1
|
Qi Y, Ren S, Ou X, Li P, Wu H, Che Y, Wang X. Ultrasound-activated sonothermal-catalytic synergistic therapy via asymmetric electron distribution for bacterial wound infections. Biomaterials 2025; 321:123338. [PMID: 40239594 DOI: 10.1016/j.biomaterials.2025.123338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
Antibiotic-resistant bacterial infections present a growing global health challenge, requiring innovative therapeutic solutions to overcome current limitations. We introduce boron-integrated bismuth oxide (B-BiO2) nanosheets with an asymmetrically distributed electronic structure for ultrasound-activated synergistic sonothermal and catalytic therapy. Boron incorporation enhances local electron density distribution, optimizing charge separation and significantly improving sonothermal and catalytic efficiency, as validated by density functional theory calculations. These nanosheets exhibit dual functionality, effectively generating localized heat and reactive oxygen species (ROS) under ultrasound, leading to 99.999 % antibacterial efficacy against multidrug-resistant pathogens by disrupting bacterial membranes, as demonstrated through all-atom simulations and in vitro experiments. The simulations further reveal that sonothermal conversion effects enhance bacterial membrane fluidity and induce structural defects, amplifying ROS-induced oxidative damage and membrane destabilization. In vivo, B-BiO2 nanosheets accelerate wound healing in methicillin-resistant Staphylococcus aureus (MRSA)-infected murine models, achieving 99.8 % closure by day 14 by reducing inflammation and promoting angiogenesis and tissue regeneration. Transcriptomic analysis highlights the activation of extracellular matrix remodeling, angiogenesis, and autophagy pathways, underscoring the nanosheets' therapeutic potential. This study establishes ultrasound-activated B-BiO2 nanosheets as a novel nanotherapeutic platform, leveraging asymmetric electron distribution to synergistically combat drug-resistant infections and promote effective wound healing.
Collapse
Affiliation(s)
- Ye Qi
- Research Institute of Biomedical and Advanced Materials, College of Life and Health, Dalian University, 10 Xuefu Street, Dalian, Liaoning, 116622, China.
| | - Shuangsong Ren
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, 193 Lianhe Road, Dalian, Liaoning, 116011, China
| | - Xiaolong Ou
- Research Institute of Biomedical and Advanced Materials, College of Life and Health, Dalian University, 10 Xuefu Street, Dalian, Liaoning, 116622, China
| | - Pisong Li
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, 6 Jiefang Street, Dalian, Liaoning, 116001, China
| | - Han Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, 193 Lianhe Road, Dalian, Liaoning, 116011, China
| | - Ying Che
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, 193 Lianhe Road, Dalian, Liaoning, 116011, China.
| | - Xinyi Wang
- Research Institute of Biomedical and Advanced Materials, College of Life and Health, Dalian University, 10 Xuefu Street, Dalian, Liaoning, 116622, China.
| |
Collapse
|
2
|
Obeid MA, Alyamani H, Alenaizat A, Tunç T, Aljabali AAA, Alsaadi MM. Nanomaterial-based drug delivery systems in overcoming bacterial resistance: Current review. Microb Pathog 2025; 203:107455. [PMID: 40057006 DOI: 10.1016/j.micpath.2025.107455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Antimicrobial resistance is one of the most serious contemporary global health concerns, threatening the effectiveness of existing antibiotics and resulting in morbidity, mortality, and economic burdens. This review examines the contribution of nanomaterial-based drug delivery systems to solving the problems associated with bacterial resistance and provides a thorough overview of their mechanisms of action, efficiency, and perspectives for the future. Owing to their unique physicochemical properties, nanomaterials reveal new ways of passing through the traditional mechanisms of bacterial defence connected to the permeability barrier of membranes, efflux pumps, and biofilm formation. This review addresses the different types of nanomaterials, including metallic nanoparticles, liposomes, and polymeric nanoparticles, in terms of their antimicrobial properties and modes of action. More emphasis has been placed on the critical discussion of recent studies on such active systems. Both in vitro and in vivo models are discussed, with particular attention paid to multidrug-resistant bacteria. This review begins by reviewing the urgency for antimicrobial resistance (AMR) by citing recent statistics, which indicate that the number of deaths and reasons for financial losses continue to increase. A background is then provided on the limitations of existing antibiotic therapies and the pressing need to develop innovative approaches. Nanomaterial-based drug delivery systems have been proposed as promising solutions because of their potential to improve drug solubility, stability, and targeted delivery, although side effects can also be mitigated. In addition to established knowledge, this review also covers ongoing debates on the continuous risks associated with the use of nanomaterials, such as toxicity and environmental impact. This discussion emphasizes the optimization of nanomaterial design to target specific bacteria, and rigorous clinical trials to establish safety and efficacy in humans. It concludes with reflections on the future directions of nanomaterial-based drug delivery systems in fighting AMR, underlining the need for an interdisciplinary approach, along with continuous research efforts to translate these promising technologies into clinical practice. As the fight against bacterial resistance reaches its peak, nanomaterials may be the key to developing next-generation antimicrobial therapies.
Collapse
Affiliation(s)
- Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O.BOX 566, Irbid, 21163, Jordan.
| | - Hanin Alyamani
- William Harvey Research Institute, Center for Microvascular Research, Queen Mary University of London, London, United Kingdom
| | | | - Tutku Tunç
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O.BOX 566, Irbid, 21163, Jordan
| | - Manal M Alsaadi
- Department of Industrial Pharmacy, Faculty of Pharmacy, University of Tripoli, PO Box, Tripoli, 13645, Libya
| |
Collapse
|
3
|
Xia Y, Yang L, Xu S, Xia Y, Peng L, Wu Y, Han X, Qin C, Li S, Yin L. Trapping effect of surface deficient cocrystal synergizes with bimetallic nanoparticles against bacterial infection in wounds. J Colloid Interface Sci 2025; 695:137805. [PMID: 40339286 DOI: 10.1016/j.jcis.2025.137805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
Metal nanoparticles exert broad-spectrum antimicrobial effects through in situ generation of reactive oxygen species (ROS). However, the limited penetrability of ROS restricts the scope of antimicrobial activity of these nanoparticles. Herein, we develop core-shell nanoparticles composed of a surface-defective cocrystal shell and a Cu/Zn bimetallic core. Surface defects are generated by etching myricetin from a berberine-dihydromyricetin cocrystal, and bacterial adsorption efficiency peaks when the cocrystal contains 40 % myricetin. Similarly, Cu2+ doping on the surface of Zn nanoparticles to form a bimetallic core can optimize the ROS generation efficiency by facilitating effective electron transfer. Nanoparticles with this combined core-shell structure not only capture bacteria effectively but also draw bacteria into the ROS-killing range of the metal particles, thereby enhancing activity against drug-resistant bacteria. The feasibility of antibacterial infection was further validated in wounds of mice infected with Escherichia coli and Staphylococcus aureus. This strategy could be used to optimize antimicrobial nanoparticles with ROS-generating functionality and could have an impact on combating bacterial resistance.
Collapse
Affiliation(s)
- Yanming Xia
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Lei Yang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Shuxian Xu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yunhan Xia
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Linxiu Peng
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yutong Wu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaopeng Han
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chao Qin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Suxin Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Lifang Yin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
4
|
Chugh S, Létisse F, Neyrolles O. The exometabolome as a hidden driver of bacterial virulence and pathogenesis. Trends Microbiol 2025; 33:546-557. [PMID: 39701858 DOI: 10.1016/j.tim.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024]
Abstract
The traditional view of metabolism as merely supplying energy and biosynthetic precursors is undergoing a paradigm shift. Metabolic dynamics not only regulates gene expression but also orchestrates cellular processes with remarkable precision. Most bacterial pathogens exhibit exceptional metabolic plasticity, enabling them to adapt to diverse environments, including hostile conditions within a host. While the role of intracellular bacterial metabolism in pathogen-host interactions has been extensively studied, the contributions of the extracellularly released or secreted bacterial metabolites (referred to here as the bacterial 'exometabolome') to metabolic adaptations and disease pathogenesis remain largely unexplored. In this review, we highlight the significant and intriguing roles of bacterial exometabolomes in drug tolerance, immune suppression, and disease pathogenesis, opening a new frontier in our understanding of bacterial-host interactions.
Collapse
Affiliation(s)
- Saurabh Chugh
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Fabien Létisse
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
5
|
Li X, Dong S, Pan Q, Liu N, Zhang Y. Antibiotic conjugates: Using molecular Trojan Horses to overcome drug resistance. Biomed Pharmacother 2025; 186:118007. [PMID: 40268370 DOI: 10.1016/j.biopha.2025.118007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 04/25/2025] Open
Abstract
Antimicrobial resistance (AMR) has become a global health crisis due to the rapid emergence of multi-drug-resistant bacteria. The paucity of novel antibiotics in the clinical pipeline has exacerbated this issue, thereby warranting the development of new antibacterial therapies. The 'Trojan Horse' strategy entails conjugating antibiotics with bioactive components that not only facilitate the entry of antibiotic molecules into bacterial cells by circumventing the membrane barriers, but also augment the effects of conventional antibiotics against recalcitrant pathogens. These Trojan Horse elements can also serve as a promising tool for repurposing drugs with hitherto unexamined antimicrobial activity, or drugs with limited clinical utility due to considerable toxic side effects. In this review, we have discussed the current state of research on antibiotic conjugates with monoclonal antibodies (mAbs), antimicrobial peptides (AMPs) and the iron-chelating siderophores. The rationale and mechanisms of different antibiotic conjugates have been summarized, and the preclinical and clinical evidence pertaining to the activity of these conjugates against drug-resistant pathogens have been reviewed. Furthermore, the challenges associated with the clinical translation of these novel antimicrobials, and the future research directions have also been discussed. While antibiotic conjugates offer an attractive alternative to conventional antimicrobials, there are several obstacles to their clinical translation. A greater understanding of the mechanisms underlying AMR, and continuing advances in genetic engineering, synthetic biology, and bioinformatics will be crucial in designing more selective, potent, and safe antibiotic conjugates for tackling multi-drug resistant (MDR) infections.
Collapse
Affiliation(s)
- Xi Li
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyuan Dong
- Department of Thoracic surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qi Pan
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China
| | - Ning Liu
- Department of Rehabilitation, the First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | - Yijie Zhang
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China.
| |
Collapse
|
6
|
Liu X, Zhao X, Qiu H, Liang W, Liu L, Sun Y, Zhao L, Wang X, Liang H. Antibacterial Activity of GO-Based Composites Enhanced by Phosphonate-Functionalized Ionic Liquids and Silver. MATERIALS (BASEL, SWITZERLAND) 2025; 18:1889. [PMID: 40333535 PMCID: PMC12028358 DOI: 10.3390/ma18081889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025]
Abstract
The development of antibiotic-independent antimicrobial materials is critical for addressing bacterial resistance to conventional antibiotics. Currently, there is a lack of comprehensive understanding of ionic liquid-modified composites in antimicrobial applications. Here, we innovatively prepared GO-based composites modified with phosphonate ionic liquids via a series of surface functionalizations. The resulting antibacterial composites exhibit significant broad-spectrum activity against both Gram-negative and Gram-positive bacteria, including drug-resistant strains, with stronger efficacy against Gram-negative species. Additionally, the material features excellent long-term reusability and the ability to inhibit/destroy biofilms, which is vital for combating persistent infections. Mechanistic studies reveal its antibacterial effects through multiple pathways: disrupting bacterial membranes, inducing ROS, and inactivating intracellular substances-mechanisms less likely to promote resistance. Overall, these phosphonate ionic liquid-modified polycationic materials demonstrate substantial potential in treating bacterial infections, offering a promising strategy to tackle antibiotic resistance challenges.
Collapse
Affiliation(s)
- Xinyu Liu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Xing Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Hongda Qiu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Weida Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Linlin Liu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Yunyu Sun
- Anhui Key Laboratory of Spin Electron and Nanomaterials of Anhui Higher Education Institutes, School of Chemistry and Chemical Engineering, Suzhou University, Suzhou 234000, China;
| | - Lingling Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| | - Xiao Wang
- Health Science Center, Ningbo University, Ningbo 315211, China;
| | - Hongze Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China; (X.Z.); (H.Q.); (W.L.); (L.L.); (L.Z.)
| |
Collapse
|
7
|
Ito H, Wada T, Ichinose G, Tanimoto J, Yoshimura J, Yamamoto T, Morita S. Barriers to the widespread adoption of diagnostic artificial intelligence for preventing antimicrobial resistance. Sci Rep 2025; 15:13113. [PMID: 40240443 PMCID: PMC12003763 DOI: 10.1038/s41598-025-95110-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Currently, antimicrobial resistance (AMR) poses a major public health challenge. The emergence of AMR, which significantly threatens public health, is primarily due to the overuse of antimicrobial agents. This study explored the possibility that the ethical dilemmas inherent in the context of AMR may hinder the adoption of diagnostic artificial intelligence (AI). We conducted a web survey across eight countries/areas to assess public preference between two hypothetical AI types: one prioritizing individual health and the other considering the global AMR threat. Our results revealed a societal preference for the utilization of both AI types, reflecting a conflict between recognizing the significance of AMR and the desire for individualized treatment. Interestingly, the survey indicated significant gender and age differences in AI preferences, and the majority of respondents opposed the idea of AI standardization in treatment. These findings highlight the challenges of incorporating AI into public health and the necessity of considering public sentiment in addressing global health issues such as AMR.
