin-resistant S. aureus biofilms in vitro and in vivo. Taking together, this work opens up new avenues for the rational design of single-atom nanozymes for precise antibiofilm therapy.
Collapse
Affiliation(s)
- Lunjie Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, 510006, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou, 510006, China
| | - Hongbin Pu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, 510006, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou, 510006, China
| | - Da-Wen Sun
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou, 510006, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou, 510006, China
- Food Refrigeration and Computerized Food Technology (FRCFT), Agriculture and Food Science Centre, University College Dublin, National University of Ireland, Belfield, Dublin, D04 V1W8, Ireland
| |
Collapse
|
14
|
Zhou S, Liu D, Fan K, Liu H, Zhang XD. Atomic-level design of biomimetic iron-sulfur clusters for biocatalysis. NANOSCALE 2024; 16:18644-18665. [PMID: 39257356 DOI: 10.1039/d4nr02883j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Designing biomimetic materials with high activity and customized biological functions by mimicking the central structure of biomolecules has become an important avenue for the development of medical materials. As an essential electron carrier, the iron-sulfur (Fe-S) clusters have the advantages of simple structure and high electron transport capacity. To rationally design and accurately construct functional materials, it is crucial to clarify the electronic structure and conformational relationships of Fe-S clusters. However, due to the complex catalytic mechanism and synthetic process in vitro, it is hard to reveal the structure-activity relationship of Fe-S clusters accurately. This review introduces the main structural types of Fe-S clusters and their catalytic mechanisms first. Then, several typical structural design strategies of biomimetic Fe-S clusters are systematically introduced. Furthermore, the development of Fe-S clusters in the biocatalytic field is enumerated, including tumor treatment, antibacterial, virus inhibition and plant photoprotection. Finally, the problems and development directions of Fe-S clusters are summarized. This review aims to guide people to accurately understand and regulate the electronic structure of Fe-S at the atomic level, which is of great significance for designing biomimetic materials with specific functions and expanding their applications in biocatalysis.
Collapse
Affiliation(s)
- Sufei Zhou
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China.
| | - Di Liu
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China.
| | - Kelong Fan
- Key Laboratory of Protein and Peptide Drugs, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Haile Liu
- Key Laboratory of Water Security and Water Environment Protection in Plateau Intersection (NWNU), Ministry of Education; Key Lab of Bioelectrochemistry and Environmental Analysis of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, China.
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China.
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| |
Collapse
|
15
|
Nasra S, Pramanik S, Oza V, Kansara K, Kumar A. Advancements in wound management: integrating nanotechnology and smart materials for enhanced therapeutic interventions. DISCOVER NANO 2024; 19:159. [PMID: 39354172 PMCID: PMC11445205 DOI: 10.1186/s11671-024-04116-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024]
Abstract
Wound management spans various techniques and materials tailored to address acute and chronic non-healing wounds, with the primary objective of achieving successful wound closure. Chronic wounds pose additional challenges, often necessitating dressings to prepare the wound bed for subsequent surgical procedures like skin grafting. Ideal dressing materials should not only expedite wound healing but also mitigate protein, electrolyte, and fluid loss while minimizing pain and infection risk. Nanotechnology has emerged as a transformative tool in wound care, revolutionizing the landscape of biomedical dressings. Its application offers remarkable efficacy in accelerating wound healing and combating bacterial infections, representing a significant advancement in wound care practices. Integration of nanotechnology into dressings has resulted in enhanced properties, including improved mechanical strength and controlled drug release, facilitating tailored therapeutic interventions. This review article comprehensively explores recent breakthroughs in wound healing therapies, with a focus on innovative medical dressings such as nano-enzymes. Additionally, the utilization of smart materials, like hydrogels and electroactive polymers, in wound dressings offers dynamic functionalities to promote tissue regeneration. Emerging concepts such as bio-fabrication, microfluidic systems, bio-responsive scaffolds, and personalized therapeutics show promise in expediting wound healing and minimizing scarring. Through an in-depth exploration of these advancements, this review aims to catalyze a paradigm shift in wound care strategies, promoting a patient-centric approach to therapeutic interventions.
Collapse
Affiliation(s)
- Simran Nasra
- Biological and Life Sciences, School of Arts a Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Sanjali Pramanik
- Biological and Life Sciences, School of Arts a Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Vidhi Oza
- Biological and Life Sciences, School of Arts a Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Krupa Kansara
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts a Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| |
Collapse
|
16
|
Chen Y, Wang N, Lv Y, Zhou C, Liang Q, Su X. Construction of the fluorescence sensing platform with a bifunctional Cu@MOF nanozyme for determination of alkaline phosphatase and its inhibitor. Talanta 2024; 278:126564. [PMID: 39018761 DOI: 10.1016/j.talanta.2024.126564] [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: 01/24/2024] [Revised: 06/08/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
In this work, a novel and sensitive fluorescence sensing system for alkaline phosphatase (ALP) was constructed using a bifunctional copper metal-organic framework (Cu@MOF) nanozyme, which had excellent oxidase-mimetic activity and fluorescence properties. Owing to the presence of 2-amino-1,4-benzenedicarboxylic acid (1,4-BDC-NH2) ligand, Cu@MOF displays excellent fluorescence performance at 444 nm. Additionally, Cu2+ endows the oxidase-like activity of Cu@MOF, which could trigger p-phenylenediamine (PPD) to be oxidized to a brown product (PPDox) and quench the photoluminescence of Cu@MOF through the inner filtration effect (IFE). As the preferential affinity of ATP for Cu2+, the catalytic activity of Cu@MOF was significantly reduced once ATP was added, thus PPD could not be oxidized and fluorescence was recovered. In the presence of ALP, ATP was hydrolyzed to adenosine and Pi, which allowed Cu@MOF to regain its catalytic activity and continued to catalyze the generation of PPDox. The fluorescence of Cu@MOF was therefore weakened once again. The ALP activity was directly proportional to the degree of decrease in fluorescence intensity. Thus, this novel fluorescence sensing strategy had a linear range of 0.5-60 U/L and the limit of detection was 0.14 U/L. The established sensing method could also be used to for ALP inhibitors screening, and achieved satisfactory results in determining the level of ALP activity in human serum.
Collapse
Affiliation(s)
- Yuhan Chen
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Nan Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Yuntai Lv
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Chenyu Zhou
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Qing Liang
- School of Life Sciences, Jilin University, Changchun, 130012, China.
| | - Xingguang Su
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun, 130012, China.
| |
Collapse
|
17
|
Welegergs GG, Ambaye AD, Jokazi M, Nwahara N, Nyokong T. Bioengineering of one dimensional hierarchical Cu 7S 4 hollow nanotubes for non-enzymatic glucose sensing applications. RSC Adv 2024; 14:27122-27131. [PMID: 39193293 PMCID: PMC11348840 DOI: 10.1039/d4ra05199h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Herein, a novel and facile eco-friendly green chemistry approach has been devised at room temperature for synthesis of 1D hierarchical Cu7S4 hollow nanotubes on Cu substrate via volatile organosulfur compounds from Allium sativum L for non-enzymatic glucose detection. Field emission scanning electron microscopy (FESEM), transmission electron microscopy (TEM), X-ray diffraction (XRD), energy dispersive X-ray spectroscopy (EDX), and X-ray spectroscopy (XPS) were employed to characterize the surface morphology, structural phase, compositional, and chemical states of the obtained samples, respectively. The SEM results confirm the formation of 1D hierarchical Cu7S4 hollow nanotubes. The XRD patterns are indexed to orthogonal anilite Cu7S4 crystal planes and the EDX spectra clearly reveal the presence of Cu and S elements. XPS spectra confirms peaks of Cu 2p and S 1s core levels, which are typical characteristics of Cu(i) and S(ii), respectively. The Brunauer-Emmett-Teller (BET) specific surface area for obtained Cu7S4 hollow nanotubes is 2.07 m2 g-1 with a pore size distribution of 27.90 nm. Using Cu7S4 hollow nanotubes, the detection of non-enzymatic glucose was conducted over a dynamic range of concentrations from 0.5 to 100 μmol L-1 and reveals a high sensitivity of 1058.33 μA mM-1cm-2 and a limit of detection (LOD) of 0.127 μmol L-1. The obtained results indicated that Cu7S4 hollow nanotubes are promising candidates for non-enzymatic glucose detection.
Collapse
Affiliation(s)
- Giday G Welegergs
- Institute for Nanotechnology Innovation, Rhodes University Makhanda 6140 South Africa
- Debre Berhan University, Department of Chemistry P. O. Box 445 Debre Berhan Ethiopia
| | - Abera D Ambaye
- Materials Science and Engineering, Bio, and Emerging Technology Institute 5954 Addis Ababa Ethiopia
| | - Mbulelo Jokazi
- Institute for Nanotechnology Innovation, Rhodes University Makhanda 6140 South Africa
| | - Nnamdi Nwahara
- Institute for Nanotechnology Innovation, Rhodes University Makhanda 6140 South Africa
| | - Tebello Nyokong
- Institute for Nanotechnology Innovation, Rhodes University Makhanda 6140 South Africa
| |
Collapse
|
18
|
Hameed S, Sharif S, Ovais M, Xiong H. Emerging trends and future challenges of advanced 2D nanomaterials for combating bacterial resistance. Bioact Mater 2024; 38:225-257. [PMID: 38745587 PMCID: PMC11090881 DOI: 10.1016/j.bioactmat.2024.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
The number of multi-drug-resistant bacteria has increased over the last few decades, which has caused a detrimental impact on public health worldwide. In resolving antibiotic resistance development among different bacterial communities, new antimicrobial agents and nanoparticle-based strategies need to be designed foreseeing the slow discovery of new functioning antibiotics. Advanced research studies have revealed the significant disinfection potential of two-dimensional nanomaterials (2D NMs) to be severed as effective antibacterial agents due to their unique physicochemical properties. This review covers the current research progress of 2D NMs-based antibacterial strategies based on an inclusive explanation of 2D NMs' impact as antibacterial agents, including a detailed introduction to each possible well-known antibacterial mechanism. The impact of the physicochemical properties of 2D NMs on their antibacterial activities has been deliberated while explaining the toxic effects of 2D NMs and discussing their biomedical significance, dysbiosis, and cellular nanotoxicity. Adding to the challenges, we also discussed the major issues regarding the current quality and availability of nanotoxicity data. However, smart advancements are required to fabricate biocompatible 2D antibacterial NMs and exploit their potential to combat bacterial resistance clinically.