Collapse
Affiliation(s)
- Hiromu Ito
- Department of International Health and Medical Anthropology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.
| | - Takayuki Wada
- Graduate School of Human Life and Ecology, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Genki Ichinose
- Department of Mathematical and Systems Engineering, Shizuoka University, Shizuoka, Japan
| | - Jun Tanimoto
- Department of Energy and Environmental Engineering, Interdisciplinary Graduate School of Engineering Sciences, Kyushu University, Fukuoka, Japan
- Department of Advanced Environmental Science and Engineering, Faculty of Engineering Sciences, Kyushu University, Fukuoka, Japan
| | - Jin Yoshimura
- Department of International Health and Medical Anthropology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- Marine Biosystems Research Center, Chiba University, Chiba, Japan
- Department of Biological Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Taro Yamamoto
- Department of International Health and Medical Anthropology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Satoru Morita
- Department of Mathematical and Systems Engineering, Shizuoka University, Shizuoka, Japan
| |
Collapse
|
8
|
Hui J, Moon W, Dong PT, Dos Anjos C, Negri L, Yan H, Wang Y, Tam J, Dai T, Anderson RR, Goverman J, Gelfand JA, Yun SH. Low-Irradiance Antimicrobial Blue Light-Bathing Therapy for Wound Infection Control. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412493. [PMID: 40229974 DOI: 10.1002/advs.202412493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/14/2025] [Indexed: 04/16/2025]
Abstract
The prevalence of antibiotic resistance and tolerance in wound infection management poses a serious and growing health threat, necessitating the exploration of alternative approaches. Antimicrobial blue light therapy offers an appealing, non-pharmacological solution. However, its practical application has been hindered by the requirement for high irradiance levels (50-200 mW/cm2), which particularly raises safety concerns. Here, a light-bathing strategy is introduced that employs prolonged, continuous exposure to blue light at an irradiance range lower by more than an order of magnitude (5 mW/cm2). This method consistently applies bacteriostatic pressure, keeping wound bioburden low, all while minimizing photothermal risks. Leveraging tailor-made, wearable light-emitting patches, preclinical trials on rat models of wound infection are conducted, demonstrating its safety and efficacy for suppressing infections induced by methicillin-resistant Staphylococcus aureus (S. aureus) and multidrug-resistant Pseudomonas aeruginosa (P. aeruginosa). The results pave a new way for the application of blue light therapy in wound care.
Collapse
Affiliation(s)
- Jie Hui
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Wonjoon Moon
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pu-Ting Dong
- Department of Microbiology, The ADA Forsyth Institute, Boston, MA, 02142, USA
| | - Carolina Dos Anjos
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Laisa Negri
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Hao Yan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Ying Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Joshua Tam
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, 02139, USA
| | - Tianhong Dai
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - R Rox Anderson
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jeremy Goverman
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
- Wound Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Jeffrey A Gelfand
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Seok-Hyun Yun
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02139, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
9
|
Chen Z, Li C, Su J, He Z, Xu J, Bian Y, Kim H, Guan X. Enhanced sulfamethoxazole removal in water and wastewater by ferrate(VI)/perborate system via borate buffering. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138261. [PMID: 40222061 DOI: 10.1016/j.jhazmat.2025.138261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Ferrate(VI) is prone to self-decomposition in water, leading to the loss of active substances Fe(V) and Fe(IV). Therefore, the use of Fe(VI) alone has limited practical applicability in municipal wastewater and industrial wastewater treatment scenarios due to its insufficient pollutant removal efficiency. This study discussed the removal efficiency of the Fe(VI)/perborate system for sulfamethoxazole and other 6 drugs in pure water within 5 minutes, and the removal efficiency of the Fe(VI)/perborate system for sulfamethoxazole in effluent and secondary effluent in a wastewater treatment plant (WWTP) within 5 minutes. Results show that the sulfamethoxazole removal efficiency reached 88.63 % in the influent of Lijiang B WWTP and 79 % in the secondary effluent of Wuhan WWTP. The removal efficiency of sulfamethoxazole in pure water reached 25.8 % in 5 minutes. This finding is explained by the buffering with the borate produced by the hydrolysis of Fe(VI)/perborate, which maintains the pH around 9 and, in turn, inhibits the reduction of active species. Moreover, H2O2, 1O2, and O2•- radicals generated by Fe(VI)/perborate accelerate the activation of Fe(VI), and the Fe2+ produced in the system participates in Fenton reactions with H2O2. This study offers a novel approach for using ferrate in practical water treatment.
Collapse
Affiliation(s)
- Zihe Chen
- School of Environment and Architecture, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Cong Li
- School of Environment and Architecture, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Jingzhen Su
- School of Environment and Architecture, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Zhenming He
- School of Environment and Architecture, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jiani Xu
- School of Environment and Architecture, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yulin Bian
- School of Environment and Architecture, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Hyunook Kim
- University of Seoul, Dept. of Environmental Engineering, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, South Korea
| | - Xiaohong Guan
- Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, Institute of Eco-Chongming, School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
10
|
Miao ZY, Lin J, Chen WM. Natural sideromycins and siderophore-conjugated natural products as inspiration for novel antimicrobial agents. Eur J Med Chem 2025; 287:117333. [PMID: 39892091 DOI: 10.1016/j.ejmech.2025.117333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/03/2025]
Abstract
The widespread emergence of multidrug-resistant (MDR) Gram-negative pathogens has posed a major challenge to clinical anti-infective therapy, and new effective treatments are urgently needed. A promising "Trojan horse" strategy involves conjugating antibiotics to siderophore molecules; the resulting siderophore-antibiotic conjugates (SACs) deliver antibiotics directly into cells by hijacking the sophisticated iron transport systems of Gram-negative bacteria, bypassing the outer membrane permeability barrier to enhance uptake and antibacterial efficacy. The clinical release of the first siderophore-antibiotic conjugate, cefiderocol, has aroused tremendous interest in the field among researchers and pharmaceutical companies. To date, most of the reported SACs have focused on the conjugation of siderophores to traditional antibacterial drugs. However, these antibacterial agents designed on the basis of the traditional antibiotic skeleton theoretically bear the risk of cross-resistance caused by shared molecular scaffolds. In this case, exploring novel natural product antibacterial conjugate scaffolds to circumvent the risk of early cross-resistance represents a presumably more sustainable approach for the development of SACs. In this review, we systematically summarize the research progress on siderophore-natural product conjugates as novel antimicrobial agents reported since 2010. Additionally, we propose challenges to be overcome and prospects for future development in this field.
Collapse
Affiliation(s)
- Zhi-Ying Miao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China
| | - Jing Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China.
| | - Wei-Min Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China.
| |
Collapse
|
11
|
Panda I, Raut S, Samal SK, Behera SK, Pradhan S. Evaluation of 1-vinyl-3-alkyl imidazolium-based ionic liquid monomers towards antibacterial activity: An in-silico & in-vitro study. Comput Biol Chem 2025; 115:108288. [PMID: 39642541 DOI: 10.1016/j.compbiolchem.2024.108288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/22/2024] [Accepted: 11/20/2024] [Indexed: 12/09/2024]
Abstract
In this study 1-vinyl-3-alkyl imidazolium-based ionic liquid monomers (ILs) with different alkyl chain lengths {R = hexyl (A), octyl (B) and decyl (C)} have been synthesized for antibacterial applications. The prepared ILs have been characterized using UV, FT-IR and NMR spectroscopy. The antibacterial activities of the synthesized ILs against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) have been examined by measuring their minimal inhibitory concentrations (MICs) and minimum bactericidal concentrations (MBCs). The results exhibit that these ILs have admirable antibacterial activities with MIC values range from < 1.2 to 12.2 μM for S. aureus and < 2.4 to 12.2 μM for E. coli. A notable dependence of antibacterial and antibiofilm efficacy on the alkyl chain length (ILC> ILB > ILA) has been observed. From in-silico evaluation, the binding energies of β-lactamase protein of S. aureus (PDB ID: 1GHP) are found to be -4.4, -4.6, -4.7 kcal/mol for IL A, IL B, and IL C. For dihydrofolate reductase (DHFR) of S. aureus and E. coli the binding energies -4.6, -4.5, -5.3 kcal/mol and -5.3, -5.4, -5.6 kcal/mol have been noted for IL A, IL B, and IL C respectively. MD simulations (100 ns) have been performed to predict the stability and understand the binding mechanism of the docked complexes.
Collapse
Affiliation(s)
- Itishree Panda
- Department of Chemistry, Faculty of Engineering and Technology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar 751030, India
| | - Sangeeta Raut
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003, India
| | - Sangram Keshari Samal
- Laboratory of Biomaterials and Regenerative Medicine for Advanced Therapies, ICMR-Regional Medical Research Center, Bhubaneswar, Odisha 751023, India
| | - Santosh Kumar Behera
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat 382355, India.
| | - Sanghamitra Pradhan
- Department of Chemistry, Faculty of Engineering and Technology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar 751030, India.
| |
Collapse
|
12
|
Zhu CY, Byun H, Do EA, Zhang Y, Tanchoco E, Beld J, Hsiao A, Zhu J. Music exposure enhances resistance to Salmonella infection by promoting healthy gut microbiota. Microbiol Spectr 2025; 13:e0237724. [PMID: 40130867 PMCID: PMC12054044 DOI: 10.1128/spectrum.02377-24] [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: 09/20/2024] [Accepted: 02/28/2025] [Indexed: 03/26/2025] Open
Abstract
Music intervention is gaining recognition as a cost-effective therapeutic for improving human health. Despite its growing application, the mechanisms through which music exerts beneficial health effects remain largely unexplored. Here, we show that music can exert beneficial effects in mice through modulating gut microbiome composition. Adult mice were exposed to ambient noise, Mozart's Flute Quartet in D Major, K. 285, or white noise over a three-week period. Afterward, we observed treatment-specific changes in the community of gut commensal bacteria in these animals. Upon subsequent challenge with the bacterial pathogen Salmonella typhimurium, control groups exhibited significant weight loss and increased Salmonella colonization, whereas the Mozart-treated group did not. 16S ribosomal RNA gene sequencing revealed that the Mozart group showed a significant increase in Lactobacillus salivarius, a probiotic known for its antibacterial properties. Further experiments confirmed that L. salivarius mitigated Salmonella infection in mice and that L. salivarius acidified local environments in in vitro culture, thus inhibiting Salmonella growth. Additionally, mice exposed to Mozart consumed more food but showed similar body weight compared to the control groups. Behavioral assessments, including open field and object location tests, revealed that Mozart-treated mice were more active, less anxious, and exhibited enhanced spatial memory. Finally, Mozart exposure was shown to significantly boost colonization of administered L. salivarius and alter gut metabolite profiles. These findings suggest that music exposure fosters healthier gut microbiota, enhancing resistance to bacterial infections and highlighting the potential of music therapy as a novel strategy to combat drug-resistant pathogen infections. IMPORTANCE Music therapy is increasingly recognized as a low-cost approach to improving health, but how it works remains unclear. Our study demonstrates that music can positively influence health by altering the gut microbiome. In a mouse model, exposure to Mozart's Flute Quartet in D Major enhanced the gut microbiota, specifically increasing levels of the beneficial bacterium Lactobacillus salivarius. This probiotic protected mice from Salmonella infection by creating an acidic environment that inhibited pathogen growth. Mozart-treated mice also showed reduced anxiety, better spatial memory, and higher food intake without weight gain, suggesting the benefits of music exposure. These findings reveal a novel link between music, gut health, and disease resistance, suggesting that music therapy could be a promising strategy for enhancing gut microbiota and combating infections, including those caused by drug-resistant bacteria.
Collapse
Affiliation(s)
- Clara Y. Zhu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Hyuntae Byun
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elyza A. Do
- Department of Microbiology & Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Yue Zhang
- Department of Microbiology & Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Ethan Tanchoco
- Department of Microbiology & Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Joris Beld
- Department of Microbiology & Immunology, College of Medicine, Drexel University, Philadelphia, Pennsylvania, USA
| | - Ansel Hsiao
- Department of Microbiology & Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Jun Zhu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Wang Y, Cui Q, Hou Y, He S, Zhao W, Lancuo Z, Sharshov K, Wang W. Metagenomic Insights into the Diverse Antibiotic Resistome of Non-Migratory Corvidae Species on the Qinghai-Tibetan Plateau. Vet Sci 2025; 12:297. [PMID: 40284799 PMCID: PMC12031176 DOI: 10.3390/vetsci12040297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Antibiotic resistance represents a global health crisis with far-reaching implications, impacting multiple domains concurrently, including human health, animal health, and the natural environment. Wild birds were identified as carriers and disseminators of antibiotic-resistant bacteria (ARB) and their associated antibiotic resistance genes (ARGs). A majority of studies in this area have concentrated on migratory birds as carriers for the spread of antibiotic resistance over long distances. However, there has been scant research on the resistome of non-migratory Corvidae species that heavily overlap with human activities, which limits our understanding of antibiotic resistance in these birds and hinders the development of effective management strategies. This study employed a metagenomics approach to examine the characteristics of ARGs and mobile genetic elements (MGEs) in five common Corvidae species inhabiting the Qinghai-Tibetan Plateau. The ARGs were classified into 20 major types and 567 subtypes. Notably, ARGs associated with multidrug resistance, including to macrolide-lincosamide-streptogramins, tetracyclines, beta-lactam, and bacitracin, were particularly abundant, with the subtypes acrB, bacA, macB, class C beta-lactamase, and tetA being especially prevalent. A total of 5 types of MGEs (166 subtypes) were identified across five groups of crows, and transposase genes, which indicated the presence of transposons, were identified as the most abundant type of MGEs. Moreover, some common opportunistic pathogens were identified as potential hosts for these ARGs and MGEs. Procrustes analysis and co-occurrence network analysis showed that the composition of the gut microbiota shaped the ARGs and MGEs, indicating a substantial association between these factors. The primary resistance mechanisms of ARGs in crows were identified as multidrug efflux pumps, alteration of antibiotic targets, and enzymatic inactivation. High-risk ARGs which were found to potentially pose significant risks to public health were also analyzed and resulted in the identification of 81 Rank I and 47 Rank II ARGs. Overall, our study offers a comprehensive characterization of the resistome in wild Corvidae species, enhancing our understanding of the potential public health risks associated with these birds.
Collapse
Affiliation(s)
- You Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China; (Y.W.); (Q.C.); (Y.H.); (Z.L.)
- College of Eco-Environmental Engineering, Qinghai University, Xining 810016, China
| | - Quanchao Cui
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China; (Y.W.); (Q.C.); (Y.H.); (Z.L.)
- College of Eco-Environmental Engineering, Qinghai University, Xining 810016, China
| | - Yuliang Hou
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China; (Y.W.); (Q.C.); (Y.H.); (Z.L.)