Collapse
Affiliation(s)
- Saima Hameed
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, PR China
- School of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, PR China
| | - Sumaira Sharif
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Muhammad Ovais
- BGI Genomics, BGI Shenzhen, Shenzhen, 518083, Guangdong, PR China
| | - Hai Xiong
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, PR China
| |
Collapse
|
19
|
Yuan Y, Chen L, Song K, Cheng M, Fang L, Kong L, Yu L, Wang R, Fu Z, Sun M, Wang Q, Cui C, Wang H, He J, Wang X, Liu Y, Jiang B, Jiang J, Wang C, Yan X, Zhang X, Gao L. Stable peptide-assembled nanozyme mimicking dual antifungal actions. Nat Commun 2024; 15:5636. [PMID: 38965232 PMCID: PMC11224359 DOI: 10.1038/s41467-024-50094-6] [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: 04/19/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024] Open
Abstract
Natural antimicrobial peptides (AMPs) and enzymes (AMEs) are promising non-antibiotic candidates against antimicrobial resistance but suffer from low efficiency and poor stability. Here, we develop peptide nanozymes which mimic the mode of action of AMPs and AMEs through de novo design and peptide assembly. Through modelling a minimal building block of IHIHICI is proposed by combining critical amino acids in AMPs and AMEs and hydrophobic isoleucine to conduct assembly. Experimental validations reveal that IHIHICI assemble into helical β-sheet nanotubes with acetate modulation and perform phospholipase C-like and peroxidase-like activities with Ni coordination, demonstrating high thermostability and resistance to enzymatic degradation. The assembled nanotubes demonstrate cascade antifungal actions including outer mannan docking, wall disruption, lipid peroxidation and subsequent ferroptotic death, synergistically killing >90% Candida albicans within 10 min on disinfection pad. These findings demonstrate an effective de novo design strategy for developing materials with multi-antimicrobial mode of actions.
Collapse
Affiliation(s)
- Ye Yuan
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lei Chen
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kexu Song
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Miaomiao Cheng
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ling Fang
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Xishan People's Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, 214105, Jiangsu, China
| | - Lingfei Kong
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lanlan Yu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Ruonan Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Zhendong Fu
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Minmin Sun
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qian Wang
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chengjun Cui
- Xishan People's Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, 214105, Jiangsu, China
| | - Haojue Wang
- Xishan People's Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, 214105, Jiangsu, China
| | - Jiuyang He
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaonan Wang
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuan Liu
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chenxuan Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xiyun Yan
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450052, China
| | - Xinzheng Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lizeng Gao
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- CAS Engineering Laboratory for Nanozyme, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
20
|
Cao H, Yuan Y, Zhao R, Shi W, Jiang J, Gao Y, Chen L, Gao L. Deciphering the Catalytic Mechanism of Peroxidase-like Activity of Iron Sulfide Nanozymes. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30958-30966. [PMID: 38833280 DOI: 10.1021/acsami.4c06024] [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: 06/06/2024]
Abstract
Iron sulfide nanomaterials represented by FeS2 and Fe3S4 nanozymes have attracted increasing attention due to their biocompatibility and peroxidase-like (POD-like) catalytic activity in disease diagnosis and treatments. However, the mechanism responsible for their POD-like activities remains unclear. Herein, taking the oxidation of 3,3,5,5-tetramethylbenzidine (TMB) by H2O2 on FeS2(100) and Fe3S4(001) surfaces, the catalytic mechanism was investigated in detail using density functional theory (DFT) calculations and experimental characterizations. Our experimental results showed that the catalytic activity of FeS2 nanozymes was significantly higher than that of Fe3S4 nanozymes. Our DFT calculations indicated that the surface iron ions of iron sulfide nanozymes could effectively catalyze the production of HO• radicals via the interactions between Fe 3d electrons and the frontier orbitals of H2O2 in the range of -10 to 5 eV. However, FeS2 nanozymes exhibited higher POD-like activity due to the surface Fe(II) binding to H2O2, forming inner-orbital complexes, which results in a larger binding energy and a smaller energy barrier for the base-like decomposition of H2O2. In contrast, the surface iron ions of Fe3S4 nanozymes bind to H2O2, forming outer-orbital complexes, which results in a smaller binding energy and a larger energy barrier for the base-like decomposition of H2O2. The charge transfer analysis showed that FeS2 nanozymes transferred 0.12 e and Fe3S4 nanozymes transferred 0.05 e from their surface iron ions to H2O2, respectively. The simulations were consistent with the experimental observations that the FeS2 nanozymes had a greater affinity for H2O2 compared to that of Fe3S4 nanozymes. This work provides a theoretical foundation for the rational design and accurate preparation of iron sulfide functional nanozymes.
Collapse
Affiliation(s)
- Haolin Cao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ye Yuan
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Runze Zhao
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wei Shi
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yang Gao
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Lei Chen
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Translational Medicine, Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou 225001, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan 450052, China
| |
Collapse
|
21
|
Deng S, Ou K, Zhang C, Yuan D, Cai X, Li F, Wang X, Yin J, Xu C, Li Y, Gong T. A one-two punch strategy for diabetic wound management based on an antibiotic-hybrid biomineralized iron sulfide nanoparticle. Acta Biomater 2024; 181:333-346. [PMID: 38643814 DOI: 10.1016/j.actbio.2024.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 04/23/2024]
Abstract
Bacterial infection and immune imbalance are the primary culprits behind chronic wounds in individuals with diabetes, impeding the progression of damaged tissues towards normal healing. To achieve a harmonious balance between pro- and anti-inflammation within these infected areas, herein, we propose a one-two punch strategy for on-demand therapy of diabetes-infected wounds, utilizing an azithromycin (AZM)-hybrid nanocomposite termed GOx@FexSy/AZM. During the infective stage, the nanocomposite facilitates the production of ROS, coupled with the burst release of AZM and H2S gas, effectively dismantling biofilms and achieving rapid sterilization. Subsequently, the hyperinflammatory response induced by antibiosis is significantly mitigated through the synergistic action of tissue H2S and the prolonged half-life of AZM. These components inhibit the activity of pro-inflammatory transcription factors (AP-1 and NF-κB) within macrophages, thereby promoting the polarization of macrophages towards a reparative M2 phenotype and facilitating tissue remodeling. By catering to the diverse requirements of wound healing at different stages, this nanocomposite accelerates a sensible transition from inflammation to the reparative phase. In summary, this one-two punch strategy gives an instructive instance for procedural treatment of diabetes wound infection. STATEMENT OF SIGNIFICANCE: The treatment of diabetic wound infection presents two major challenges: the diminished antibacterial efficacy arising from biofilm formation and bacterial resistance, as well as the inadequate transition of the wound microenvironment from pro-inflammatory to anti-inflammatory states after bacterial clearance. In this work, a biomineralized iron sulfide nanocomposite was prepared to mediate cascade catalytic (ROS storm) / antibiotic (AZM) / gas (H2S) triple-synergetic antibacterial therapy during the initial stage of bacterial infection, achieving the goal of rapid bactericidal effect; Subsequently, the residual H2S and long half-life AZM would inhibit the key pro-inflammatory transcription factors and promote the macrophages polarization to reparative M2, which effectively mediated tissue repair after hyperinflammatory reactions, leading to orderly treatment of hyperglycemic infected wounds.
Collapse
Affiliation(s)
- Shuangpiao Deng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Kaixin Ou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Chenyu Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Department of Pharmacy, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangzhou, 510095, PR China
| | - Daojing Yuan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Xiaowen Cai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Fengtan Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Xuetao Wang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Jing Yin
- Department of Cerebrovascular Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, PR China.
| | - Chuanshan Xu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Yanli Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Teng Gong
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
22
|
Huang XL. Unveiling the role of inorganic nanoparticles in Earth's biochemical evolution through electron transfer dynamics. iScience 2024; 27:109555. [PMID: 38638571 PMCID: PMC11024932 DOI: 10.1016/j.isci.2024.109555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
This article explores the intricate interplay between inorganic nanoparticles and Earth's biochemical history, with a focus on their electron transfer properties. It reveals how iron oxide and sulfide nanoparticles, as examples of inorganic nanoparticles, exhibit oxidoreductase activity similar to proteins. Termed "life fossil oxidoreductases," these inorganic enzymes influence redox reactions, detoxification processes, and nutrient cycling in early Earth environments. By emphasizing the structural configuration of nanoparticles and their electron conformation, including oxygen defects and metal vacancies, especially electron hopping, the article provides a foundation for understanding inorganic enzyme mechanisms. This approach, rooted in physics, underscores that life's origin and evolution are governed by electron transfer principles within the framework of chemical equilibrium. Today, these nanoparticles serve as vital biocatalysts in natural ecosystems, participating in critical reactions for ecosystem health. The research highlights their enduring impact on Earth's history, shaping ecosystems and interacting with protein metal centers through shared electron transfer dynamics, offering insights into early life processes and adaptations.