- College of Eco-Environmental Engineering, Qinghai University, Xining 810016, China
| | - Shunfu He
- Xining Wildlife Park of Qinghai Province, Xining 810016, China; (S.H.); (W.Z.)
| | - Wenxin Zhao
- Xining Wildlife Park of Qinghai Province, Xining 810016, China; (S.H.); (W.Z.)
| | - Zhuoma Lancuo
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China; (Y.W.); (Q.C.); (Y.H.); (Z.L.)
| | - Kirill Sharshov
- Federal Research Center of Fundamental and Translational Medicine, Novosibirsk 630117, Russia;
| | - Wen Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China; (Y.W.); (Q.C.); (Y.H.); (Z.L.)
| |
Collapse
|
14
|
Hu L, Luo Y, Yang J, Cheng C. Botanical Flavonoids: Efficacy, Absorption, Metabolism and Advanced Pharmaceutical Technology for Improving Bioavailability. Molecules 2025; 30:1184. [PMID: 40076406 PMCID: PMC11902153 DOI: 10.3390/molecules30051184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Flavonoids represent a class of natural plant secondary metabolites with multiple activities including antioxidant, antitumor, anti-inflammatory, and antimicrobial properties. However, due to their structural characteristics, they often exhibit low bioavailability in vivo. In this review, we focus on the in vivo study of flavonoids, particularly the effects of gut microbiome on flavonoids, including common modifications such as methylation, acetylation, and dehydroxylation, etc. These modifications aim to change the structural characteristics of the original substances to enhance absorption and bioavailability. In order to improve the bioavailability of flavonoids, we discuss two feasible methods, namely dosage form modification and chemical modification, and hope that these approaches will offer new insights into the application of flavonoids for human health. In this article, we also introduce the types, plant sources, and efficacy of flavonoids. In conclusion, this is a comprehensive review on how to improve the bioavailability of flavonoids.
Collapse
Affiliation(s)
- Lei Hu
- Jiangxi Key Laboratory for Sustainable Utilization of Chinese Materia Medica Resources, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; (L.H.); (Y.L.); (J.Y.)
- Lushan Xinglin Institute for Medicinal Plants, Jiujiang Xinglin Key Laboratory for Traditional Chinese Medicines, Jiujiang 332900, China
| | - Yiqing Luo
- Jiangxi Key Laboratory for Sustainable Utilization of Chinese Materia Medica Resources, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; (L.H.); (Y.L.); (J.Y.)
- Lushan Xinglin Institute for Medicinal Plants, Jiujiang Xinglin Key Laboratory for Traditional Chinese Medicines, Jiujiang 332900, China
| | - Jiaxin Yang
- Jiangxi Key Laboratory for Sustainable Utilization of Chinese Materia Medica Resources, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; (L.H.); (Y.L.); (J.Y.)
- Lushan Xinglin Institute for Medicinal Plants, Jiujiang Xinglin Key Laboratory for Traditional Chinese Medicines, Jiujiang 332900, China
| | - Chunsong Cheng
- Jiangxi Key Laboratory for Sustainable Utilization of Chinese Materia Medica Resources, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; (L.H.); (Y.L.); (J.Y.)
- Lushan Xinglin Institute for Medicinal Plants, Jiujiang Xinglin Key Laboratory for Traditional Chinese Medicines, Jiujiang 332900, China
| |
Collapse
|
15
|
Xu Z, Liu J, Zhuang Y. The anti-Acinetobacter baumannii therapeutic potential of azole hybrids: A mini-review. Arch Pharm (Weinheim) 2025; 358:e2400592. [PMID: 40040257 DOI: 10.1002/ardp.202400592] [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: 07/19/2024] [Revised: 12/19/2024] [Accepted: 02/11/2025] [Indexed: 03/06/2025]
Abstract
Acinetobacter baumannii is one of the major causes of severe hospital- and community-acquired infections, posing a significant threat to human lives. A. baumannii has already generated resistance to almost all of the currently available antibiotics, but no new class of antibacterials have been launched for the treatment of infections caused by A. baumannii in the last half century, creating an urgent need to develop novel antibacterials. Azoles as a broad class of five-membered nitrogen-containing aromatic heterocycles are privileged pharmacophores widely found in pharmaceuticals. Azoles could target on diverse enzymes, proteins, and receptors in A. baumannii via various noncovalent interactions. Particularly, azole hybrids have potential advantages in increasing therapeutic efficacy and circumventing drug resistance, representing useful scaffolds for the discovery of novel anti-A. baumannii agents. This review outlines the current scenario of the antibacterial therapeutic potential of azole hybrids against A. baumannii, developed from 2020 onwards, aiming to provide potential candidates for further preclinical/clinical evaluations and facilitate the rational design of more effective candidates.
Collapse
Affiliation(s)
- Zhi Xu
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, People's Republic of China
| | - Junna Liu
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, People's Republic of China
| | - Yafei Zhuang
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, People's Republic of China
| |
Collapse
|
16
|
Lu X, Xu X, Ding Y, Gong X, Ming L, Dai X, Gu C, Wang J, Zhao J, Gao M, Yin H, Wang Z, Wang X, Wang L, Zhang D, Zhang M, Huang J. Discovery and optimization of tetrahydroacridine derivatives as a novel class of antibiotics against multidrug-resistant Gram-positive pathogens by targeting type I signal peptidase and disrupting bacterial membrane. Eur J Med Chem 2025; 283:117101. [PMID: 39626521 DOI: 10.1016/j.ejmech.2024.117101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/27/2024] [Accepted: 11/08/2024] [Indexed: 01/03/2025]
Abstract
Increasing antimicrobial resistance underscores the urgent need for new antibiotics with unique mechanisms. Type I signal peptidase (SPase I) is crucial for bacterial survival and a promising target for antibiotics. Herein we designed and synthesized innovative tetrahydroacridine-9-carboxylic acid derivatives by optimizing the initial hit compound SP11 based on virtual screening. Structure-activity relationship (SAR) studies and bioactivity assessments identified compound C09 as a standout, showing excellent in vitro antimicrobial activity against MRSA and other multidrug-resistant Gram-positive pathogens. C09 targets SPase I with a favorable affinity, disrupts bacterial cell membranes, and eradicates biofilms, reducing resistance risk. In vivo tests in a murine MRSA skin infection model demonstrated significant efficacy. Additionally, C09 has good liver microsome metabolic stability, safe hemolytic activity and mammalian cytotoxicity, as well as a good in vivo safety profile. Overall, our findings highlight the potential of tetrahydroacridine-9-carboxylic acid derivatives as a novel class of antibiotics against multidrug-resistant Gram-positive bacteria.
Collapse
Affiliation(s)
- Xiaolin Lu
- School of Science, China Pharmaceutical University, Nanjing, 211198, China; School of Pharmacy, Shanxi Medical University, Taiyuan, 030001, China
| | - Xianghan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Risk Assessment Center of Veterinary Drug Residue and Antimicrobial Resistance, Center for Veterinary Drug Research and Evaluation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Sanya Institute of Nanjing Agricultural University, Nanjing Agricultural University, Sanya, 572025, China
| | - Yushi Ding
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin Gong
- School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Liqin Ming
- School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Xingyang Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Risk Assessment Center of Veterinary Drug Residue and Antimicrobial Resistance, Center for Veterinary Drug Research and Evaluation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Congying Gu
- School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiayi Wang
- School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiaqi Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Risk Assessment Center of Veterinary Drug Residue and Antimicrobial Resistance, Center for Veterinary Drug Research and Evaluation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Sanya Institute of Nanjing Agricultural University, Nanjing Agricultural University, Sanya, 572025, China
| | - Mengkang Gao
- School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Hao Yin
- School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhi Wang
- School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoming Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Risk Assessment Center of Veterinary Drug Residue and Antimicrobial Resistance, Center for Veterinary Drug Research and Evaluation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Liping Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Risk Assessment Center of Veterinary Drug Residue and Antimicrobial Resistance, Center for Veterinary Drug Research and Evaluation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Dayong Zhang
- School of Science, China Pharmaceutical University, Nanjing, 211198, China.
| | - Menghan Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Jinhu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Risk Assessment Center of Veterinary Drug Residue and Antimicrobial Resistance, Center for Veterinary Drug Research and Evaluation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Sanya Institute of Nanjing Agricultural University, Nanjing Agricultural University, Sanya, 572025, China.
| |
Collapse
|
17
|
Davis M, Schermuly A, Rajkhowa A, Thursky K, Warren N, Flowers P. Antibiotic Economies: The Economisation of Antibiotic Use in Australia and Implications for the Mitigation of Antimicrobial Resistance. SOCIOLOGY OF HEALTH & ILLNESS 2025; 47:e70011. [PMID: 39902639 PMCID: PMC11791877 DOI: 10.1111/1467-9566.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 11/07/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025]
Abstract
This paper examines how economic rationalities shape antibiotic usage with the aim of expanding options for the reduction of antimicrobial resistance (AMR). Antibiotic usage is typically attributed to the individual behaviours of patients, pet owners and prescribers, an emphasis that has neglected sociological explanations, particularly the economic rationalities that are transforming healthcare. We used sociological theory of pharmaceutical capitalisation and economisation to explore in-depth interviews on antibiotic usage with scientists, policymakers, prescribers, patients and pet owners in Australia. Antibiotics attracted values in terms of cost to the patient and pet owner, profit for the clinic, how the drugs saved time away from work and childcare, and how they eased the pressures of self-care, parenting and pet ownership. Economic transactions that are only partially under individual patient and prescriber control shape antibiotic use. In these circumstances, antibiotic use is influenced by other social agents-for example, business managers and clinic owners-decentring prescriber authority. Adoption of socio-economic values of antibiotic usage and inclusion of its other economic agents is needed to improve AMR intervention effectiveness.
Collapse
Affiliation(s)
- M. Davis
- School of Social Sciences and Centre to Impact AMRMonash UniversityClaytonAustralia
| | - A. Schermuly
- School of Social Sciences and Centre to Impact AMRMonash UniversityClaytonAustralia
| | - A. Rajkhowa
- Department of Infectious DiseasesNational Centre for Antimicrobial StewardshipMelbourne Medical SchoolUniversity of MelbourneParkvilleAustralia
| | - K. Thursky
- Department of Infectious DiseasesNational Centre for Antimicrobial StewardshipMelbourne Medical SchoolUniversity of MelbourneParkvilleAustralia
| | - N. Warren
- School of Social Sciences and Centre to Impact AMRMonash UniversityClaytonAustralia
| | - P. Flowers
- Department of PsychologyUniversity of StrathclydeGlasgowUK
| |
Collapse
|
18
|
Saleh M, El-Moghazy A, Elgohary AH, Saber WIA, Helmy YA. Revolutionizing Nanovaccines: A New Era of Immunization. Vaccines (Basel) 2025; 13:126. [PMID: 40006673 PMCID: PMC11860605 DOI: 10.3390/vaccines13020126] [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: 11/16/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Infectious diseases continue to pose a significant global health threat. To combat these challenges, innovative vaccine technologies are urgently needed. Nanoparticles (NPs) have unique properties and have emerged as a promising platform for developing next-generation vaccines. Nanoparticles are revolutionizing the field of vaccine development, offering a new era of immunization. They allow the creation of more effective, stable, and easily deliverable vaccines. Various types of NPs, including lipid, polymeric, metal, and virus-like particles, can be employed to encapsulate and deliver vaccine components, such as mRNA or protein antigens. These NPs protect antigens from degradation, target them to specific immune cells, and enhance antigen presentation, leading to robust and durable immune responses. Additionally, NPs can simultaneously deliver multiple vaccine components, including antigens, and adjuvants, in a single formulation, simplifying vaccine production and administration. Nanovaccines offer a promising approach to combat food- and water-borne bacterial diseases, surpassing traditional formulations. Further research is needed to address the global burden of these infections. This review highlights the potential of NPs to revolutionize vaccine platforms. We explore their mechanisms of action, current applications, and emerging trends. The review discusses the limitations of nanovaccines, innovative solutions and the potential role of artificial intelligence in developing more effective and accessible nanovaccines to combat infectious diseases.
Collapse
Affiliation(s)
- Mohammed Saleh
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Ahmed El-Moghazy
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA 92521, USA
| | - Adel H. Elgohary
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - WesamEldin I. A. Saber
- Microbial Activity Unit, Department of Microbiology, Soils, Water and Environment Research Institute, Agricultural Research Center, Giza 12619, Egypt
| | - Yosra A. Helmy
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40546, USA
| |
Collapse
|
19
|
Koshani R, Yeh SL, He Z, Narasimhalu N, Vom Steeg LG, Sim DG, Woods RJ, Read AF, Sheikhi A. Polymeric Anti-Antibiotic Microparticles to Prevent Antibiotic Resistance Evolution. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2407549. [PMID: 39828608 DOI: 10.1002/smll.202407549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/23/2024] [Indexed: 01/22/2025]
Abstract
Vancomycin (VAN) and daptomycin (DAP) are among the last-resort antibiotics for treating multidrug-resistant Gram-positive bacterial infections. They are administered intravenously (IV); however, ≈5 - 10% of the total IV dose is released in the gastrointestinal (GI) tract via biliary excretion, driving resistance emergence in commensal Enterococcus faecium (E. faecium) populations. Here, it is reported that sevelamer (SEV), a Food and Drug Administration (FDA)-approved anion-exchange polymeric microparticle, captures anionic DAP within minutes and cationic VAN within hours, inactivating the antibacterial efficacy of DAP and VAN. In vitro SEV-mediated VAN or DAP transient removal is successfully described by a diffusion-adsorption mechanism. In vivo oral SEV treatment effectively prevented VAN resistance enrichment following the VAN treatment of E. faecium-colonized mice. This work shows, for the first time, that the adjuvant SEV therapy prevents antimicrobial resistance in nosocomial pathogens by eliminating off-target antibiotics. It is envisioned that SEV may protect DAP and VAN from resistance development, potentially addressing the long-lasting antimicrobial resistance.