Collapse
Affiliation(s)
- Xiao-Lan Huang
- Center for Clean Water Technology, School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, NY 11794-6044, USA
| |
Collapse
|
23
|
Cui F, Li L, Wang D, Li J, Li T. Nanomaterials with Enzyme-like Properties for Combatting Foodborne Pathogen Infections: Classifications, Mechanisms, and Applications in Food Preservation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10179-10194. [PMID: 38685503 DOI: 10.1021/acs.jafc.4c00872] [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: 05/02/2024]
Abstract
During the transportation and storage of food, foodborne spoilage caused by bacterial and biofilm infection is prone to occur, leading to issues such as short shelf life, economic loss, and sensory quality instability. Therefore, the development of novel and efficient antibacterial agents capable of efficiently inhibiting bacteria throughout various stages of food processing, transportation, and storage is strongly recommended by researchers. The emergence of nanozymes is considered to be an effective candidate for inhibiting foodborne bacteria agents in the food industry. As potent antibacterial agents, nanozymes have the advantages of low cost, high stability, strong broad-spectrum antibacterial ability, and biocompatibility. Herein, we aim to summarize the classification status of various nanozymes. Furthermore, the general catalytic bacteriostatic mechanism of nanozymes against intracellular bacteria, planktonic bacteria, and biofilm activities are highlighted, mainly concerning the destruction of cell walls and/or membranes, reactive oxygen species regulation, HOBr/Cl generation, damage of intracellular components, and so forth. In particular, the review focuses on the pivotal role of nanozymes as antibacterial agents and delivery vehicles in the fields of food preservation applications. We look forward to the future prospects, especially in the field of food preservation, to promote broader applications based on antimicrobial nanozymes.
Collapse
Affiliation(s)
- Fangchao Cui
- National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, College of Food Science and Technology, Bohai University, Jinzhou, Liaoning 121013, China
| | - Lanling Li
- National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, College of Food Science and Technology, Bohai University, Jinzhou, Liaoning 121013, China
| | - Dangfeng Wang
- National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, College of Food Science and Technology, Bohai University, Jinzhou, Liaoning 121013, China
| | - Jianrong Li
- National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, College of Food Science and Technology, Bohai University, Jinzhou, Liaoning 121013, China
| | - Tingting Li
- Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, Dalian, Liaoning 116029, China
| |
Collapse
|
24
|
Shan J, Jin X, Zhang C, Huang M, Xing J, Li Q, Cui Y, Niu Q, Chen XL, Wang X. Metal natural product complex Ru-procyanidins with quadruple enzymatic activity combat infections from drug-resistant bacteria. Acta Pharm Sin B 2024; 14:2298-2316. [PMID: 38799629 PMCID: PMC11121202 DOI: 10.1016/j.apsb.2023.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 05/29/2024] Open
Abstract
Bacterial infection hampers wound repair by impeding the healing process. Concurrently, inflammation at the wound site triggers the production of reactive oxygen species (ROS), causing oxidative stress and damage to proteins and cells. This can lead to chronic wounds, posing severe risks. Therefore, eliminating bacterial infection and reducing ROS levels are crucial for effective wound healing. Nanozymes, possessing enzyme-like catalytic activity, can convert endogenous substances into highly toxic substances, such as ROS, to combat bacteria and biofilms without inducing drug resistance. However, the current nanozyme model with single enzyme activity falls short of meeting the complex requirements of antimicrobial therapy. Thus, developing nanozymes with multiple enzymatic activities is essential. Herein, we engineered a novel metalloenzyme called Ru-procyanidin nanoparticles (Ru-PC NPs) with diverse enzymatic activities to aid wound healing and combat bacterial infections. Under acidic conditions, due to their glutathione (GSH) depletion and peroxidase (POD)-like activity, Ru-PC NPs combined with H2O2 exhibit excellent antibacterial effects. However, in a neutral environment, the Ru-PC NPs, with catalase (CAT) activity, decompose H2O2 to O2, alleviating hypoxia and ensuring a sufficient oxygen supply. Furthermore, Ru-PC NPs possess exceptional antioxidant capacity through their superior superoxide dismutase (SOD) enzyme activity, effectively scavenging excess ROS and reactive nitrogen species (RNS) in a neutral environment. This maintains the balance of the antioxidant system and prevents inflammation. Ru-PC NPs also promote the polarization of macrophages from M1 to M2, facilitating wound healing. More importantly, Ru-PC NPs show good biosafety with negligible toxicity. In vivo wound infection models have confirmed the efficacy of Ru-PC NPs in inhibiting bacterial infection and promoting wound healing. The focus of this work highlights the quadruple enzymatic activity of Ru-PC NPs and its potential to reduce inflammation and promote bacteria-infected wound healing.
Collapse
Affiliation(s)
- Jie Shan
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Xu Jin
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Cong Zhang
- Division of Gastroenterology, the First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Muchen Huang
- The Second Clinical Medical College, Anhui Medical University, Hefei 230022, China
| | - Jianghao Xing
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qingrong Li
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Yuyu Cui
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qiang Niu
- The Second Clinical Medical College, Anhui Medical University, Hefei 230022, China
| | - Xu Lin Chen
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| |
Collapse
|
25
|
Zuo J, Quan Y, Li J, Li Y, Song D, Li X, Wang Y, Yi L, Wang Y. Tackling Antibiotic Resistance: Exploring 5-Fluorouracil as a Promising Antimicrobial Strategy for the Treatment of Streptococcus suis Infection. Animals (Basel) 2024; 14:1286. [PMID: 38731290 PMCID: PMC11083182 DOI: 10.3390/ani14091286] [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: 03/15/2024] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Streptococcus suis (S. suis) is a zoonotic pathogen with a global distribution, which causes serious diseases in both humans and animals and economic losses in the swine industry. As antibiotic resistance increases, there is an urgent imperative to explore novel antibacterial alternatives. In the present study, we selected the anticancer drug 5-fluorouracil (5-FU) approved by the Food and Drug Administration (FDA) as a candidate drug to treat S. suis infections. The results showed that various pathogens, especially S. suis, are more sensitive to 5-FU. Moreover, the cytotoxicity of 5-FU is relatively low. Extensive in vitro assays demonstrated the pronounced bacteriostatic and bactericidal efficacy of 5-FU against susceptible and multidrug-resistant S. suis strains. Its mechanisms of action include damage to the bacterial cell walls and membranes, resulting in the leakage of intracellular components, and the inhibition of thymidylate synthase (TS), leading to a depletion of deoxythymidine triphosphate (dTTP) pools, ultimately causing thymine-less death and lethal DNA damage in bacteria. Gene-knockout experiments further showed that 5-FU played a role by inhibiting the thyA gene-encoding thymidine synthase. Finally, we determined that S. suis infections can be alleviated by 5-FU in the mouse infection model. This study emphasizes the antibacterial potential of 5-FU against S. suis and provides evidence for its targeting of bacterial membrane damage and DNA damage. In summary, 5-FU can control S. suis infection and is expected to become a new alternative to antibiotics.
Collapse
Affiliation(s)
- Jing Zuo
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (J.Z.); (Y.Q.); (Y.L.); (D.S.); (X.L.); (Y.W.)
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China;
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (J.Z.); (Y.Q.); (Y.L.); (D.S.); (X.L.); (Y.W.)
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China;
| | - Jinpeng Li
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China;
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Yue Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (J.Z.); (Y.Q.); (Y.L.); (D.S.); (X.L.); (Y.W.)
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China;
| | - Dong Song
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (J.Z.); (Y.Q.); (Y.L.); (D.S.); (X.L.); (Y.W.)
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China;
| | - Xingping Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (J.Z.); (Y.Q.); (Y.L.); (D.S.); (X.L.); (Y.W.)
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China;
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (J.Z.); (Y.Q.); (Y.L.); (D.S.); (X.L.); (Y.W.)
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China;
| | - Li Yi
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China;
- College of Life Science, Luoyang Normal University, Luoyang 471934, China
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (J.Z.); (Y.Q.); (Y.L.); (D.S.); (X.L.); (Y.W.)
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China;
| |
Collapse
|
26
|
Han J, Chen Y, Xiang X, Wang T, Shen J, Zhang N, Liang C, Liu X, Ma X. A Comparative Analysis of the Antibacterial Spectrum of Ultrasmall Manganese Ferrite Nanozymes with Varied Surface Modifications. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38489475 DOI: 10.1021/acsami.3c16490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Bacterial infectious diseases pose a significant global challenge. However, conventional antibacterial agents exhibit limited therapeutic effectiveness due to the emergence of drug resistance, necessitating the exploration of novel antibacterial strategies. Nanozymes have emerged as a highly promising alternative to antibiotics, owing to their particular catalytic activities against pathogens. Herein, we synthesized ultrasmall-sized MnFe2O4 nanozymes with different charges (MnFe2O4-COOH, MnFe2O4-PEG, MnFe2O4-NH2) and assessed their antibacterial capabilities. It was found that MnFe2O4 nanozymes exhibited both antibacterial and antibiofilm properties attributed to their excellent peroxidase-like activities and small sizes, enabling them to penetrate biofilms and interact with bacteria. Moreover, MnFe2O4 nanozymes effectively expedite wound healing within 12 days and facilitate tissue repair and regeneration while concurrently reducing inflammation. MnFe2O4-COOH displayed favorable antibacterial activity against Gram-positive bacteria, with 80% bacterial removal efficiency against MRSA by interacting with phosphatidylglycerol (PG) and cardiolipin (CL) of the membrane. By interacting with negatively charged bacteria surfaces, MnFe2O4-NH2 demonstrated the most significant and broad-spectrum antibacterial activity, with 95 and 85% removal efficiency against methicillin-resistant Staphylococcus aureus (MRSA) and P. aeruginosa, respectively. MnFe2O4-PEG dissipated membrane potential and reduced ATP levels in MRSA and P. aeruginosa, showing relatively broad-spectrum antibacterial activity. To conclude, MnFe2O4 nanozymes offer a promising therapeutic approach for treating wound infections.