Collapse
Affiliation(s)
- Roya Koshani
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Shang-Lin Yeh
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Zeming He
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Naveen Narasimhalu
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Landon G Vom Steeg
- Department of Biology and Entomology, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA
| | - Derek G Sim
- Department of Biology and Entomology, The Pennsylvania State University, University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Robert J Woods
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrew F Read
- Department of Biology and Entomology, The Pennsylvania State University, University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Amir Sheikhi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Chemistry, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Neurosurgery, College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| |
Collapse
|
20
|
Wen Y, Huang Y, Xia Z, Wu M, Zhi Y. Zosurabalpin like petrichor: a novel antibiotic class with unprecedented target towards Acinetobacter baumannii. Sci Bull (Beijing) 2025; 70:7-9. [PMID: 39147649 DOI: 10.1016/j.scib.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Affiliation(s)
- Yajin Wen
- The Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yongye Huang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Zhenwei Xia
- The Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Min Wu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| | - Yinghao Zhi
- Wenzhou Traditional Chinese Medicine Hospital of Zhejiang Chinese Medical University, Wenzhou 325000, China.
| |
Collapse
|
21
|
Yang F, Ge Y, Zhang Y, Cui Z, Lin S, Ni W, Sun Z, Shen D, Zhu J, Liu L, Zhao S, Huang N, Sun F, Lu Y, Shi S, Li J. NIR-Activated Hydrogel with Dual-Enhanced Antibiotic Effectiveness for Thorough Elimination of Antibiotic-Resistant Bacteria. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2952-2965. [PMID: 39760335 DOI: 10.1021/acsami.4c16291] [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: 01/07/2025]
Abstract
Antibiotic resistance has become a critical health crisis globally. Traditional strategies using antibiotics can lead to drug-resistance, while inorganic antimicrobial agents can cause severe systemic toxicity. Here, we have developed a dual-antibiotic hydrogel delivery system (PDA-Ag@Levo/CMCS), which can achieve controlled release of clinical antibiotics levofloxacin (Levo) and classic nanoscale antibiotic silver nanoparticles (AgNPs), effectively eliminating drug-resistant P. aeruginosa. Benefiting from the photothermal (PTT) effect of polydopamine (PDA), the local high temperature generated by PDA-Ag@Levo/CMCS can quickly kill bacteria through continuous and responsive release of dual-antibiotics to restore sensitivity to ineffective antibiotics. Moreover, AgNPs could significantly improve the efficiency of traditional antibiotics by disrupting bacterial membranes and reducing their toxicity to healthy tissues. A clever combination of PTT and drug-combination therapy can effectively eliminate biofilms and drug-resistant bacteria. Mechanism studies have shown that PDA-Ag@Levo might eliminate drug-resistant P. aeruginosa by disrupting biofilm formation and protein synthesis, and inhibit the resistance mutation of P. aeruginosa by promoting the expression of related genes, such as rpoS, dinB, and mutS. Collectively, the synergistic effect of this dual-antibiotic hydrogel combined with PTT provides a creative strategy for eliminating drug-resistant bacteria in chronic infection wounds.
Collapse
Affiliation(s)
- Fengjiao Yang
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
- Department of Central Laboratory, Clinical Medicine Scientific and Technical Innovation Park, Shanghai Tenth People's Hospital, Shanghai 200435, China
| | - Yuqi Ge
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
| | - Yue Zhang
- Department of Central Laboratory, Clinical Medicine Scientific and Technical Innovation Park, Shanghai Tenth People's Hospital, Shanghai 200435, China
| | - Zhongqi Cui
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
| | - Shiyang Lin
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
| | - Wenxuan Ni
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
| | - Zijiu Sun
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
| | - Dandan Shen
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
| | - Jichao Zhu
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou 313000, China
- Affiliated Central Hospital of Huzhou University, Huzhou Central Hospital, Huzhou 313000, China
| | - Li Liu
- Department of Clinical Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
- Department of Central Laboratory, Clinical Medicine Scientific and Technical Innovation Park, Shanghai Tenth People's Hospital, Shanghai 200435, China
| | - Shasha Zhao
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
- Department of Central Laboratory, Clinical Medicine Scientific and Technical Innovation Park, Shanghai Tenth People's Hospital, Shanghai 200435, China
| | - Nan Huang
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
- Department of Central Laboratory, Clinical Medicine Scientific and Technical Innovation Park, Shanghai Tenth People's Hospital, Shanghai 200435, China
| | - Fenyong Sun
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
| | - Yingying Lu
- Department of Clinical Laboratory, Shanghai Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Shuo Shi
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
| | - Jinghua Li
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, School of Chemical Science and Engineering, Tongji University, Shanghai 200072, China
| |
Collapse
|
22
|
Mendoza-Guido B, Rivera-Montero L, Barrantes K, Chacon L. Plasmid and integron-associated antibiotic resistance in Escherichia coli isolated from domestic wastewater treatment plants. FEMS Microbiol Lett 2025; 372:fnaf041. [PMID: 40246693 DOI: 10.1093/femsle/fnaf041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/17/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025] Open
Abstract
The rapid dissemination of antibiotic resistance genes (ARGs) represents a significant global threat, with wastewater treatment plants (WWTPs) playing an important role as reservoirs and propagation hubs. In this study, we performed whole-genome sequencing and bioinformatic analyses on eight multidrug-resistant Escherichia coli isolates previously obtained from domestic WWTPs in Costa Rica. We identified 61 ARGs (23 unique), with 40 located on plasmids, and 21 on chromosomal sequences, seven of which were within integrons. Several ARGs were associated with resistance to clinically and veterinary important antibiotics, including sulfamethoxazole/trimethoprim, beta-lactams, and tetracyclines. One hundred twenty-one virulence-associated genes (29 unique) were detected, with 16 located on plasmids. Notably, the presence of virulence factors such as ompT and hlyF genes alongside ARGs on plasmids underscores the transmissible pathogenic potential of WWTP-associated E. coli strains. These findings highlight the role of small domestic WWTPs in disseminating pathogenic and multidrug-resistant bacteria and their mobile genetic elements, emphasizing the need for further research to understand how these discharges impact aquatic environments.
Collapse
Affiliation(s)
- Bradd Mendoza-Guido
- Instituto de Investigaciones en Salud (INISA), Universidad de Costa Rica, San José 11501, Costa Rica
- Programa de Doctorado en Ciencias, Universidad de Costa Rica, San José 11501, Costa Rica
| | - Luis Rivera-Montero
- Instituto de Investigaciones en Salud (INISA), Universidad de Costa Rica, San José 11501, Costa Rica
| | - Kenia Barrantes
- Instituto de Investigaciones en Salud (INISA), Universidad de Costa Rica, San José 11501, Costa Rica
- Doctorado en Ciencias Naturales para el Desarrollo (DOCINADE), Instituto Tecnológico de Costa Rica, Universidad Nacional and Universidad Estatal a Distancia, San José 474-2050, Costa Rica
| | - Luz Chacon
- Instituto de Investigaciones en Salud (INISA), Universidad de Costa Rica, San José 11501, Costa Rica
| |
Collapse
|
23
|
Castagliuolo G, Di Napoli M, Zangmo T, Szpunar J, Ronga L, Zanfardino A, Varcamonti M, Tesauro D. Antimicrobial Activity and Mode of Action of N-Heterocyclic Carbene Silver(I) Complexes. Molecules 2024; 30:76. [PMID: 39795133 PMCID: PMC11722542 DOI: 10.3390/molecules30010076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
Silver drugs have played a vital role in human healthcare for the treatment of infections for many centuries. Currently, due to antibiotic resistance, a potential scenario or the application of silver complexes may arise as substitutes for conventional antibiotics. In this perspective, N-heterocyclic carbene (NHC) ligands have been selected as carrier molecules for silver ions. In this study, we selected two mono NHC-silver halide complexes: bromo[1,3-diethyl-4,5-bis(4-methoxyphenyl)imidazol-2-ylidene]silver(I) (Ag4MC) and chloro[2-pyridin- N-(2-ethylacetylamido)-2-yl-2H-imidazol-2-ylidene]silver(I) (Ag5MC), and two cationic bis NHC silver complexes: bis[1,3-diethyl-4,5-bis(4-methoxyphenyl)imidazol-2-ylidene]silver(I) (Ag4BC) and bis[2-pyridin-N-(2-ethylacetylamido)-2-yl-2H-imidazol-2-ylidene]silver(I) (Ag5BC). The inhibitory properties of the four complexes were evaluated for their antimicrobial potential against a set of Gram (+) and Gram (-) bacterial strains and the fungus C. albicans. In addition, further investigations were made using fluorescence and scanning electron microscopy (SEM) in order to gain more insights into the mechanism of action. Some preliminary information on the Ag target was obtained by analyzing the cytosol of E. coli treated with Ag5MC by size-exclusion chromatography (SEC) coupled with inductively coupled plasma mass spectrometry (ICP-MS).
Collapse
Affiliation(s)
- Giusy Castagliuolo
- Department of Biology, University of Naples “Federico II”, Via Cynthia 26, 80126 Naples, Italy; (G.C.); (M.D.N.); (M.V.)
| | - Michela Di Napoli
- Department of Biology, University of Naples “Federico II”, Via Cynthia 26, 80126 Naples, Italy; (G.C.); (M.D.N.); (M.V.)
| | - Tshering Zangmo
- IPREM (Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux), CNRS, E2S UPPA, Université de Pau et des Pays de l’Adour, CEDEX 9, 64012 Pau, France; (T.Z.); (J.S.); (L.R.)
| | - Joanna Szpunar
- IPREM (Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux), CNRS, E2S UPPA, Université de Pau et des Pays de l’Adour, CEDEX 9, 64012 Pau, France; (T.Z.); (J.S.); (L.R.)
| | - Luisa Ronga
- IPREM (Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux), CNRS, E2S UPPA, Université de Pau et des Pays de l’Adour, CEDEX 9, 64012 Pau, France; (T.Z.); (J.S.); (L.R.)
| | - Anna Zanfardino
- Department of Biology, University of Naples “Federico II”, Via Cynthia 26, 80126 Naples, Italy; (G.C.); (M.D.N.); (M.V.)
| | - Mario Varcamonti
- Department of Biology, University of Naples “Federico II”, Via Cynthia 26, 80126 Naples, Italy; (G.C.); (M.D.N.); (M.V.)
| | - Diego Tesauro
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB), University of Naples “Federico II”, Via Montesano 49, 80131 Naples, Italy;
| |
Collapse
|
24
|
Abdullah SJ, Guan JS, Mu Y, Bhattacharjya S. Single Disulfide Bond in Host Defense Thanatin Analog Peptides: Antimicrobial Activity, Atomic-Resolution Structures and Target Interactions. Int J Mol Sci 2024; 26:51. [PMID: 39795909 PMCID: PMC11720011 DOI: 10.3390/ijms26010051] [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: 11/25/2024] [Revised: 12/21/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Host defense antimicrobial peptides (AMPs) are promising lead molecules with which to develop antibiotics against drug-resistant bacterial pathogens. Thanatin, an inducible antimicrobial peptide involved in the host defense of Podisus maculiventris insects, is gaining considerable attention in the generation of novel classes of antibiotics. Thanatin or thanatin-based analog peptides are extremely potent in killing bacterial pathogens in the Enterobacteriaceae family, including drug-resistant strains of Escherichia coli and Klebsiella pneumoniae. A single disulfide bond that covalently links two anti-parallel β-strands in thanatin could be pivotal to its selective antibacterial activity and mode of action. However, potential correlations of the disulfide covalent bond with structure, activity and target binding in thanatin peptides are currently unclear to. Here, we examined a 16-residue designed thanatin peptide, namely disulfide-bonded VF16QK, and its Cys to Ser substituted variant, VF16QKSer, to delineate their structure-activity relationships. Bacterial growth inhibitory activity was only detected for the disulfide-bonded VF16QK peptide. Mechanistically, both peptides vastly differ in their bacterial cell permeabilizations, atomic-resolution structures, interactions with the LPS-outer membrane and target periplasmic protein LptAm binding. In particular, analysis of the 3-D structures of the two peptides revealed an altered folded conformation for the VF16QKSer peptide that was correlated with diminished LPS-outer membrane permeabilization and target interactions. Analysis of docked complexes of LPS-thanatin peptides indicated potential structural requirements and conformational adaptation for antimicrobial activity. Collectively, these observations contrast with those for the disulfide-bonded β-hairpin antimicrobial protegrin and tachyplesin peptides, where disulfide bonds are dispensable for activity. We surmise that the atomistic structures and associated molecular interactions presented in this work can be utilized to design novel thanatin-based antibiotics.
Collapse
Affiliation(s)
| | | | | | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| |
Collapse
|
25
|
Panáček D, Belza J, Hochvaldová L, Baďura Z, Zoppellaro G, Šrejber M, Malina T, Šedajová V, Paloncýová M, Langer R, Zdražil L, Zeng J, Li L, Zhao E, Chen Z, Xiong Z, Li R, Panáček A, Večeřová R, Kučová P, Kolář M, Otyepka M, Bakandritsos A, Zbořil R. Single Atom Engineered Antibiotics Overcome Bacterial Resistance. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410652. [PMID: 39308225 PMCID: PMC11635910 DOI: 10.1002/adma.202410652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/10/2024] [Indexed: 12/13/2024]
Abstract
The outbreak of antibiotic-resistant bacteria, or "superbugs", poses a global public health hazard due to their resilience against the most effective last-line antibiotics. Identifying potent antibacterial agents capable of evading bacterial resistance mechanisms represents the ultimate defense strategy. This study shows that -the otherwise essential micronutrient- manganese turns into a broad-spectrum potent antibiotic when coordinated with a carboxylated nitrogen-doped graphene. This antibiotic material (termed NGA-Mn) not only inhibits the growth of a wide spectrum of multidrug-resistant bacteria but also heals wounds infected by bacteria in vivo and, most importantly, effectively evades bacterial resistance development. NGA-Mn exhibits up to 25-fold higher cytocompatibility to human cells than its minimum bacterial inhibitory concentration, demonstrating its potential as a next-generation antibacterial agent. Experimental findings suggest that NGA-Mn acts on the outer side of the bacterial cell membrane via a multimolecular collective binding, blocking vital functions in both Gram-positive and Gram-negative bacteria. The results underscore the potential of single-atom engineering toward potent antibiotics, offering simultaneously a long-sought solution for evading drug resistance development while being cytocompatible to human cells.