Collapse
Affiliation(s)
- Junhua Han
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Yingxian Chen
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Xin Xiang
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Tingting Wang
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Nan Zhang
- Center for Regenerative and Reconstructive Medicine, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
| | - Chen Liang
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, P. R. China
| | - Xiaoli Liu
- Center for Regenerative and Reconstructive Medicine, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, P. R. China
| | - Xiaowei Ma
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
- Sanya Institute of China Agricultural University, Sanya, Hainan 572025, P. R. China
| |
Collapse
|
27
|
Chen C, Chen L, Mao C, Jin L, Wu S, Zheng Y, Cui Z, Li Z, Zhang Y, Zhu S, Jiang H, Liu X. Natural Extracts for Antibacterial Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306553. [PMID: 37847896 DOI: 10.1002/smll.202306553] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/23/2023] [Indexed: 10/19/2023]
Abstract
Bacteria-induced epidemics and infectious diseases are seriously threatening the health of people around the world. In addition, antibiotic therapy has been inducing increasingly more serious bacterial resistance, which makes it urgent to develop new treatment strategies to combat bacteria, including multidrug-resistant bacteria. Natural extracts displaying antibacterial activity and good biocompatibility have attracted much attention due to greater concerns about the safety of synthetic chemicals and emerging drug resistance. These antibacterial components can be isolated and utilized as antimicrobials, as well as transformed, combined, or wrapped with other substances by using modern assistive technologies to fight bacteria synergistically. This review summarizes recent advances in natural extracts from three kinds of sources-plants, animals, and microorganisms-for antibacterial applications. This work discusses the corresponding antibacterial mechanisms and the future development of natural extracts in antibacterial fields.
Collapse
Affiliation(s)
- Cuihong Chen
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Lin Chen
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Congyang Mao
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
| | - Liguo Jin
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Shuilin Wu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Zhaoyang Li
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Shengli Zhu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Hui Jiang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Xiangmei Liu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
| |
Collapse
|
28
|
Zhang L, Wang H, Qu X. Biosystem-Inspired Engineering of Nanozymes for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2211147. [PMID: 36622946 DOI: 10.1002/adma.202211147] [Citation(s) in RCA: 97] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Indexed: 06/17/2023]
Abstract
Nanozymes with intrinsic enzyme-mimicking activities have shown great potential to become surrogates of natural enzymes in many fields by virtue of their advantages of high catalytic stability, ease of functionalization, and low cost. However, due to the lack of predictable descriptors, most of the nanozymes reported in the past have been obtained mainly through trial-and-error strategies, and the catalytic efficacy, substrate specificity, as well as practical application effect under physiological conditions, are far inferior to that of natural enzymes. To optimize the catalytic efficacies and functions of nanozymes in biomedical settings, recent studies have introduced biosystem-inspired strategies into nanozyme design. In this review, recent advances in the engineering of biosystem-inspired nanozymes by leveraging the refined catalytic structure of natural enzymes, simulating the behavior changes of natural enzymes in the catalytic process, and mimicking the specific biological processes or living organisms, are introduced. Furthermore, the currently involved biomedical applications of biosystem-inspired nanozymes are summarized. More importantly, the current opportunities and challenges of the design and application of biosystem-inspired nanozymes are discussed. It is hoped that the studies of nanozymes based on bioinspired strategies will be beneficial for constructing the new generation of nanozymes and broadening their biomedical applications.
Collapse
Affiliation(s)
- Lu Zhang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Huan Wang
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Xiaogang Qu
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| |
Collapse
|
29
|
Bao W, Tian L, Wang H, Tang A, Yang H. Breaking through the pH Limitation of Fe 1-xS Nanozymes Using Component-Modulated Coupled Nanoclay. Inorg Chem 2024; 63:3366-3375. [PMID: 38323570 DOI: 10.1021/acs.inorgchem.3c03934] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Overcoming the intrinsic low activity of most peroxidase mimics under neutral pH is crucial but still extremely challenging for the detection of disease markers in biological samples. Here, we chose nanoclay (i.e., montmorillonite K10, MK10) as a carrier to modulate the structure of Fe1-xS nanozyme components through an interfacial modulation strategy, aiming at breaking the neutral pH limitation of Fe1-xS. MK10 with abundant hydroxyl groups on its surface acts as a carrier to increase the ratio of Fe(II) and S(II-) content in surface Fe1-xS. We verify that Fe(II)-promoted surface hydroxyl radical generation and S(II-)-promoted regeneration of Fe(II) play key roles in endowing peroxidase-like activity to Fe1-xS at neutral pH. As expected, Fe1-xS/MK10 exhibited 11-fold higher Vmax and 52-fold higher catalytic efficiency than bare Fe1-xS. As a proof of concept, the sensor constructed based on Fe1-xS/MK10 achieved colorimetric detection of xanthine under neutral conditions with a linear range of 5-300 μM and a limit of detection of 2.49 μM. Finally, we achieved highly sensitive detection of xanthine in serum using the constructed biosensor. Our contribution is the novel use of a nanoclay-mediated interfacial modulation strategy for boosting the peroxidase-mimicking activity and breaking the pH limitation, which contributes to the in situ detection of disease markers by nanozymes under physiological conditions.
Collapse
Affiliation(s)
- Wenxin Bao
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan 430074, China
| | - Luyuan Tian
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan 430074, China
| | - Hao Wang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan 430074, China
| | - Aidong Tang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan 430074, China
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Huaming Yang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan 430074, China
- Hunan Key Laboratory of Mineral Materials and Application, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China
| |
Collapse
|
30
|
Huang XL, Harmer JR, Schenk G, Southam G. Inorganic Fe-O and Fe-S oxidoreductases: paradigms for prebiotic chemistry and the evolution of enzymatic activity in biology. Front Chem 2024; 12:1349020. [PMID: 38389729 PMCID: PMC10881703 DOI: 10.3389/fchem.2024.1349020] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Oxidoreductases play crucial roles in electron transfer during biological redox reactions. These reactions are not exclusive to protein-based biocatalysts; nano-size (<100 nm), fine-grained inorganic colloids, such as iron oxides and sulfides, also participate. These nanocolloids exhibit intrinsic redox activity and possess direct electron transfer capacities comparable to their biological counterparts. The unique metal ion architecture of these nanocolloids, including electron configurations, coordination environment, electron conductivity, and the ability to promote spontaneous electron hopping, contributes to their transfer capabilities. Nano-size inorganic colloids are believed to be among the earliest 'oxidoreductases' to have 'evolved' on early Earth, playing critical roles in biological systems. Representing a distinct type of biocatalysts alongside metalloproteins, these nanoparticles offer an early alternative to protein-based oxidoreductase activity. While the roles of inorganic nano-sized catalysts in current Earth ecosystems are intuitively significant, they remain poorly understood and underestimated. Their contribution to chemical reactions and biogeochemical cycles likely helped shape and maintain the balance of our planet's ecosystems. However, their potential applications in biomedical, agricultural, and environmental protection sectors have not been fully explored or exploited. This review examines the structure, properties, and mechanisms of such catalysts from a material's evolutionary standpoint, aiming to raise awareness of their potential to provide innovative solutions to some of Earth's sustainability challenges.
Collapse
Affiliation(s)
- Xiao-Lan Huang
- NYS Center for Clean Water Technology, School of Marine and Atmospheric Sciences, Stony Brook, NY, United States
| | - Jeffrey R Harmer
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Gerhard Schenk
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Sustainable Minerals Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Gordon Southam
- Sustainable Minerals Institute, The University of Queensland, Brisbane, QLD, Australia
- School of the Environment, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
31
|
Mu Q, Deng H, An X, Liu G, Liu C. Designing nanodiscs as versatile platforms for on-demand therapy. NANOSCALE 2024; 16:2220-2234. [PMID: 38192208 DOI: 10.1039/d3nr05457h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Nowadays, there has been an increasing utilization of nanomedicines for disease treatment. Nanodiscs (NDs) have emerged as a novel platform technology that garners significant attention in biomedical research and drug discovery. NDs are nanoscale phospholipid bilayer discs capable of incorporating membrane proteins and lipids within a native-like environment. They are assembled using amphiphilic biomacromolecular materials, such as apolipoprotein A1 or membrane scaffold proteins (MSPs), peptides, and styrene-maleic acid polymers (SMAs). NDs possess well-defined sizes and shapes, offering a stable, homogeneous, and biologically relevant environment for studying membrane proteins and lipids. Their unique properties have made them highly desirable for diverse applications, including cancer immunotherapy, vaccine development, antibacterial and antiviral therapy, and treating Alzheimer's disease (AD) and diabetes-related conditions. This review discusses the classifications, advantages, and applications of NDs in disease therapy.
Collapse
Affiliation(s)
- Qianwen Mu
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Haolan Deng
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaoyu An
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chao Liu
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| |
Collapse
|
32
|
Maroto-Tello A, Ayllón T, Aguinaga-Casañas MA, Ariza JJ, Penelo S, Baños A, Ortiz-Díez G. In Vitro Activity of Allium cepa Organosulfur Derivatives against Canine Multidrug-Resistant Strains of Staphylococcus spp. and Enterobacteriaceae. Vet Sci 2024; 11:26. [PMID: 38250932 PMCID: PMC10820550 DOI: 10.3390/vetsci11010026] [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: 10/27/2023] [Revised: 12/06/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND The increase of multi-resistant bacteria, especially Staphylococcus spp. and Enterobacteriaceae, constitutes a challenge in veterinary medicine. The rapid growth of resistance is outpacing antibiotic discovery. Innovative strategies are needed, including the use of natural products like Allium species (Allium sativum L. and Allium cepa L.), which have been used empirically for centuries to treat infectious diseases in humans and farm and aquaculture animals due to their antibacterial properties. METHODS This study aimed to evaluate the in vitro activity of two Allium-derived compounds, propyl propane thiosulfinate (PTS) and propyl propane thiosulfonate (PTSO), against multi-resistant Staphylococcus spp. (n = 30) and Enterobacteriaceae (n = 26) isolated from dogs referred to a veterinary teaching hospital in Madrid. RESULTS AND DISCUSSION The results indicated the in vitro efficacy of PTSO/PTS against the tested bacterial strains, and 56.7% of Staphylococcus pseudintermedius and 53.8% of Enterobacteriaceae showed sensitivity to PTS and PTSO compared with classic antibiotics. In addition, 50% of S. pseudintermedius strains resistant to erythromycin, ibofloxacin, difloxacin and orbifloxacin and 50% of Enterobacteriaceae strains resistant to tetracycline and doxycycline were sensitive to PTS and PTSO. Although studies are needed to verify their efficacy in vivo, the combined use of PTS and PTSO exhibits promise in enhancing bacterial sensitivity against S. pseudintermedius and Enterobacteriaceae infections, providing a first insight into the potential of both compounds in veterinary practice.