Collapse
Affiliation(s)
- David Panáček
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
- Nanotechnology CentreCentre for Energy and Environmental TechnologiesVŠB–Technical University of Ostrava17. listopadu 2172/15Ostrava‐Poruba708 00Czech Republic
| | - Jan Belza
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
| | - Lucie Hochvaldová
- Department of Physical ChemistryFaculty of SciencePalacký University Olomouc17. listopadu 1192/12Olomouc771 46Czech Republic
| | - Zdeněk Baďura
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
- Nanotechnology CentreCentre for Energy and Environmental TechnologiesVŠB–Technical University of Ostrava17. listopadu 2172/15Ostrava‐Poruba708 00Czech Republic
| | - Giorgio Zoppellaro
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
- Nanotechnology CentreCentre for Energy and Environmental TechnologiesVŠB–Technical University of Ostrava17. listopadu 2172/15Ostrava‐Poruba708 00Czech Republic
| | - Martin Šrejber
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
| | - Tomáš Malina
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
- Nanotechnology CentreCentre for Energy and Environmental TechnologiesVŠB–Technical University of Ostrava17. listopadu 2172/15Ostrava‐Poruba708 00Czech Republic
| | - Veronika Šedajová
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
| | - Markéta Paloncýová
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
| | - Rostislav Langer
- IT4InnovationsVŠB‐Technical University of Ostrava17. listopadu 2172/15Ostrava‐Poruba708 00Czech Republic
| | - Lukáš Zdražil
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
- Nanotechnology CentreCentre for Energy and Environmental TechnologiesVŠB–Technical University of Ostrava17. listopadu 2172/15Ostrava‐Poruba708 00Czech Republic
| | - Jianrong Zeng
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research InstituteChinese Academy of SciencesShanghai201204P. R. China
| | - Lina Li
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research InstituteChinese Academy of SciencesShanghai201204P. R. China
| | - En Zhao
- Jiangsu Co‐Innovation Center of Efficient Processing and Utilization of Forest ResourcesInternational Innovation Center for Forest Chemicals and MaterialsCollege of Chemical EngineeringNanjing Forestry UniversityLongpan Road 159Nanjing210037P. R. China
| | - Zupeng Chen
- Jiangsu Co‐Innovation Center of Efficient Processing and Utilization of Forest ResourcesInternational Innovation Center for Forest Chemicals and MaterialsCollege of Chemical EngineeringNanjing Forestry UniversityLongpan Road 159Nanjing210037P. R. China
| | - Zhiqiang Xiong
- State Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Ruibin Li
- State Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou Medical CollegeSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Aleš Panáček
- Department of Physical ChemistryFaculty of SciencePalacký University Olomouc17. listopadu 1192/12Olomouc771 46Czech Republic
| | - Renata Večeřová
- Department of MicrobiologyFaculty of Medicine and DentistryPalacký University OlomoucHněvotínská 3Olomouc779 00Czech Republic
| | - Pavla Kučová
- Department of MicrobiologyFaculty of Medicine and DentistryPalacký University OlomoucHněvotínská 3Olomouc779 00Czech Republic
| | - Milan Kolář
- Department of MicrobiologyFaculty of Medicine and DentistryPalacký University OlomoucHněvotínská 3Olomouc779 00Czech Republic
| | - Michal Otyepka
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
- IT4InnovationsVŠB‐Technical University of Ostrava17. listopadu 2172/15Ostrava‐Poruba708 00Czech Republic
| | - Aristides Bakandritsos
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
- Nanotechnology CentreCentre for Energy and Environmental TechnologiesVŠB–Technical University of Ostrava17. listopadu 2172/15Ostrava‐Poruba708 00Czech Republic
| | - Radek Zbořil
- Regional Centre of Advanced Technologies and MaterialsCzech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucŠlechtitelů 241/27Olomouc‐Holice783 71Czech Republic
- Nanotechnology CentreCentre for Energy and Environmental TechnologiesVŠB–Technical University of Ostrava17. listopadu 2172/15Ostrava‐Poruba708 00Czech Republic
| |
Collapse
|
26
|
Abdinia FS, Javadi K, Rajabnia M, Ferdosi-Shahandashti E. A Comprehensive Study on the Distribution of Integrons and Their Gene Cassettes in Clinical Isolates. DNA Cell Biol 2024; 43:579-595. [PMID: 39419631 DOI: 10.1089/dna.2024.0175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Antibiotic resistance is a significant global health concern, leading to increased morbidity, mortality, and health care costs. Integrons are genetic elements that could acquire and express gene cassettes, including those that confer antibiotic resistance. This comprehensive study focused on the distribution of integrons and their gene cassettes in clinical isolates. This study explored the structure and classification of integrons with particular emphasis on Class I, II, III, and IV integrons. It also discussed the role of integrons in antibiotic resistance. The findings of this study contribute to a better understanding of the mechanisms underlying antibiotic resistance and provide valuable insights for developing strategies to combat this public health crisis.
Collapse
Affiliation(s)
- Fatemeh Sarina Abdinia
- Department of Nanotechnology, Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Kasra Javadi
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Mehdi Rajabnia
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Elaheh Ferdosi-Shahandashti
- Biomedical and Microbial Advanced Technologies Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
27
|
Prakash H, Chahal S, Sindhu J, Tyagi P, Sharma D, Guin M, Srivastava N, Singh K. Diastereomeric pure pyrazolyl-indolyl dihydrofurans: Unveiling isomeric selectivity in antibacterial action via in vitro and in silico insights. Bioorg Med Chem Lett 2024; 114:130005. [PMID: 39454968 DOI: 10.1016/j.bmcl.2024.130005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/19/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Developing pure diastereoisomeric molecular hybrids for the selective inhibition of bacterial growth opened new avenues for combating the ever-increasing microbial resistance. Considering this, a series of diastereoisomeric pure pyrazolyl-dihydrofurans (7a-7y) were synthesized and characterized using NMR, LCMS, and X-ray crystallography. DFT based method was used to explore the configurational stability of cis over trans isomeric form. Considering 7a and 8a as representative isomeric forms with same structural framework, the difference in their bio-efficacy against bacterial and fungal strains was assessed using serial dilution method. The relatively high inhibition of bacterial growth by the cis isomeric form (7a) (MIC = 1.562 µg/mL), amoxicillin (MIC = 3.125 µg/mL) inspired us to broaden the substrate scope for synthesizing a series of pure diastereoisomeric cis forms as selective anti-bacterial agents. However, both the isomers displayed antifungal activity less than the standard drug (Fluconazole) employed in the study. All the reactions proceeded smoothly and yielded a diverse array of dihydrofuran derivatives. The developed synthetics were found to be non-cytotoxic against mouse fibroblast cells and didn't affect the seed germination of Brassica nigra seeds when treated at 1 mg/mL concentration. The experimentally determined in vitro results were further validated using in silico molecular docking and dynamics studies.
Collapse
Affiliation(s)
- Hari Prakash
- Jubilant Biosys Ltd., Knowledge Park-II, Greater Noida 201310, Uttar Pradesh, India; Department of Chemistry, Sharda School of Basic Sciences & Research, Sharda University, Knowledge Park-III, Greater Noida 201310, Uttar Pradesh, India
| | - Sandhya Chahal
- Department of Chemistry, Chaudhary Ranbir Singh University, Jind, Haryana 126102, India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, Chaudhary Charan Singh Haryana Agricultural University, Hisar 125004, India
| | - Prateek Tyagi
- Department of Chemistry, Zakir Husain Delhi College, New Delhi, Delhi 110002, India
| | - Deepansh Sharma
- Department of Life Sciences, J.C. Bose University, Science and Technology, YMCA, Faridabad 126001, India
| | - Mridula Guin
- Department of Chemistry, Sharda School of Basic Sciences & Research, Sharda University, Knowledge Park-III, Greater Noida 201310, Uttar Pradesh, India
| | - Noopur Srivastava
- Department of Chemistry, Sharda School of Basic Sciences & Research, Sharda University, Knowledge Park-III, Greater Noida 201310, Uttar Pradesh, India.
| | - Kuldeep Singh
- Jubilant Biosys Ltd., Knowledge Park-II, Greater Noida 201310, Uttar Pradesh, India.
| |
Collapse
|
28
|
Allel K, Peters A, Haghparast-Bidgoli H, Spencer-Sandino M, Conejeros J, Garcia P, Pouwels KB, Yakob L, Munita JM, Undurraga EA. Excess burden of antibiotic-resistant bloodstream infections: evidence from a multicentre retrospective cohort study in Chile, 2018-2022. LANCET REGIONAL HEALTH. AMERICAS 2024; 40:100943. [PMID: 39605961 PMCID: PMC11600772 DOI: 10.1016/j.lana.2024.100943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 10/13/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024]
Abstract
Background Antibiotic-resistant bloodstream infections (ARB BSI) cause an enormous disease and economic burden. We assessed the impact of ARB BSI caused by high- and critical-priority pathogens in hospitalised Chilean patients compared to BSI caused by susceptible bacteria. Methods We conducted a retrospective cohort study from 2018 to 2022 in three Chilean hospitals and measured the association of ARB BSI with in-hospital mortality, length of hospitalisation (LOS), and intensive care unit (ICU) admission. We focused on BSI caused by Acinetobacter baumannii, Enterobacterales, Staphylococcus aureus, Enterococcus species, and Pseudomonas aeruginosa. We addressed confounding using propensity scores, inverse probability weighting, and multivariate regressions. We stratified by community- and hospital-acquired BSI and assessed total hospital and productivity costs. Findings We studied 1218 adult patients experiencing 1349 BSI episodes, with 47.3% attributed to ARB. Predominant pathogens were Staphylococcus aureus (33% Methicillin-resistant 'MRSA'), Enterobacterales (50% Carbapenem-resistant 'CRE'), and Pseudomonas aeruginosa (65% Carbapenem-resistant 'CRPA'). Approximately 80% of BSI were hospital-acquired. ARB was associated with extended LOS (incidence risk ratio IRR = 1.14, 95% CI = 1.05-1.24), increased ICU admissions (odds ratio OR = 1.25; 1.07-1.46), and higher mortality (OR = 1.42, 1.20-1.68) following index blood culture across all BSI episodes. In-hospital mortality risk, adjusted for time-varying and fixed confounders, was 1.35-fold higher (1.16-1.58) for ARB patients, with higher hazard ratios for hospital-acquired MRSA and CRE at 1.37 and 1.48, respectively. Using a societal perspective and a 5% discount rate, we estimated excess costs for ARB at $12,600 per patient, with an estimated annual excess burden of 2270 disability-adjusted life years (DALYs) and $9.6 (5.0-16.4) million. Interpretation It is urgent to develop and implement interventions to reduce the burden of ARB BSIs, particularly from MRSA and CRE. Funding Agencia Nacional de Investigación y Desarrollo ANID, Chile.
Collapse
Affiliation(s)
- Kasim Allel
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Institute for Global Health, University College London, London, UK
- Department of Infectious Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Anne Peters
- Multidisciplinary Initiative for Collaborative Research on Bacterial Resistance (MICROB-R), Santiago, Chile
- Genomics and Resistant Microbes (GeRM), Facultad de Medicina Clínica Alemana, Instituto de Ciencias e Innovación en Medicina (ICIM), Universidad del Desarrollo, Santiago, Chile
| | | | - Maria Spencer-Sandino
- Multidisciplinary Initiative for Collaborative Research on Bacterial Resistance (MICROB-R), Santiago, Chile
- Genomics and Resistant Microbes (GeRM), Facultad de Medicina Clínica Alemana, Instituto de Ciencias e Innovación en Medicina (ICIM), Universidad del Desarrollo, Santiago, Chile
| | - Jose Conejeros
- Department of Infectious Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Escuela de Gobierno, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia Garcia
- Multidisciplinary Initiative for Collaborative Research on Bacterial Resistance (MICROB-R), Santiago, Chile
- Departamento de Laboratorios Clínicos, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Koen B. Pouwels
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
| | - Laith Yakob
- Disease Control Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Jose M. Munita
- Multidisciplinary Initiative for Collaborative Research on Bacterial Resistance (MICROB-R), Santiago, Chile
- Genomics and Resistant Microbes (GeRM), Facultad de Medicina Clínica Alemana, Instituto de Ciencias e Innovación en Medicina (ICIM), Universidad del Desarrollo, Santiago, Chile
- Hospital Padre Hurtado, Santiago, Chile
| | - Eduardo A. Undurraga
- Multidisciplinary Initiative for Collaborative Research on Bacterial Resistance (MICROB-R), Santiago, Chile
- Escuela de Gobierno, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Investigación para la Gestión Integrada del Riesgo de Desastres (CIGIDEN), Santiago, Chile
| |
Collapse
|
29
|
Wang P, Bai Q, Liu X, Zhao M, Chen L, Hu F, Ye J, Chen X, Wang KN, Liu B, Mao D. Nucleus-Targeting Photosensitizers Enhance Neutrophil Extracellular Traps for Efficient Eradication of Multidrug-Resistant Bacterial Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400304. [PMID: 39529561 DOI: 10.1002/adma.202400304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Neutrophil extracellular traps (NETs) are web-like complexes of DNA and proteins that are extruded by activated neutrophils and play critical roles as major components of the innate immune response against pathogen invasion. However, some microbes have developed strategies to evade NET attacks, leading to impaired immune defenses and persistent infections. In this study, an engineered neutrophil strategy for enhancing the antibacterial activity of NETs is developed. A nucleus-targeting photosensitizer (NCP) with strong reactive oxygen species production and a strong DNA-binding capacity is synthesized. NCP-loaded neutrophils are subsequently constructed via direct incubation of NCP with neutrophils, and the NCP is closely inserted into the nucleus DNA. Upon activation by bacteria-related toxins, NCP-coupled NETs can be released rapidly, actively trapping bacteria and providing a high local concentration of NCP around them. Both in vitro and in vivo results revealed that NCP-coupled NETs can effectively eradicate various multidrug-resistant bacteria and biofilms through photodynamic therapy, overcome bacterial immune evasion, and promote tissue recovery from severe wound infections. This design can significantly strengthen NET function, providing a non-antibiotic alternative platform for treating bacterial infectious diseases.
Collapse
Affiliation(s)
- Peng Wang
- Department of Burns, Wound Repair & Reconstruction, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Qingqing Bai
- Department of Burns, Wound Repair & Reconstruction, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xianglong Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 117585, Singapore
| | - Minyang Zhao
- Department of Burns, Wound Repair & Reconstruction, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Lei Chen
- Department of Burns, Wound Repair & Reconstruction, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Fang Hu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Jinzhou Ye
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Xinhan Chen
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Kang-Nan Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 117585, Singapore
| | - Duo Mao
- Department of Burns, Wound Repair & Reconstruction, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, 510080, China
| |
Collapse
|
30
|
Hussein S, Ahmed SK, Mohammed SM, Qurbani K, Ali S, Saber AF, Khdir K, Shareef S, Rasool AH, Mousa S, Sidiq AS, Hamzah H. Recent developments in antibiotic resistance: an increasing threat to public health. ANNALS OF ANIMAL SCIENCE 2024. [DOI: 10.2478/aoas-2024-0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Antibiotic resistance (ABR) is a major global health threat that puts decades of medical progress at risk. Bacteria develop resistance through various means, including modifying their targets, deactivating drugs, and utilizing efflux pump systems. The main driving forces behind ABR are excessive antibiotic use in healthcare and agriculture, environmental contamination, and gaps in the drug development process. The use of advanced detection technologies, such as next-generation sequencing (NGS), clustered regularly interspaced short palindromic repeats (CRISPR)-based diagnostics, and metagenomics, has greatly improved the identification of resistant pathogens. The consequences of ABR on public health are significant, increased mortality rates, the endangerment of modern medical procedures, and resulting in higher healthcare expenses. It has been expected that ABR could potentially drive up to 24 million individuals into extreme poverty by 2030. Mitigation strategies focus on antibiotic stewardship, regulatory measures, research incentives, and raising public awareness. Furthermore, future research directions involve exploring the potential of CRISPR-Cas9 (CRISPR-associated protein 9), nanotechnology, and big data analytics as new antibiotic solutions. This review explores antibiotic resistance, including mechanisms, recent trends, drivers, and technological advancements in detection. It also evaluates the implications for public health and presents strategies for mitigating resistance. The review emphasizes the significance of future directions and research needs, stressing the necessity for sustained and collaborative efforts to tackle this issue.