Collapse
Affiliation(s)
- Alba Maroto-Tello
- Departamento de Microbiología, DMC Research Center, 18620 Granada, Spain; (A.M.-T.); (M.A.A.-C.); (A.B.)
| | - Tania Ayllón
- Facultad de Ciencias de la Salud, Universidad Alfonso X el Sabio, 28691 Madrid, Spain
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense, 28040 Madrid, Spain
| | | | - Juan José Ariza
- Departamento de Microbiología, Campus Fuente Nueva, Universidad de Granada, 18001 Granada, Spain;
| | - Silvia Penelo
- Servicio de Urgencias, Hospitalización y UCI, Hospital Clínico Veterinario Complutense, Universidad Complutense, 28040 Madrid, Spain
| | - Alberto Baños
- Departamento de Microbiología, DMC Research Center, 18620 Granada, Spain; (A.M.-T.); (M.A.A.-C.); (A.B.)
- Departamento de Microbiología, Campus Fuente Nueva, Universidad de Granada, 18001 Granada, Spain;
| | - Gustavo Ortiz-Díez
- Departamento de Medicina y Cirugía, Facultad de Veterinaria, Universidad Complutense, 28040 Madrid, Spain;
| |
Collapse
|
33
|
Xing Z, Guo J, Wu Z, He C, Wang L, Bai M, Liu X, Zhu B, Guan Q, Cheng C. Nanomaterials-Enabled Physicochemical Antibacterial Therapeutics: Toward the Antibiotic-Free Disinfections. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303594. [PMID: 37626465 DOI: 10.1002/smll.202303594] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/17/2023] [Indexed: 08/27/2023]
Abstract
Bacterial infection continues to be an increasing global health problem with the most widely accepted treatment paradigms restricted to antibiotics. However, the overuse and misuse of antibiotics have triggered multidrug resistance of bacteria, frustrating therapeutic outcomes, and leading to higher mortality rates. Even worse, the tendency of bacteria to form biofilms on living and nonliving surfaces further increases the difficulty in confronting bacteria because the extracellular matrix can act as a robust barrier to prevent the penetration of antibiotics and resist environmental damage. As a result, the inability to eliminate bacteria and biofilms often leads to persistent infection, implant failure, and device damage. Therefore, it is of paramount importance to develop alternative antimicrobial agents while avoiding the generation of bacterial resistance to prevent the large-scale growth of bacterial resistance. In recent years, nano-antibacterial materials have played a vital role in the antibacterial field because of their excellent physical and chemical properties. This review focuses on new physicochemical antibacterial strategies and versatile antibacterial nanomaterials, especially the mechanism and types of 2D antibacterial nanomaterials. In addition, this advanced review provides guidance on the development direction of antibiotic-free disinfections in the antibacterial field in the future.
Collapse
Affiliation(s)
- Zhenyu Xing
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Jiusi Guo
- Department of Orthodontics, Department of Endodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zihe Wu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Chao He
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Liyun Wang
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingru Bai
- Department of Orthodontics, Department of Endodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xikui Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Bihui Zhu
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuyue Guan
- Department of Geriatrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
34
|
Sun C, Zhou X, Liu C, Deng S, Song Y, Yang J, Dai J, Ju Y. An Integrated Therapeutic and Preventive Nanozyme-Based Microneedle for Biofilm-Infected Diabetic Wound Healing. Adv Healthc Mater 2023; 12:e2301474. [PMID: 37479531 DOI: 10.1002/adhm.202301474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/28/2023] [Indexed: 07/23/2023]
Abstract
The healing of biofilm-infected diabetic wounds characterized by a deteriorative tissue microenvironment represents a substantial clinical challenge. Current treatments remain unsatisfactory due to the limited antibiofilm efficacy caused by weak tissue and biofilm permeability of drugs and the risk of reinfection during the healing process. To address these issues, an integrated therapeutic and preventive nanozyme-based microneedle (denoted as Fe2 C/GOx@MNs) is engineered. The dissolvable tips with enough mechanical strength can deliver and rapidly release Fe2 C nanoparticles (NPs)/glucose oxidase (GOx) in the biofilm active regions, enhancing tissue and biofilm permeability of Fe2 C NPs/GOx, ultimately achieving highly efficient biofilm elimination. Meanwhile, the chitosan backing layer can not only act as an excellent physical barrier between the wound bed and the external environment, but also prevent the bacterial reinvasion during wound healing with its superior antibacterial property. Significantly, the biofilm elimination and reinfection prevention abilities of Fe2 C/GOx@MNs on wound healing are proved on methicillin-resistant Staphylococcus aureus-biofilm-infected diabetic mouse model with full-thickness wound. Together, these results demonstrate the promising clinical application of Fe2 C/GOx@MNs in biofilm-infected wound healing.
Collapse
Affiliation(s)
- Caixia Sun
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xinyu Zhou
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Cong Liu
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Shuyue Deng
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuhan Song
- Nanjing Institute for Food and Drug Control, Nanjing, 210038, China
| | - Jun Yang
- Nanjing Institute for Food and Drug Control, Nanjing, 210038, China
| | - Jianjun Dai
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yanmin Ju
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
35
|
Shen B, Yang L, Xu H, Zhang Y, Ming D, Zhu L, Wang Y, Jiang L. Detection and treatment of biofilm-induced periodontitis by histidine-doped FeSN nanozyme with ultra-high peroxidase-like activity. J Colloid Interface Sci 2023; 650:211-221. [PMID: 37402327 DOI: 10.1016/j.jcis.2023.06.188] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023]
Abstract
Pathogenic biofilm induced oral diseases have posed a significant treat to human health, such as periodontitis resulting from the formation of bacterial biofilm on teeth and gums. The traditional treatment methods such as mechanical debridement and antibiotic therapy encounter the poor therapeutic effect. Recently, numerous nanozymes with excellent antibacterial effect have been widely used in the treatment of oral diseases. In this study, a novel iron-based nanozyme (FeSN) generated by histidine-doped FeS2 with high peroxidase-like (POD-like) activity was designed for the oral biofilm removal and treatment of periodontitis. FeSN exhibited an extremely high POD-like activity, and enzymatic reaction kinetics and theoretical calculations had demonstrated its catalytic efficiency to be approximately 30 times than that of FeS2. The antibacterial experiments showed that FeSN had robust antibacterial activity against Fusobacterium nucleatum in the presence of H2O2, causing a reduction in the levels of glutathione reductase and ATP in bacterial cells, while increasing the level of oxidase coenzyme. The ultrahigh POD-like activity of FeSN allowed for easy detection of pathogenic biofilms and promoted the breakdown of biofilm structure. Furthermore, FeSN demonstrated excellent biocompatibility and low cytotoxicity to human fibroblast cells. In a rat model of periodontitis, FeSN exhibited significant therapeutic effects by reducing the extent of biofilm formation, inflammation, and alveolar bone loss. Taken together, our results suggested that FeSN, generated by self-assembly of two amino acids, represented a promising approach for biofilm removal and periodontitis treatment. This method has the potential to overcome the limitations of current treatments and provide an effective alternative for periodontitis treatment.
Collapse
Affiliation(s)
- Bowen Shen
- College of Biotechnology and Pharmaceutical Engineering, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Lei Yang
- College of Biotechnology and Pharmaceutical Engineering, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Hengyue Xu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Yangheng Zhang
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Dengming Ming
- College of Biotechnology and Pharmaceutical Engineering, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Liying Zhu
- School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Yuxian Wang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China.
| | - Ling Jiang
- College of Biotechnology and Pharmaceutical Engineering, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China; College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
36
|
Cao S, Long Y, Xiao S, Deng Y, Ma L, Adeli M, Qiu L, Cheng C, Zhao C. Reactive oxygen nanobiocatalysts: activity-mechanism disclosures, catalytic center evolutions, and changing states. Chem Soc Rev 2023; 52:6838-6881. [PMID: 37705437 DOI: 10.1039/d3cs00087g] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Benefiting from low costs, structural diversities, tunable catalytic activities, feasible modifications, and high stability compared to the natural enzymes, reactive oxygen nanobiocatalysts (RONBCs) have become dominant materials in catalyzing and mediating reactive oxygen species (ROS) for diverse biomedical and biological applications. Decoding the catalytic mechanism and structure-reactivity relationship of RONBCs is critical to guide their future developments. Here, this timely review comprehensively summarizes the recent breakthroughs and future trends in creating and decoding RONBCs. First, the fundamental classification, activity, detection method, and reaction mechanism for biocatalytic ROS generation and elimination have been systematically disclosed. Then, the merits, modulation strategies, structure evolutions, and state-of-art characterisation techniques for designing RONBCs have been briefly outlined. Thereafter, we thoroughly discuss different RONBCs based on the reported major material species, including metal compounds, carbon nanostructures, and organic networks. In particular, we offer particular insights into the coordination microenvironments, bond interactions, reaction pathways, and performance comparisons to disclose the structure-reactivity relationships and mechanisms. In the end, the future challenge and perspectives for RONBCs are also carefully summarised. We envision that this review will provide a comprehensive understanding and guidance for designing ROS-catalytic materials and stimulate the wide utilisation of RONBCs in diverse biomedical and biological applications.
Collapse
Affiliation(s)
- Sujiao Cao
- Department of Medical Ultrasound, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Yanping Long
- Department of Medical Ultrasound, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
- Department of Chemistry and Biochemistry, Freie Universitat Berlin, Takustrasse 3, Berlin 14195, Germany
| | - Sutong Xiao
- Department of Medical Ultrasound, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
| | - Yuting Deng
- Department of Medical Ultrasound, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
| | - Lang Ma
- Department of Medical Ultrasound, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
| | - Mohsen Adeli
- Department of Chemistry and Biochemistry, Freie Universitat Berlin, Takustrasse 3, Berlin 14195, Germany
| | - Li Qiu
- Department of Medical Ultrasound, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
- Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| | - Chong Cheng
- Department of Medical Ultrasound, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
- Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| | - Changsheng Zhao
- Department of Medical Ultrasound, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
- Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| |
Collapse
|
37
|
Hou J, Xianyu Y. Tailoring the Surface and Composition of Nanozymes for Enhanced Bacterial Binding and Antibacterial Activity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302640. [PMID: 37322391 DOI: 10.1002/smll.202302640] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/17/2023] [Indexed: 06/17/2023]
Abstract
With the advantages of diverse structures, tunable enzymatic activity, and high stability, nanozymes are widely used in medicine, chemistry, food, environment, and other fields. As an alternative to traditional antibiotics, nanozymes attract more and more attention from the scientific researchers in recent years. Developing nanozymes-based antibacterial materials opens up a new avenue for the bacterial disinfection and sterilization. In this review, the classification of nanozymes and their antibacterial mechanisms are discussed. The surface and composition of nanozymes are critical for the antibacterial efficacy, which can be tailored to enhance both the bacterial binding and the antibacterial activity. On the one hand, the surface modification of nanozymes enables binding and targeting of bacteria that improves the antibacterial performance of nanozymes including the biochemical recognition, the surface charge, and the surface topography. On the other hand, the composition of nanozymes can be modulated to achieve enhanced antibacterial performance including the single nanozyme-mediated synergistic and multiple nanozymes-mediated cascade catalytic antibacterial applications. In addition, the current challenges and future prospects of tailoring nanozymes for antibacterial applications are discussed. This review can provide insights into the design of future nanozymes-based materials for the antibacterial treatments.
Collapse
Affiliation(s)
- Jinjie Hou
- State Key Laboratory of Fluid Power and Mechatronic Systems, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Yunlei Xianyu
- State Key Laboratory of Fluid Power and Mechatronic Systems, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, P. R. China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital, Hangzhou, 310016, P. R. China
- Ningbo Research Institute, Zhejiang University, Ningbo, 315100, P. R. China
| |
Collapse
|
38
|
Shi Y, Li H, Chu D, Lin W, Wang X, Wu Y, Li K, Wang H, Li D, Xu Z, Gao L, Li B, Chen H. Rescuing Nucleus Pulposus Cells From Senescence via Dual-Functional Greigite Nanozyme to Alleviate Intervertebral Disc Degeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300988. [PMID: 37400370 PMCID: PMC10477883 DOI: 10.1002/advs.202300988] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/23/2023] [Indexed: 07/05/2023]
Abstract
High levels of reactive oxygen species (ROS) lead to progressive deterioration of mitochondrial function, resulting in tissue degeneration. In this study, ROS accumulation induced nucleus pulposus cells (NPCs) senescence is observed in degenerative human and rat intervertebral disc, suggesting senescence as a new therapeutic target to reverse intervertebral disc degeneration (IVDD). By targeting this, dual-functional greigite nanozyme is successfully constructed, which shows the ability to release abundant polysulfides and presents strong superoxide dismutase and catalase activities, both of which function to scavenge ROS and maintain the tissue at physical redox level. By significantly lowering the ROS level, greigite nanozyme rescues damaged mitochondrial function in IVDD models both in vitro and in vivo, rescues NPCs from senescence and alleviated the inflammatory response. Furthermore, RNA-sequencing reveals ROS-p53-p21 axis is responsible for cellular senescence-induced IVDD. Activation of the axis abolishes greigite nanozyme rescued NPCs senescence phenotype, as well as the alleviated inflammatory response to greigite nanozyme, which confirms the role of ROS-p53-p21 axis in greigite nanozyme's function to reverse IVDD. In conclusion, this study demonstrates that ROS-induced NPCs senescence leads to IVDD and the dual-functional greigite nanozyme holds strong potential to reverse this process, providing a novel strategy for IVDD management.
Collapse
Affiliation(s)
- Yu Shi
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Hanwen Li
- Orthopedic InstituteDepartment of Orthopedic SurgeryFirst Affiliated HospitalSuzhou Medical CollegeSoochow UniversityNo. 899 Pinghai RoadSuzhou215000P. R. China
| | - Dongchuan Chu
- Department of RadiologyAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
| | - Wenzheng Lin
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Xinglong Wang
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Yin Wu
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Ke Li
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Huihui Wang
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Dandan Li
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Zhuobin Xu
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Lizeng Gao
- CAS Engineering Laboratory for NanozymeInstitute of BiophysicsChinese Academy of SciencesNo. 15 Datun RoadBeijing100101P. R. China
| | - Bin Li
- Orthopedic InstituteDepartment of Orthopedic SurgeryFirst Affiliated HospitalSuzhou Medical CollegeSoochow UniversityNo. 899 Pinghai RoadSuzhou215000P. R. China
| | - Hao Chen
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
| |
Collapse
|
39
|
Ding H, Ao C, Zhang X. Potential use of garlic products in ruminant feeding: A review. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 14:343-355. [PMID: 37635929 PMCID: PMC10448032 DOI: 10.1016/j.aninu.2023.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 01/23/2023] [Accepted: 04/06/2023] [Indexed: 08/29/2023]
Abstract
The addition of antibiotics as growth promoters to ruminant feed can result in bacterial resistance and antibiotic residues in ruminant products. Correspondingly, there is serious public concern regarding the presence of antibiotic residue in ruminant products and the consequent threat to human health. As a result, the addition of plants and their products to ruminant feeds, as an alternative to antibiotics, has received much attention recently. Garlic and its products are rich in organosulphur compounds, which have a variety of biological activities and have been widely used as natural additives in animal production. This review presents recent knowledge on the addition of garlic products (powder, skin, oil, leaf and extracts) to the diets of ruminants. In this paper, garlic products are evaluated with respect to their chemical composition, bioactive compounds, and their impacts on the rumen ecosystem, antioxidant status, immune response, parasitic infection, growth performance and product quality of ruminants. This review provides valuable guidance and a theoretical basis for the development of garlic products as green, highly efficient and safe additives, with the aims of promoting ruminant growth and health, reducing methane emissions and improving ruminant product quality. Garlic extracts have the potential to control parasite infections by decreasing the faecal egg count. Garlic powder, oil and allicin are able to reduce the methane emissions of ruminants. Organosulphur compounds such as allicin, which is present in garlic products, have the potential to inhibit membrane lipid synthesis of the archaeal community, thus influencing the population of methanogenic archaea and resulting in a reduction in methane emissions. Some garlic products are also able to increase the average daily gain (garlic skin, water extract, and leaf) and the feed conversion ratio (garlic skin and leaf) of ruminants. Garlic stalk silage fed to sheep has the potential to improve the nutritional value of mutton by increasing the concentrations of linoleic and linolenic acids and essential amino acids. Sheep fed a diet containing garlic powder or oil are able to produce milk with higher concentrations of the conjugated linoleic acids and n-3 fatty acids, which has health benefits for consumers, due to the widely recognized positive impact of n-3 polyunsaturated fatty acids and conjugated linoleic acids on human heart health, improving platelet aggregation, vasodilation and thrombotic tendency. Overall, garlic products have the potential to enhance growth performance and product quality and reduce parasite infections, as well as methane emissions of ruminants.
Collapse
Affiliation(s)
- He Ding
- Institute of Grassland Research, Chinese Academy of Agricultural Sciences, Hohhot 010010, China
| | - Changjin Ao
- Key Laboratory of Animal Feed and Nutrition of Inner Mongolia Autonomous Region, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xiaoqing Zhang
- Institute of Grassland Research, Chinese Academy of Agricultural Sciences, Hohhot 010010, China
| |
Collapse
|
40
|
Xu Z, Zhu Y, Xie M, Liu K, Cai L, Wang H, Li D, Chen H, Gao L. Mackinawite nanozymes as reactive oxygen species scavengers for acute kidney injury alleviation. J Nanobiotechnology 2023; 21:281. [PMID: 37598162 PMCID: PMC10439570 DOI: 10.1186/s12951-023-02034-7] [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: 04/26/2023] [Accepted: 07/31/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Iron sulfide nanomaterials have been successfully employed as therapeutic agents for bacterial infection therapy and catalytic-ferroptosis synergistic tumor therapy due to their unique structures, physiochemical properties, and biocompatibility. However, biomedical research and understanding of the biological functions of iron sulfides are insufficient, and how iron sulfide nanomaterials affect reactive oxygen species (ROS) in diseases remains unknown. Acute kidney injury (AKI) is associated with high levels of ROS, and therefore nanomedicine-mediated antioxidant therapy has emerged as a novel strategy for its alleviation. RESULTS Here, mackinawite nanozymes were synthesized from glutathione (GSH) and iron ions (Fe3+) (denoted as GFeSNs) using a hydrothermal method, and then evaluated as ROS scavengers for ROS-related AKI treatment. GFeSNs showed broad-spectrum ROS scavenging ability through synergistic interactions of multiple enzymes-like and hydrogen polysulfide-releasing properties. Furthermore, both in vitro and in vivo experiments demonstrated that GFeSNs exhibited outstanding cytoprotective effects against ROS-induced damage at extremely low doses and significantly improved treatment outcomes in AKI. CONCLUSIONS Given the synergetic antioxidant properties and high biocompatibility, GFeSNs exhibit great potential for the treatment of AKI and other ROS-associated diseases.