Collapse
Affiliation(s)
- Safin Hussein
- Department of Biology, College of Science , University of Raparin , Rania, Sulaymaniyah, Kurdistan Region, 46012 , Iraq
| | - Sirwan Khalid Ahmed
- College of Nursing , University of Raparin , Rania, Sulaymaniyah, Kurdistan Region, 46012 , Iraq
| | - Saman M. Mohammed
- Department of Biology, College of Education , University of Sulaimani , Sulaymaniyah, Kurdistan Region, 46001 , Iraq
| | - Karzan Qurbani
- Department of Biology, College of Science , University of Raparin , Rania, Sulaymaniyah, Kurdistan Region, 46012 , Iraq
| | - Seenaa Ali
- Department of Medical Laboratory, College of Health and Medical Technology , Sulaimani Polytechnic University , Sulaymaniyah, Kurdistan Region, 46001 , Iraq
| | - Abdulmalik Fareeq Saber
- Department of Psychiatric and Mental Health Nursing, College of Nursing , Hawler Medical University , Erbil, Kurdistan Region, 44001 , Iraq
| | - Karokh Khdir
- Department of Biology, College of Education , University of Sulaimani , Sulaymaniyah, Kurdistan Region, 46001 , Iraq
| | - Salar Shareef
- Department of Medical Laboratory Science, College of Science , University of Raparin , Rania, Sulaymaniyah, Kurdistan Region, 46012 , Iraq
| | - Aram H. Rasool
- Department of Medical Laboratory Science, College of Health Sciences , University of Human Development , Sulaymaniyah, Kurdistan Region, 46001 , Iraq
| | - Sumayah Mousa
- Department of Medical Laboratory Science, College of Science , Komar University of Science and Technology , Sulaymaniyah, Kurdistan Region, 46001 , Iraq
| | - Avin S. Sidiq
- Department of Anesthesia, College of Health Sciences , Cihan University Sulaimaniya , Sulaymaniyah, Kurdistan Region, 46001 , Iraq
| | - Haider Hamzah
- Department of Biology, College of Science , University of Sulaimani , Sulaymaniyah, Kurdistan Region, 46001 , Iraq
| |
Collapse
|
31
|
Luo L, Cai Y, Su Y, Li C, Tian G, Wang X, Wu Z, Chen W, Zhang T, Zhang Z. Novel Tree Shrew-Derived Antimicrobial Peptide with Broad-Spectrum Antibacterial Activity. ACS OMEGA 2024; 9:45279-45288. [PMID: 39554445 PMCID: PMC11561621 DOI: 10.1021/acsomega.4c06857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/19/2024]
Abstract
The number of cationic residues and net charge are critical for the activity of antimicrobial peptides (AMPs) due to their role in facilitating initial electrostatic interactions with negatively charged bacterial membranes. A cathelicidin AMP (TC-33) has been identified from the Chinese tree shrew in our previous work, which exhibited weak antimicrobial activity, likely due to its moderately cationic nature. In the current study, based on TC-33, we designed a novel AMP by peptide truncation and Glu substitutions to increase its net cationic charge from +4 to +8. The resulting peptide, TC-LAR-18, showed 4-128-fold enhanced antimicrobial activity relative to TC-33 without causing hemolysis and cytotoxicity within 100 μg/mL. TC-LAR-18 effectively eliminated both planktonic and biofilm-associated bacteria, demonstrating rapid bactericidal effects due to its ability to quickly penetrate and disrupt bacterial cell membranes with a low propensity to induce resistance. Notably, TC-LAR-18 provided substantial protection against skin bacterial infection in mice, underscoring its therapeutic potential. These findings not only highlight the importance of positively charged residues for the antibacterial activity of AMPs but also present a useful drug candidate for combating multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Lin Luo
- Institute
of Medical Biology, Chinese Academy of Medical
Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
- Third
Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Third Affiliated Hospital of Kunming Medical University
(Yunnan Cancer Hospital), Kunming 650118, Yunnan, China
| | - Ying Cai
- Institute
of Medical Biology, Chinese Academy of Medical
Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| | - Yunhan Su
- School
of Basic Medical Sciences, Kunming Medical
University, Kunming 650500, Yunnan, China
| | - Chenxi Li
- Third
Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Third Affiliated Hospital of Kunming Medical University
(Yunnan Cancer Hospital), Kunming 650118, Yunnan, China
| | - Gengzhou Tian
- Department
of Breast Surgery, First Affiliated Hospital
of Kunming Medical University, Kunming 650223, Yunnan, China
| | - Xingyu Wang
- Institute
of Medical Biology, Chinese Academy of Medical
Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
- School
of Life Sciences, Yunnan University, Kunming 650500, China
| | - Zhongxiang Wu
- Institute
of Medical Biology, Chinese Academy of Medical
Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| | - Wenlin Chen
- Third
Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Third Affiliated Hospital of Kunming Medical University
(Yunnan Cancer Hospital), Kunming 650118, Yunnan, China
| | - Tianyu Zhang
- Institute
of Medical Biology, Chinese Academy of Medical
Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| | - Zhiye Zhang
- Institute
of Medical Biology, Chinese Academy of Medical
Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| |
Collapse
|
32
|
Huang Y, Li J, Yu Z, Li J, Liang K, Deng Y. Elaborated Bio-Heterojunction With Robust Sterilization Effect for Infected Tissue Regeneration via Activating Competent Cell-Like Antibacterial Tactic. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2414111. [PMID: 39397342 DOI: 10.1002/adma.202414111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Indexed: 10/15/2024]
Abstract
Phototherapy, such as photothermal therapy (PTT) and photodynamic therapy (PDT) has been a powerful strategy to combat bacterial infection. However, the compact cell membranes of pathogenic bacteria, especially drug-resistant bacteria, significantly diminish the efficiency of heat conduction and impede the entrance of reactive oxygen species (ROS) into cells, resulting in unsatisfactory sterilization. Enlightened by the membrane feature of competent bacteria, herein a MXene/CaO2 bio-heterojunction (MC bio-HJ) is elaborated to achieve rapid disinfection and promote infected tissue regeneration through activating competent cell-like antibacterial tactics. The bio-HJ first compels pathogenic bacteria to become a competent cell-like stage through the coordination of Ca2+ and membrane phospholipid, and potentiates the membrane permeability. Assisted by near infrared (NIR) irradiation, the heat and ROS generated from PTT and PDT of bio-HJ easily pass through bacterial membrane and drastically perturb bacterial metabolism, leading to rapid disinfection. More importantly, employing two in vivo infected model of mice, it have corroborated that the MC bio-HJs not only effectively accelerate MRSA-infected cutaneous regeneration, but also considerably boost osseointegration in an infected bone defect after coating on orthopedic implants. As envisaged, this work demonstrates a novel therapeutic tactic with robust antibacterial effect to remedy infected tissue regeneration through activating competent cell-like stage.
Collapse
Affiliation(s)
- Yixuan Huang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Jialun Li
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
| | - Zhaohan Yu
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
| | - Jiyao Li
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
| | - Kunneng Liang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Department of Cariology and Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610065, China
| | - Yi Deng
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, 999077, China
| |
Collapse
|
33
|
Li M, Wang C, Yu Q, Chen H, Ma Y, Wei L, Wu MX, Yao M, Lu M. A wearable and stretchable dual-wavelength LED device for home care of chronic infected wounds. Nat Commun 2024; 15:9380. [PMID: 39477919 PMCID: PMC11525593 DOI: 10.1038/s41467-024-53579-6] [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: 06/18/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Phototherapy can offer a safe and non-invasive solution against infections, while promoting wound healing. Conventional phototherapeutic devices are bulky and limited to hospital use. To overcome these challenges, we developed a wearable, flexible red and blue LED (r&bLED) patch controlled by a mobile-connected system, enabling safe self-application at home. The patch exhibits excellent skin compatibility, flexibility, and comfort, with high safety under system supervision. Additionally, we synthesized a sprayable fibrin gel (F-gel) containing blue light-sensitive thymoquinone and red light-synergistic NADH. Combined with bLED, thymoquinone eradicated microbes and biofilms within minutes, regardless of antibiotic resistance. Furthermore, NADH and rLED synergistically improved macrophage and endothelial cell mitochondrial function, promoting wound healing, reducing inflammation, and enhancing angiogenesis, as validated in infected diabetic wounds in mice and minipigs. This innovative technology holds great promise for revolutionizing at-home phototherapy for chronic infected wounds.
Collapse
Affiliation(s)
- Ming Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenxi Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qiang Yu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haoyi Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingying Ma
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA.
| | - Min Yao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Min Lu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
34
|
Koncz M, Stirling T, Hadj Mehdi H, Méhi O, Eszenyi B, Asbóth A, Apjok G, Tóth Á, Orosz L, Vásárhelyi BM, Ari E, Daruka L, Polgár TF, Schneider G, Zalokh SA, Számel M, Fekete G, Bohár B, Nagy Varga K, Visnyovszki Á, Székely E, Licker MS, Izmendi O, Costache C, Gajic I, Lukovic B, Molnár S, Szőcs-Gazdi UO, Bozai C, Indreas M, Kristóf K, Van der Henst C, Breine A, Pál C, Papp B, Kintses B. Genomic surveillance as a scalable framework for precision phage therapy against antibiotic-resistant pathogens. Cell 2024; 187:5901-5918.e28. [PMID: 39332413 DOI: 10.1016/j.cell.2024.09.009] [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: 01/17/2024] [Revised: 05/15/2024] [Accepted: 09/04/2024] [Indexed: 09/29/2024]
Abstract
Phage therapy is gaining increasing interest in the fight against critically antibiotic-resistant nosocomial pathogens. However, the narrow host range of bacteriophages hampers the development of broadly effective phage therapeutics and demands precision approaches. Here, we combine large-scale phylogeographic analysis with high-throughput phage typing to guide the development of precision phage cocktails targeting carbapenem-resistant Acinetobacter baumannii, a top-priority pathogen. Our analysis reveals that a few strain types dominate infections in each world region, with their geographical distribution remaining stable within 6 years. As we demonstrate in Eastern Europe, this spatiotemporal distribution enables preemptive preparation of region-specific phage collections that target most local infections. Finally, we showcase the efficacy of phage cocktails against prevalent strain types using in vitro and animal infection models. Ultimately, genomic surveillance identifies patients benefiting from the same phages across geographical scales, thus providing a scalable framework for precision phage therapy.
Collapse
Affiliation(s)
- Mihály Koncz
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary
| | - Tamás Stirling
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary; Doctoral School of Biology, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Hiba Hadj Mehdi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary; Doctoral School of Biology, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Orsolya Méhi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Bálint Eszenyi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - András Asbóth
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary; Department of Genetics, ELTE Eötvös Loránd University, Pázmány Péter stny. 1/C, 1117 Budapest, Hungary
| | - Gábor Apjok
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Ákos Tóth
- National Center for Public Health and Pharmacy, Albert Flórián út 2-6, 1097 Budapest, Hungary
| | - László Orosz
- Department of Medical Microbiology, University of Szeged, Szent-Györgyi Albert Medical School, Dom tér 10, 6720 Szeged, Hungary
| | - Bálint Márk Vásárhelyi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Eszter Ari
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; Department of Genetics, ELTE Eötvös Loránd University, Pázmány Péter stny. 1/C, 1117 Budapest, Hungary; HCEMM-BRC Metabolic Systems Biology Group, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Tamás Ferenc Polgár
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; Theoretical Medicine Doctoral School, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - György Schneider
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Szigeti út 12, 7624 Pécs, Hungary
| | - Sif Aldin Zalokh
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Mónika Számel
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Gergely Fekete
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Metabolic Systems Biology Group, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Balázs Bohár
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, 10th Floor Commonwealth Building Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Karolina Nagy Varga
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Ádám Visnyovszki
- South-Pest Central Hospital National Institute of Hematology and Infectious Diseases, Nagyvárad tér 1, 1097 Budapest, Hungary; Doctoral School of Interdisciplinary Medical Sciences, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Edit Székely
- George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Str. Gheorghe Marinescu 38, 540142 Targu Mures, Romania; County Emergency Clinical Hospital of Targu Mures, Str. Dr. Gh. Marinescu 50, 540136 Targu Mures, Romania
| | - Monica-Sorina Licker
- Microbiology Department, Multidisciplinary Research Center on Antimicrobial Resistance, "Victor Babes" University of Medicine and Pharmacy, Str. Eftimie Murgu 2, 300041 Timisoara, Romania; Microbiology Laboratory, "Pius Branzeu" Emergency Clinical County Hospital, Str. Liviu Rebreanu 156, 300723 Timisoara, Romania
| | - Oana Izmendi
- Microbiology Department, Multidisciplinary Research Center on Antimicrobial Resistance, "Victor Babes" University of Medicine and Pharmacy, Str. Eftimie Murgu 2, 300041 Timisoara, Romania; Microbiology Laboratory, "Pius Branzeu" Emergency Clinical County Hospital, Str. Liviu Rebreanu 156, 300723 Timisoara, Romania; Doctoral School, "Victor Babes" University of Medicine and Pharmacy, Str. Eftimie Murgu 2, 300041 Timisoara, Romania
| | - Carmen Costache
- Department of Microbiology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj-Napoca, Str. Victor Babes 8, 400347 Cluj-Napoca, Romania
| | - Ina Gajic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia
| | - Bojana Lukovic
- Academy of Applied Studies Belgrade, College of Health Sciences, Bulevar Zorana Djindjica 152a, Belgrade, Serbia
| | - Szabolcs Molnár
- Emergency County Hospital Miercurea-Ciuc, Str. Doctor Dénes László 2, 530173 Miercurea Ciuc, Romania
| | | | - Csilla Bozai
- County Emergency Hospital Satu Mare, Str. Ravensburg 1-3, 440192 Satu Mare, Romania
| | - Marina Indreas
- Bacau County Emergency Hospital, Str. Haret Spiru 2-4, 600114 Bacau, Romania
| | - Katalin Kristóf
- Institute of Laboratory Medicine, Semmelweis University, Üllői út 78/b, 1083 Budapest, Hungary
| | - Charles Van der Henst
- Microbial Resistance and Drug Discovery, VIB-VUB Center for Structural Biology, VIB, Flanders Institute for Biotechnology, Pleinlaan 2, Building E-3, 1050 Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Elsene, 1050 Brussels, Belgium
| | - Anke Breine
- Microbial Resistance and Drug Discovery, VIB-VUB Center for Structural Biology, VIB, Flanders Institute for Biotechnology, Pleinlaan 2, Building E-3, 1050 Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Elsene, 1050 Brussels, Belgium
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Metabolic Systems Biology Group, Temesvári Krt. 62, 6726 Szeged, Hungary; National Laboratory for Health Security, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary.