Collapse
Affiliation(s)
- Zhuobin Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Yufei Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Mengke Xie
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Kankan Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Liangliang Cai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Huihui Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Dandan Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| | - Hao Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
41
|
Huang X, Lu C, Zhang W, Liu L, Zha Z, Miao Z. Chiral Sulfur Nanosheets for Dual-Selective Inhibition of Gram-Positive Bacteria. ACS NANO 2023; 17:14893-14903. [PMID: 37466081 DOI: 10.1021/acsnano.3c03458] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Elemental sulfur is the oldest known antimicrobial agent. However, conventional sulfur in the clinic suffers from poor aqueous solubility and limited antibacterial activity, greatly hindering its practical use. Herein, we report a reform strategy coupling dimension engineering with chirality transfer to convert conventional 3D sulfur particles into chiral 2D sulfur nanosheets (S-NSs), which exhibit 50-fold improvement of antibacterial capability and dual-selective inhibition against Gram-positive bacteria. Benefiting from the inherent selectivity of S-NSs and chirality selectivity from decorated d-histidine, the obtained chiral S-NSs are proven to precisely kill Gram-positive drug-resistant bacteria, while no obvious bacterial inhibition is observed for Gram-negative bacteria. Mechanism studies reveal that S-NSs produce numerous reactive oxygen specipoes and hydrogen sulfide after incubation with bacteria, thus causing bacterial membrane destruction, respiratory chain damage, and ATP production inhibition. Upon spraying chiral S-NSs dispersions onto MRSA-infected wounds, the skin healing process was greatly accelerated in 8 days due to metabolism inhibition and oxidative damage of bacteria, indicating the excellent treatment efficiency of MRSA-infected wounds. This work converts the traditional well-known sulfur into modern antibacterial agents with a superior Gram-selectivity bactericidal capability.
Collapse
Affiliation(s)
- Xiang Huang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| | - Chenxin Lu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| | - Wenjie Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| | - Lulu Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| | - Zhaohua Miao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| |
Collapse
|
42
|
Miao X, Yin Y, Chen Y, Bi W, Yin Y, Chen S, Peng D, Gao L, Qin T, Liu X. Bidirectionally Regulating Viral and Cellular Ferroptosis with Metastable Iron Sulfide Against Influenza Virus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206869. [PMID: 37092591 PMCID: PMC10265104 DOI: 10.1002/advs.202206869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/29/2023] [Indexed: 05/03/2023]
Abstract
Influenza virus with numerous subtypes and frequent variation limits the development of high-efficacy and broad-spectrum antiviral strategy. Here, a novel multi-antiviral metastable iron sulfides (mFeS) against various influenza A/B subtype viruses is developed. This work finds that mFeS induces high levels of lipid peroxidation and •OH free radicals in the conservative viral envelope, which depends on Fe2+ . This phenomenon, termed as a viral ferroptosis, results in the loss of viral infectibility and pathogenicity in vitro and in vivo, respectively. Furthermore, the decoction of mFeS (Dc(mFeS)) inhibits cellular ferroptosis-dependent intracellular viral replication by correcting the virus-induced reprogrammed sulfur metabolism, a conserved cellular metabolism. Notably, personal protective equipment (PPE) that is loaded with mFeS provides good antiviral protection. Aerosol administration of mFeS combined with the decoction (mFeS&Dc) has a potential therapeutic effect against H1N1 lethal infection in mice. Collectively, mFeS represents an antiviral alternative with broad-spectrum activity against intracellular and extracellular influenza virus.
Collapse
Affiliation(s)
- Xinyu Miao
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safetythe Ministry of Education of ChinaYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Yinyan Yin
- College of MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education InstitutionsYangzhou UniversityYangzhouJiangsu225009P. R. China
- Guangling CollegeYangzhou UniversityYangzhouJiangsu225000P. R. China
| | - Yulian Chen
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Wenhui Bi
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Yuncong Yin
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Sujuan Chen
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Daxin Peng
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safetythe Ministry of Education of ChinaYangzhou UniversityYangzhouJiangsu225009P. R. China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou UniversityYangzhouJiangsu225009P. R. China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry DiseaseYangzhouJiangsu225009P. R. China
| | - Lizeng Gao
- CAS Engineering Laboratory for NanozymeInstitute of BiophysicsChinese Academy of SciencesBeijing100101P. R. China
| | - Tao Qin
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safetythe Ministry of Education of ChinaYangzhou UniversityYangzhouJiangsu225009P. R. China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou UniversityYangzhouJiangsu225009P. R. China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry DiseaseYangzhouJiangsu225009P. R. China
| | - Xiufan Liu
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safetythe Ministry of Education of ChinaYangzhou UniversityYangzhouJiangsu225009P. R. China
| |
Collapse
|
43
|
Dediu V, Ghitman J, Gradisteanu Pircalabioru G, Chan KH, Iliescu FS, Iliescu C. Trends in Photothermal Nanostructures for Antimicrobial Applications. Int J Mol Sci 2023; 24:9375. [PMID: 37298326 PMCID: PMC10253355 DOI: 10.3390/ijms24119375] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The rapid development of antimicrobial resistance due to broad antibiotic utilisation in the healthcare and food industries and the non-availability of novel antibiotics represents one of the most critical public health issues worldwide. Current advances in nanotechnology allow new materials to address drug-resistant bacterial infections in specific, focused, and biologically safe ways. The unique physicochemical properties, biocompatibility, and wide range of adaptability of nanomaterials that exhibit photothermal capability can be employed to develop the next generation of photothermally induced controllable hyperthermia as antibacterial nanoplatforms. Here, we review the current state of the art in different functional classes of photothermal antibacterial nanomaterials and strategies to optimise antimicrobial efficiency. The recent achievements and trends in developing photothermally active nanostructures, including plasmonic metals, semiconductors, and carbon-based and organic photothermal polymers, and antibacterial mechanisms of action, including anti-multidrug-resistant bacteria and biofilm removal, will be discussed. Insights into the mechanisms of the photothermal effect and various factors influencing photothermal antimicrobial performance, emphasising the structure-performance relationship, are discussed. We will examine the photothermal agents' functionalisation for specific bacteria, the effects of the near-infrared light irradiation spectrum, and active photothermal materials for multimodal synergistic-based therapies to minimise side effects and maintain low costs. The most relevant applications are presented, such as antibiofilm formation, biofilm penetration or ablation, and nanomaterial-based infected wound therapy. Practical antibacterial applications employing photothermal antimicrobial agents, alone or in synergistic combination with other nanomaterials, are considered. Existing challenges and limitations in photothermal antimicrobial therapy and future perspectives are presented from the structural, functional, safety, and clinical potential points of view.
Collapse
Affiliation(s)
- Violeta Dediu
- National Research and Development Institute in Microtechnologies—IMT Bucharest, 126A Erou Iancu Nicolae Street, 077190 Voluntari, Romania;
| | - Jana Ghitman
- eBio-hub Research-Center, University “Politehnica” of Bucharest, 6 Iuliu Maniu Boulevard, Campus Building, 061344 Bucharest, Romania; (J.G.); (G.G.P.)
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania
| | - Gratiela Gradisteanu Pircalabioru
- eBio-hub Research-Center, University “Politehnica” of Bucharest, 6 Iuliu Maniu Boulevard, Campus Building, 061344 Bucharest, Romania; (J.G.); (G.G.P.)
- Academy of Romanian Scientists, 54 Splaiul Independentei, 050094 Bucharest, Romania
- Research Institute of University of Bucharest, University of Bucharest, 050095 Bucharest, Romania
| | - Kiat Hwa Chan
- Division of Science, Yale-NUS College, 16 College Avenue West, Singapore 138527, Singapore;
- NUS College, National University of Singapore, 18 College Avenue East, Singapore 138593, Singapore
| | - Florina Silvia Iliescu
- National Research and Development Institute in Microtechnologies—IMT Bucharest, 126A Erou Iancu Nicolae Street, 077190 Voluntari, Romania;
| | - Ciprian Iliescu
- National Research and Development Institute in Microtechnologies—IMT Bucharest, 126A Erou Iancu Nicolae Street, 077190 Voluntari, Romania;
- eBio-hub Research-Center, University “Politehnica” of Bucharest, 6 Iuliu Maniu Boulevard, Campus Building, 061344 Bucharest, Romania; (J.G.); (G.G.P.)
- Academy of Romanian Scientists, 54 Splaiul Independentei, 050094 Bucharest, Romania
| |
Collapse
|
44
|
Duan Y, Sun J. Preparation of Iron-Based Sulfides and Their Applications in Biomedical Fields. Biomimetics (Basel) 2023; 8:biomimetics8020177. [PMID: 37218763 DOI: 10.3390/biomimetics8020177] [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: 03/15/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Recently, iron-based sulfides, including iron sulfide minerals and biological iron sulfide clusters, have attracted widespread interest, owing to their excellent biocompatibility and multi-functionality in biomedical applications. As such, controlled synthesized iron sulfide nanomaterials with elaborate designs, enhanced functionality and unique electronic structures show numerous advantages. Furthermore, iron sulfide clusters produced through biological metabolism are thought to possess magnetic properties and play a crucial role in balancing the concentration of iron in cells, thereby affecting ferroptosis processes. The electrons in the Fenton reaction constantly transfer between Fe2+ and Fe3+, participating in the production and reaction process of reactive oxygen species (ROS). This mechanism is considered to confer advantages in various biomedical fields such as the antibacterial field, tumor treatment, biosensing and the treatment of neurodegenerative diseases. Thus, we aim to systematically introduce recent advances in common iron-based sulfides.
Collapse
Affiliation(s)
- Yefan Duan
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210009, China
| | - Jianfei Sun
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210009, China
| |
Collapse
|
45
|
Karnwal A, Kumar G, Pant G, Hossain K, Ahmad A, Alshammari MB. Perspectives on Usage of Functional Nanomaterials in Antimicrobial Therapy for Antibiotic-Resistant Bacterial Infections. ACS OMEGA 2023; 8:13492-13508. [PMID: 37091369 PMCID: PMC10116640 DOI: 10.1021/acsomega.3c00110] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/28/2023] [Indexed: 05/03/2023]
Abstract
The clinical applications of nanotechnology are emerging as widely popular, particularly as a potential treatment approach for infectious diseases. Diseases associated with multiple drug-resistant organisms (MDROs) are a global concern of morbidity and mortality. The prevalence of infections caused by antibiotic-resistant bacterial strains has increased the urgency associated with researching and developing novel bactericidal medicines or unorthodox methods capable of combating antimicrobial resistance. Nanomaterial-based treatments are promising for treating severe bacterial infections because they bypass antibiotic resistance mechanisms. Nanomaterial-based approaches, especially those that do not rely on small-molecule antimicrobials, display potential since they can bypass drug-resistant bacteria systems. Nanoparticles (NPs) are small enough to pass through the cell membranes of pathogenic bacteria and interfere with essential molecular pathways. They can also target biofilms and eliminate infections that have proven difficult to treat. In this review, we described the antibacterial mechanisms of NPs against bacteria and the parameters involved in targeting established antibiotic resistance and biofilms. Finally, yet importantly, we talked about NPs and the various ways they can be utilized, including as delivery methods, intrinsic antimicrobials, or a mixture.
Collapse
Affiliation(s)
- Arun Karnwal
- Department
of Microbiology, School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Gaurav Kumar
- Department
of Microbiology, School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Gaurav Pant
- Department
of Microbiology, Graphic Era (Deemed to
be University), Dehradun, Uttarakhand 248002, India
| | - Kaizar Hossain
- Department
of Environmental Science, Asutosh College, University of Calcutta, 92, Shyama Prasad Mukherjee Road, Bhowanipore, Kolkata 700026, West
Bengal, India
| | - Akil Ahmad
- Department
of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohammed B. Alshammari
- Department
of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
46
|
Li Y, Wang Q, Qu X, Zhang Q, Zhang X. A metalloporphyrin and hydantoin functionalized nanozyme with synergistically enhanced bacterial inhibition. Biomater Sci 2023; 11:1785-1796. [PMID: 36648752 DOI: 10.1039/d2bm01337a] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
An elaborate design of multimodal antibacterial agents has been revealed to be a promising strategy to address bacterial resistance, originating from the abuse of antibiotics. In this work, we have developed a positively charged and porous material, FePPOPHydantoin, as a disinfectant via introducing 1,3-dibromo-5,5-dimethylhydantoin (Hydantoin) and porphyrin iron units into a polymer framework. The extended π conjugated networks of FePPOPHydantoin endowed the material with strong near-infrared (NIR) absorption, high density of surface catalytic active centers, superior stability, and reproducibility. FePPOPHydantoin exhibits high peroxidase mimetic and photo-Fenton activity, which can catalyze the biologically allowable maximum concentrations of hydrogen peroxide (100 μM) to produce a vast amount of hydroxyl radicals. Simultaneously, the effective electrostatic interaction between the positively charged FePPOPHydantoin and the negatively charged bacteria facilitates the binding of FePPOPHydantoin on the bacterial membrane, restricting bacteria within the destruction range of hydroxyl radicals and thus making the bacteria more vulnerable. Finally, further close contact between bacteria and Hydantoin units in FePPOPHydantoin gave the material an antibacterial efficiency of over 99.999%. Compared with chemical therapy, photo-Fenton therapy, or peroxidase catalytic therapy alone, FePPOPHydantoin had a noteworthy multi-amplified antibacterial efficiency. Furthermore, FePPOPHydantoin exhibited good biocompatibility and negligible cytotoxicity. The in vivo antibacterial therapy on the Staphylococcus aureus (S. aureus) infected mouse wound model clearly proved the effectiveness of FePPOPHydantoin for fighting bacterial infections. This work highlights opportunities for the design of nanozymes with enhanced bacteriostatic activity, providing a new avenue for the construction of novel antibiotics.
Collapse
Affiliation(s)
- Yanhong Li
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Quanbo Wang
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China.
| | - Xinyan Qu
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China.
| | - Qiang Zhang
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Xiaomei Zhang
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| |
Collapse
|
47
|
Rong F, Wang T, Zhou Q, Peng H, Yang J, Fan Q, Li P. Intelligent polymeric hydrogen sulfide delivery systems for therapeutic applications. Bioact Mater 2023; 19:198-216. [PMID: 35510171 PMCID: PMC9034248 DOI: 10.1016/j.bioactmat.2022.03.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 12/21/2022] Open
Abstract
Hydrogen sulfide (H2S) plays an important role in regulating various pathological processes such as protecting mammalian cell from harmful injuries, promoting tissue regeneration, and regulating the process of various diseases caused by physiological disorders. Studies have revealed that the physiological effects of H2S are highly associated with its concentrations. At relatively low concentration, H2S shows beneficial functions. However, long-time and high-dose donation of H2S would inhibit regular biological process, resulting in cell dysfunction and apoptosis. To regulate the dosage of H2S delivery for precision medicine, H2S delivery systems with intelligent characteristics were developed and a variety of biocompatibility polymers have been utilized to establish intelligent polymeric H2S delivery systems, with the abilities to specifically target the lesions, smartly respond to pathological microenvironments, as well as real-timely monitor H2S delivery and lesion conditions by incorporating imaging-capable moieties. In this review, we focus on the design, preparation, and therapeutic applications of intelligent polymeric H2S delivery systems in cardiovascular therapy, inflammatory therapy, tissue regenerative therapy, cancer therapy and bacteria-associated therapy. Strategies for precise H2S therapies especially imaging-guided H2S theranostics are highlighted. Since H2S donors with stimuli-responsive characters are vital components for establishing intelligent H2S delivery systems, the development of H2S donors is also briefly introduced. H2S is an endogenous gasotransmitter that plays important role in regulating various physiological and pathological pathways. Controlled H2S delivery is vital since the therapeutic effects of H2S are highly associated with its concentrations. Intelligent polymeric H2S delivery systems possess specific targeting, stimuli responsive and imaging guided capabilities, representing a strategic option for next generation of therapies.
Collapse
|
48
|
Han S, Li Y, Gao H. Generation and Physiology of Hydrogen Sulfide and Reactive Sulfur Species in Bacteria. Antioxidants (Basel) 2022; 11:antiox11122487. [PMID: 36552695 PMCID: PMC9774590 DOI: 10.3390/antiox11122487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Sulfur is not only one of the most abundant elements on the Earth, but it is also essential to all living organisms. As life likely began and evolved in a hydrogen sulfide (H2S)-rich environment, sulfur metabolism represents an early form of energy generation via various reactions in prokaryotes and has driven the sulfur biogeochemical cycle since. It has long been known that H2S is toxic to cells at high concentrations, but now this gaseous molecule, at the physiological level, is recognized as a signaling molecule and a regulator of critical biological processes. Recently, many metabolites of H2S, collectively called reactive sulfur species (RSS), have been gradually appreciated as having similar or divergent regulatory roles compared with H2S in living organisms, especially mammals. In prokaryotes, even in bacteria, investigations into generation and physiology of RSS remain preliminary and an understanding of the relevant biological processes is still in its infancy. Despite this, recent and exciting advances in the fields are many. Here, we discuss abiotic and biotic generation of H2S/RSS, sulfur-transforming enzymes and their functioning mechanisms, and their physiological roles as well as the sensing and regulation of H2S/RSS.
Collapse
|
49
|
Shi S, Zhang Q, Sun H, Su Z, Dan J, Liang Y, Kang Y, Du T, Sun J, Wang J, Zhang W. Glucose Oxidase-Integrated Metal-Polyphenolic Network as a Microenvironment-Activated Cascade Nanozyme for Hyperglycemic Wound Disinfection. ACS Biomater Sci Eng 2022; 8:5145-5154. [PMID: 36344935 DOI: 10.1021/acsbiomaterials.2c00985] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The high systemic blood glucose concentration of hyperglycemic wound microenvironment (WME) severely impedes the disinfection and healing of infected skin wounds. Herein, a WME-activated smart natural product, integrated GOx-GA-Fe nanozyme (GGFzyme), is engineered, which combines a nanozyme and natural enzyme to promote reactive oxygen species (ROS) generation in situ for hyperglycemic wound disinfection. GGFzyme can consume a high concentration of glucose in hyperglycemia wounds and generate H2O2. The conversion of glucose into gluconic acid not avails starvation treatment but reduces the pH of WME to elevate the catalytic activities of both the nanozyme (GA-Fe) and natural enzyme (GOx). And H2O2 is then high efficiently catalyzed into •OH and O2•- in situ to combat pathogenic bacteria and promote wound disinfection. The high catalytic antibacterial capacity and superior biosafety, combined with beneficial WME modulation, demonstrate that GGFzyme is a promising therapeutic agent for hyperglycemic wounds.
Collapse
Affiliation(s)
- Shuo Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Qiuping Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Hao Sun
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Zehui Su
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Jie Dan
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Yanmin Liang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Yi Kang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Ting Du
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Jing Sun
- Qinghai Provincial Key Laboratory of Qinghai-Tibet Plateau Biological Resources, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, Qinghai, China
| | - Jianlong Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Wentao Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| |
Collapse
|
50
|
Cong Y, Baimanov D, Zhou Y, Chen C, Wang L. Penetration and translocation of functional inorganic nanomaterials into biological barriers. Adv Drug Deliv Rev 2022; 191:114615. [PMID: 36356929 DOI: 10.1016/j.addr.2022.114615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/23/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
With excellent physicochemical properties, inorganic nanomaterials (INMs) have exhibited a series of attractive applications in biomedical fields. Biological barriers prevent successful delivery of nanomedicine in living systems that limits the development of nanomedicine especially for sufficient delivery of drugs and effective therapy. Numerous researches have focused on overcoming these biological barriers and homogeneity of organisms to enhance therapeutic efficacy, however, most of these strategies fail to resolve these challenges. In this review, we present the latest progress about how INMs interact with biological barriers and penetrate these barriers. We also summarize that both native structure and components of biological barriers and physicochemical properties of INMs contributed to the penetration capacity. Knowledge about the relationship between INMs structure and penetration capacity will guide the design and application of functional and efficient nanomedicine in the future.
Collapse
Affiliation(s)
- Yalin Cong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China & Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Didar Baimanov
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China & Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China; Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, PR China
| | - Yunlong Zhou
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, PR China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China & Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; GBA Research Innovation Institute for Nanotechnology, Guangzhou 510700, Guangdong, PR China; Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | - Liming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China & Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|