| | - Bálint Kintses
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary.
| |
Collapse
|
35
|
Yan Y, Xu N, Wang X, Shi L, Huang Q, Wang J, Li X, Ni T, Yang Z, Guo W. Mesoporous polydopamine/copper sulfide hybrid nanocomposite for highly efficient NIR-triggered bacterial inactivation. Int J Biol Macromol 2024; 277:134238. [PMID: 39084434 DOI: 10.1016/j.ijbiomac.2024.134238] [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: 04/10/2024] [Revised: 06/08/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Polydopamine has gained considerable attention in the biomaterial domain owing to its excellent biocompatibility, antioxidant activity, photothermal effect and adhesion property. Herein, copper sulfide (Cu2-xS) wrapped in mesoporous polydopamine (MPDA) was synthesized through in-situ polymerization, followed by the surface modification with cationic polyethyleneimine (PEI). The mussel-inspired MPDA matrix successfully prevented the oxidation and agglomeration of Cu2-xS nanoparticles, and regulated the release of copper ions and reactive oxygen species (ROS) levels. Surface-modified PEI endow MPDA@Cu2-xS with positive charges, facilitating their rapid contact with negatively charged bacteria through electrostatic interactions. The pH-dependent Cu+/Cu2+ release and NIR-responsive ROS generation were confirmed using molecular probes and electron spin resonance (ESR). The MPDA@Cu2-xS/PEI showed significantly enhanced antibacterial activity and reduced cytotoxicity for NIH3T3 cells. Under NIR irradiation (1.0 W/cm2, 10 min), germicidal efficiency against Escherichia coli (E. coli) and Staphyloccocus aureus (S. aureus) could reach 100 % and 99.94 %, respectively. The exceptional antibacterial activities of MPDA@Cu2-xS/PEI was mainly attributed to the synergistic photothermal effect, controlled release of copper ions and ROS generation, as well as electrostatic interaction. More importantly, the MPDA@Cu2-xS/PEI composite exhibited excellent biocompatibility and biosafety. Overall, this organic/inorganic hybrid holds great potential as a promising candidate for wound treatment.
Collapse
Affiliation(s)
- Yunhui Yan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China.
| | - Na Xu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Xian Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Li Shi
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Qianqian Huang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Jia Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Xiangrong Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Tianjun Ni
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhijun Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China
| | - Wei Guo
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China; Xinxiang engineering technology research center of functional medicine nanomaterials, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
36
|
Do VH, Nguyen VS, Nguyen SH, Le DQ, Nguyen TT, Nguyen CH, Ho TH, Vo NS, Nguyen T, Nguyen HA, Cao MD. PanKA: Leveraging population pangenome to predict antibiotic resistance. iScience 2024; 27:110623. [PMID: 39228791 PMCID: PMC11369404 DOI: 10.1016/j.isci.2024.110623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/14/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024] Open
Abstract
Machine learning has the potential to be a powerful tool in the fight against antimicrobial resistance (AMR), a critical global health issue. Machine learning can identify resistance mechanisms from DNA sequence data without prior knowledge. The first step in building a machine learning model is a feature extraction from sequencing data. Traditional methods like single nucleotide polymorphism (SNP) calling and k-mer counting yield numerous, often redundant features, complicating prediction and analysis. In this paper, we propose PanKA, a method using the pangenome to extract a concise set of relevant features for predicting AMR. PanKA not only enables fast model training and prediction but also improves accuracy. Applied to the Escherichia coli and Klebsiella pneumoniae bacterial species, our model is more accurate than conventional and state-of-the-art methods in predicting AMR.
Collapse
Affiliation(s)
- Van Hoan Do
- Center for Applied Mathematics and Informatics, Le Quy Don Technical University, Hanoi, Vietnam
| | - Van Sang Nguyen
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Vietnam
| | | | - Duc Quang Le
- Faculty of IT, Hanoi University of Civil Engineering, Hanoi, Vietnam
| | - Tam Thi Nguyen
- Oxford University Clinical Research Unit, Hanoi, Vietnam
| | - Canh Hao Nguyen
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Kyoto, Japan
| | - Tho Huu Ho
- Department of Medical Microbiology, The 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
- Department of Genomics & Cytogenetics, Institute of Biomedicine & Pharmacy, Vietnam Military Medical University, Hanoi, Vietnam
| | - Nam S. Vo
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Vietnam
| | | | | | | |
Collapse
|
37
|
Guo L, Kuipers OP, Broos J. An Engineered Nisin Analogue with a Hydrophobic Moiety Attached at Position 17 Selectively Inhibits Enterococcus faecium Strains. ACS Chem Biol 2024; 19:2023-2031. [PMID: 39254256 PMCID: PMC11420946 DOI: 10.1021/acschembio.4c00337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Antibiotic resistance is one of the most challenging global public health concerns. It results from the misuse and overuse of broad-spectrum antibiotics, which enhance the dissemination of resistance across diverse bacterial species. Antibiotics like nisin and teixobactin do not target an essential protein and employ a dual mode of action antibacterial mechanism, thereby being less prone to induce resistance. There is a need for the development of a potent narrow-spectrum dual-mode-acting antibiotic against human pathogens. Using nisin, a lantibiotic with potent antimicrobial activity against many pathogens, as a template, the unnatural amino acid azidohomoalanine was introduced at selected positions and subsequently modified using click chemistry with 14 alkyne-moiety containing tails. A novel nisin variant, compound 47, featuring a benzyl group-containing tail, exhibited potent activity against various (drug-resistant) E. faecium strains with an MIC value (3.8 mg/L) similar to nisin, whereas its activity toward other pathogens like Staphylococcus aureus and Bacillus cereus was significantly reduced. Like nisin, the mode of action of compound 47 results from the inhibition of cell wall synthesis by binding to lipid II and nisin-lipid II hybrid-pore formation in the outer membrane. The resistance of compound 47 against proteolytic degradation is markedly enhanced compared to nisin. Like nisin, compound 47 was hardly hemolytic even at a very high dose. Collectively, a modified nisin variant is presented with significantly enhanced target organism specificity and stability.
Collapse
Affiliation(s)
- Longcheng Guo
- Department of Molecular Genetics,
Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, 9747 AG, The Netherlands
| | - Oscar P. Kuipers
- Department of Molecular Genetics,
Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, 9747 AG, The Netherlands
| | - Jaap Broos
- Department of Molecular Genetics,
Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, 9747 AG, The Netherlands
| |
Collapse
|
38
|
Alfonso I. Supramolecular chemical biology: designed receptors and dynamic chemical systems. Chem Commun (Camb) 2024; 60:9692-9703. [PMID: 39129537 DOI: 10.1039/d4cc03163f] [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: 08/13/2024]
Abstract
Supramolecular chemistry focuses on the study of species joined by non-covalent interactions, and therefore on dynamic and relatively ill-defined structures. Despite being a well-developed field, it has to face important challenges when dealing with the selective recognition of biomolecules in highly competitive biomimetic media. However, supramolecular interactions reside at the core of chemical biology systems, since many processes in nature are governed by weak, non-covalent, strongly dynamic contacts. Therefore, there is a natural connection between these two research fields, which are not frequently related or share interests. In this feature article, I will highlight our most recent results in the molecular recognition of biologically relevant species, following different conceptual approaches from the most conventional design of elaborated receptors to the less popular dynamic combinatorial chemistry methodology. Selected illustrative examples from other groups will be also included. The discussion has been focused mainly on systems with potential biomedical applications.
Collapse
Affiliation(s)
- Ignacio Alfonso
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC), The Spanish National Research Council (CSIC), Jordi Girona 18-26, 08034, Barcelona, Spain.
| |
Collapse
|
39
|
Yadav P, Fatimah N, Sahoo SC, Kumari S, Berry S, Reenu, Kumar Pinnaka A, Bhalla A. Design, Synthesis and Biological Evaluation of C3‐Indolyl/(3‐chloro‐indolyl)‐ C4‐aryl/heteroaryl‐azetidin‐2‐ones. ChemMedChem 2024; 19. [DOI: 10.1002/cmdc.202400157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Indexed: 01/04/2025]
Abstract
AbstractHerein, trans‐ and cis‐azetidin‐2‐ones 3–6 were strategically synthesized, capitalizing on the bioactivity of azetidin‐2‐ones and indole pharmacophore, followed by a comprehensive characterization using a diverse array of spectroscopic techniques. The sixteen azetidin‐2‐ones were examined for antimicrobial activities against both Gram‐negative (P. aeruginosa, E. coli, A. baumannii) and Gram‐positive bacteria (S. aureus, E. faecium, B. cereus), as well as against C. albicans and C. tropicalis fungal strains. The highly potent compounds (5 a, 6 b, 6 d) demonstrated maximum inhibition against all multidrug‐resistant strains, with minimum inhibitory concentrations ranging from 0.97–3.9 μg/mL, surpassing the potency of standard ampicillin (MIC: 3.12–50 μg/mL). Moreover, 6 b and 6 d exhibited significant inhibitory effects on C. albicans (MIC: 0.97 μg/mL), comparable to fluconazole. The presence of C3‐(3‐chloro‐indolyl) scaffold, combined with diverse electronic effects at N1/C4‐centers, particularly the inclusion of thiophen‐2‐yl motif, greatly influenced the activity of target compounds. Assessment of 4 d, 4 i–k and 6 d on THLE‐2 cell lines revealed their preferential safety. Molecular docking studies revealed seven compounds with active dual targeting of DNA GyrB and PBP2a proteins, demonstrating a potent broad‐spectrum antibacterial effect. In silico ADME analysis affirms positive drug‐likeness and favorable pharmacokinetic characteristics of indole‐derived hybrids, indicating a promising potential for addressing challenges in evolving multidrug resistance.
Collapse
Affiliation(s)
- Pooja Yadav
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University Chandigarh 160014 India
| | - Nasreen Fatimah
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University Chandigarh 160014 India
| | - S. C. Sahoo
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University Chandigarh 160014 India
| | - Sumeeta Kumari
- Microbial Type Culture Collection and Gene Bank CSIR-Microbial Type Culture Collection and Gene Bank Chandigarh 160036 India
| | - Shiwani Berry
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University Chandigarh 160014 India
- Department of Chemistry and Chemical Sciences Central University of Himachal Pradesh Shahpur Kangra 176206 India
| | - Reenu
- Department of Chemistry Govt. Home Science College Chandigarh 160011 India
| | - Anil Kumar Pinnaka
- Microbial Type Culture Collection and Gene Bank CSIR-Microbial Type Culture Collection and Gene Bank Chandigarh 160036 India
| | - Aman Bhalla
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University Chandigarh 160014 India
| |
Collapse
|
40
|
Gonçalves ASC, Leitão MM, Fernandes JR, Saavedra MJ, Pereira C, Simões M, Borges A. Photodynamic activation of phytochemical-antibiotic combinations for combatting Staphylococcus aureus from acute wound infections. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 258:112978. [PMID: 39002192 DOI: 10.1016/j.jphotobiol.2024.112978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024]
Abstract
Staphylococcus aureus is characterized by its high resistance to conventional antibiotics, particularly methicillin-resistant (MRSA) strains, making it a predominant pathogen in acute and chronic wound infections. The persistence of acute S. aureus wound infections poses a threat by increasing the incidence of their chronicity. This study investigated the potential of photodynamic activation using phytochemical-antibiotic combinations to eliminate S. aureus under conditions representative of acute wound infections, aiming to mitigate the risk of chronicity. The strategy applied takes advantage of the promising antibacterial and photosensitising properties of phytochemicals, and their ability to act as antibiotic adjuvants. The antibacterial activity of selected phytochemicals (berberine, curcumin, farnesol, gallic acid, and quercetin; 6.25-1000 μg/mL) and antibiotics (ciprofloxacin, tetracycline, fusidic acid, oxacillin, gentamicin, mupirocin, methicillin, and tobramycin; 0.0625-1024 μg/mL) was screened individually and in combination against two S. aureus clinical strains (methicillin-resistant and -susceptible-MRSA and MSSA). The photodynamic activity of the phytochemicals was assessed using a light-emitting diode (LED) system with blue (420 nm) or UV-A (365 nm) variants, at 30 mW/cm2 (light doses of 9, 18, 27 J/cm2) and 5.5 mW/cm2 (light doses of 1.5, 3.3 and 5.0 J/cm2), respectively. Notably, all phytochemicals restored antibiotic activity, with 9 and 13 combinations exhibiting potentiating effects on MSSA and MRSA, respectively. Photodynamic activation with blue light (420 nm) resulted in an 8- to 80-fold reduction in the bactericidal concentration of berberine against MSSA and MRSA, while curcumin caused 80-fold reduction for both strains at the light dose of 18 J/cm2. Berberine and curcumin-antibiotic combinations when subjected to photodynamic activation (420 nm light, 10 min, 18 J/cm2) reduced S. aureus culturability by ≈9 log CFU/mL. These combinations lowered the bactericidal concentration of antibiotics, achieving a 2048-fold reduction for gentamicin and 512-fold reduction for tobramycin. Overall, the dual approach involving antimicrobial photodynamic inactivation and selected phytochemical-antibiotic combinations demonstrated a synergistic effect, drastically reducing the culturability of S. aureus and restoring the activity of gentamicin and tobramycin.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; Environmental Health Department, Portuguese National Health Institute Doutor Ricardo Jorge, Porto, Portugal
| | - Miguel M Leitão
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007, Porto, Portugal
| | - José R Fernandes
- CQVR-Vila Real Chemistry Center, University of Trás-os-Montes e Alto Douro, Portugal; Physical Department, University of Trás-os-Montes and Alto Douro, Quinta dos Prados, 5000-801 Vila Real, Portugal
| | - Maria José Saavedra
- Antimicrobials, Biocides and Biofilms Unit (AB2Unit), Laboratory of Medical Microbiology, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; Animal and Veterinary Research Center (CECAV)-Al4AnimalS, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; Center Interdisciplinar of Marine and Environmental Research (CIIMAR), University of Porto, 4450-208 Matosinhos, Portugal; Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB)-Inov4Agro, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Cristiana Pereira
- Environmental Health Department, Portuguese National Health Institute Doutor Ricardo Jorge, Porto, Portugal; Environmental Hygiene and Human Biomonitoring Unit, Department of Health Protection, Laboratoire National de Santé, Luxembourg
| | - Manuel Simões
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; DEQ-Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; DEQ-Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal.
| |
Collapse
|
41
|
Jin C, Yang S, Ma H, Zhang X, Zhang K, Zou W. Ubiquitous nanocolloids suppress the conjugative transfer of plasmid-mediated antibiotic resistance in aqueous environment. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 355:124231. [PMID: 38801878 DOI: 10.1016/j.envpol.2024.124231] [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: 03/26/2024] [Revised: 05/12/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Nanocolloids (Nc) are widespread in natural water environment, whereas the potential effects of Nc on dissemination of antibiotic resistance remain largely unknown. In this study, Nc collected from the Yellow River in Henan province was tested for its ability to influence the conjugative transfer of resistant plasmid in aqueous environment. The results revealed that the conjugative transfer of RP4 plasmid between Escherichia coli was down-regulated by 52%-91% upon exposure to 1-10 mg/L Nc and the reduction became constant when the dose became higher (20-200 mg/L). Despite the exposure of Nc activated the anti-oxidation and SOS response in bacteria through up-regulating genes involved in glutathione biosynthesis and DNA recombination, the inhibition on the synthesis and secretion of extracellular polysaccharide induced the prevention of cell-cell contact, leading to the reduction of plasmid transfer. This was evidenced by the decreased bacterial adhesion and lowered levels of genes and metabolites relevant to transmembrane transport and D-glucose phosphorylation, as clarified in phenotypic, transcriptomics and metabolomics analysis of E. coli. The significant down-regulation of glycolysis/gluconeogenesis and TCA cycle was associated with the shortage of ATP induced by Nc. The up-regulation of global regulatory genes (korA and trbA) and the reduction of plasmid genes (trfAp, trbBp, and traG) expression also contributed to the suppressed conjugation of RP4 plasmid. The obtained findings remind that the role of ubiquitous colloidal particles is nonnegligible when practically and comprehensively assessing the risk of antibiotic resistance in the environment.
Collapse
Affiliation(s)
- Caixia Jin
- School of Environment, Key Laboratory of Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory of Environmental Pollution Control, Henan Normal University, Xinxiang, 453007, China
| | - Shuo Yang
- School of Environment, Key Laboratory of Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory of Environmental Pollution Control, Henan Normal University, Xinxiang, 453007, China
| | - Haiwen Ma
- School of Environment, Key Laboratory of Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory of Environmental Pollution Control, Henan Normal University, Xinxiang, 453007, China
| | - Xingli Zhang
- School of Environment, Key Laboratory of Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory of Environmental Pollution Control, Henan Normal University, Xinxiang, 453007, China
| | - Kai Zhang
- School of Geographic Sciences, Henan Key Laboratory for Synergistic Prevention of Water and Soil Environmental Pollution, Xinyang Normal University, 464000, China
| | - Wei Zou
- School of Environment, Key Laboratory of Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory of Environmental Pollution Control, Henan Normal University, Xinxiang, 453007, China.
| |
Collapse
|
42
|
Vargas-Lizarazo AY, Ali MA, Mazumder NA, Kohli GM, Zaborska M, Sons T, Garnett M, Senanayake IM, Goodson BM, Vargas-Muñiz JM, Pond A, Jensik PJ, Olson ME, Hamilton-Brehm SD, Kohli P. Electrically polarized nanoscale surfaces generate reactive oxygenated and chlorinated species for deactivation of microorganisms. SCIENCE ADVANCES 2024; 10:eado5555. [PMID: 39093965 PMCID: PMC11636998 DOI: 10.1126/sciadv.ado5555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024]
Abstract
Because of the decreasing supply of new antibiotics, recent outbreaks of infectious diseases, and the emergence of antibiotic-resistant microorganisms, it is imperative to develop new effective strategies for deactivating a broad spectrum of microorganisms and viruses. We have implemented electrically polarized nanoscale metallic (ENM) coatings that deactivate a wide range of microorganisms including Gram-negative and Gram-positive bacteria with greater than 6-log reduction in less than 10 minutes of treatment. The electrically polarized devices were also effective in deactivating lentivirus and Candida albicans. The key to the high deactivation effectiveness of ENM devices is electrochemical production of micromolar cuprous ions, which mediated reduction of oxygen to hydrogen peroxide. Formation of highly damaging species, hydroxyl radicals and hypochlorous acid, from hydrogen peroxide contributed to antimicrobial properties of the ENM devices. The electric polarization of nanoscale coatings represents an unconventional tool for deactivating a broad spectrum of microorganisms through in situ production of reactive oxygenated and chlorinated species.
Collapse
Affiliation(s)
- Annie Y. Vargas-Lizarazo
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - M. Aswad Ali
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - Nehal A. Mazumder
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | | | - Miroslava Zaborska
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - Tyler Sons
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA
| | - Michelle Garnett
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA
| | - Ishani M. Senanayake
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - Boyd M. Goodson
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
| | - José M. Vargas-Muñiz
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA
| | - Amber Pond
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA
| | - Philip J. Jensik
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA
| | - Michael E. Olson
- Department of Medical Microbiology, Immunology and Cell Biology, School of Medicine, Southern Illinois University, Springfield, IL 62702, USA
| | | | - Punit Kohli
- School of Chemical and Biomolecular Sciences, Southern Illinois University, Carbondale, IL 62901, USA
- Integrated Microscopy and Graphics Expertise (IMAGE) Center, Southern Illinois University, Carbondale, IL 62901, USA
| |
Collapse
|
43
|
Zheng J, Meng W, Chen S, Cui Z, Xian X, Tian J, Krysko DV, Li B, Zhang W. A near-infrared broad-spectrum antimicrobial nanoplatform powered by bacterial metabolic activity for enhanced antimicrobial photodynamic-immune therapy. Acta Biomater 2024; 184:335-351. [PMID: 38936751 DOI: 10.1016/j.actbio.2024.06.024] [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: 04/24/2024] [Revised: 05/14/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
The emergence of antimicrobial-resistant bacterial infections poses a significant threat to public health, necessitating the development of innovative and effective alternatives to antibiotics. Photodynamic therapy (PDT) and immunotherapy show promise in combating bacteria. However, PDT's effectiveness is hindered by its low specificity to bacteria, while immunotherapy struggles to eliminate bacteria in immunosuppressive environments. In this work, we introduce an innovative near-infrared antimicrobial nanoplatform (ZFC) driven by bacterial metabolism. ZFC, comprising d-cysteine-functionalized pentafluorophenyl bacteriochlorin (FBC-Cy) coordinated with Zn2+, is designed for antimicrobial photodynamic-immune therapy (aPIT) against systemic bacterial infections. By specifically targeting bacteria via d-amino acid incorporation into bacterial surface peptidoglycans during metabolism, ZFC achieves precise bacterial clearance in wound and pulmonary infections, exhibiting an antimicrobial efficacy of up to 90 % with minimal damage to normal cells under 750 nm light. Additionally, ZFC enhances the activation of antigen-presenting cells by 3.2-fold compared to control groups. Furthermore, aPIT induced by ZFC triggers systemic immune responses and establishes immune memory, resulting in a 1.84-fold increase in antibody expression against bacterial infections throughout the body of mice. In conclusion, aPIT prompted by ZFC presents a approach to treating bacterial infections, offering a broad-spectrum solution for systemic bacterial infections. STATEMENT OF SIGNIFICANCE: The new concept demonstrated focuses on an innovative near-infrared antimicrobial nanoplatform (ZFC) for antimicrobial photodynamic-immune therapy (aPIT), highlighting its reliance on bacterial metabolism and its non-damaging effect on normal tissues. ZFC efficiently targets deep-tissue bacterial infections by harnessing bacterial metabolism, thereby enhancing therapeutic efficacy while sparing normal tissues from harm. This approach not only clears bacterial infections effectively but also induces potent adaptive immune responses, leading to the eradication of distant bacterial infections. By emphasizing ZFC's unique mechanism driven by bacterial metabolism and its tissue-sparing properties, this work underscores the potential for groundbreaking advancements in antimicrobial therapy. Such advancements hold promise for minimizing collateral damage to healthy tissues, thereby improving treatment outcomes and mitigating the threat of antimicrobial resistance. This integrated approach represents a significant progress forward in the development of next-generation antimicrobial therapies with enhanced precision and efficacy.
Collapse
Affiliation(s)
- Jiahao Zheng
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Wangyang Meng
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Suwen Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Zepeng Cui
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Xueying Xian
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Jia Tian
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
44
|
Yan W, Bai R, Zhang Q, Jiang Y, Chen G, Zhang Y, Wu Y, Guo X, Xiao Y, Zhao F. Metagenomic insights into ecological risk of antibiotic resistome and mobilome in riverine plastisphere under impact of urbanization. ENVIRONMENT INTERNATIONAL 2024; 190:108946. [PMID: 39151267 DOI: 10.1016/j.envint.2024.108946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Microplastics (MPs) are of increasing concern due to their role as reservoirs for antibiotic resistance genes (ARGs) and pathogens. To date, few studies have explored the influence of anthropogenic activities on ARGs and mobile genetic elements (MGEs) within various riverine MPs, in comparison to their natural counterparts. Here an in-situ incubation was conducted along heavily anthropogenically-impacted Houxi River to characterize the geographical pattern of antibiotic resistome, mobilome and pathogens inhabiting MPs- and leaf-biofilms. The metagenomics result showed a clear urbanization-driven profile in the distribution of ARGs, MGEs and pathogens, with their abundances sharply increasing 4.77 to 19.90 times from sparsely to densely populated regions. The significant correlation between human fecal marker crAssphage and ARG (R2 = 0.67, P=0.003) indicated the influence of anthropogenic activity on ARG proliferation in plastisphere and natural leaf surfaces. And mantel tests and random forest analysis revealed the impact of 17 socio-environmental factors, e.g., population density, antibiotic concentrations, and pore volume of materials, on the dissemination of ARGs. Partial least squares-path modeling further unveiled that intensifying human activities not only directly boosted ARGs abundance but also exerted a comparable indirect impact on ARGs propagation. Furthermore, the polyvinylchloride plastisphere created a pathogen-friendly habitat, harboring higher abundances of ARGs and MGEs, while polylactic acid are not likely to serve as vectors for pathogens in river, with a lower resistome risk score than that in leaf-biofilms. This study highlights the diverse ecological risks associated with the dissemination of ARGs and pathogens in varied MPs, offering insights for the policymaking of usage and control of plastics within urbanization.
Collapse
Affiliation(s)
- Weifu Yan
- CAS Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, Fujian 361021, China
| | - Rui Bai
- CAS Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, Fujian 361021, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Qinghua Zhang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Yuhao Jiang
- Academy of Forest Inventory and Planning, National Forestry and Grassland Administration, Beijing 100714, China
| | - Geng Chen
- CAS Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, Fujian 361021, China
| | - Yanru Zhang
- Fujian Key Laboratory of Pollution Control & Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Yicheng Wu
- Fujian Engineering and Research Center of Rural Sewage Treatment and Water Safety, Xiamen University of Technology, Xiamen 361024, China
| | - Xuetao Guo
- College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yong Xiao
- CAS Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, Fujian 361021, China.
| | - Feng Zhao
- CAS Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, Fujian 361021, China.
| |
Collapse
|
45
|
Pramanik A, Rai S, Gates K, Kolawole OP, Kundu S, Kasani-Akula P, Singh J, Dasary J, Zhang H, Han FX, Ray PC. Sunlight-Driven Photothermally Boosted Photocatalytic Eradication of Superbugs Using a Plasmonic Gold Nanoparticle-Decorated WO 3 Nanowire-Based Heterojunction. ACS OMEGA 2024; 9:32256-32267. [PMID: 39072127 PMCID: PMC11270714 DOI: 10.1021/acsomega.4c05327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 07/30/2024]
Abstract
Superbug infections are currently one of the biggest global health problems in our society. Herein, we report the design of a plasmonic gold nanoparticle (GNP)-decorated WO3 nanowire-based heterojunction for the proficient usage of sunlight-based renewable energy to inactivate 100% superbugs via photothermally boosted photocatalytic action. Additionally, a synergistic photothermal and photocatalytic approach has been used for sunlight-driven complete eradication of carbapenem-resistant Enterobacteriaceae Escherichia coli (CRE E. coli) and methicillin-resistant Staphylococcus aureus (MRSA) superbugs. Interestingly, photocatalytic activity of methylene blue (MB) dye degradation in the presence of 670 nm near-infrared light shows that photothermally boosted photocatalytic performance is much superior to that of only a photocatalytic or photothermal process. The observed higher photocatalytic performance for the heterojunction is because the plasmonic GNP enhanced the absorption capability at 670 nm and increased the temperature of the photocatalyst surface, which reduces the activation energy of the degradation reaction. Similarly, sunlight-driven photocatalytic experiments show 100% degradation of MB after 60 min of sunlight irradiation. Moreover, sunlight-based photocatalytic inactivation of MRSA and CRE E. coli experiments show 100% inactivation after 60 min of light irradiation.
Collapse
Affiliation(s)
- Avijit Pramanik
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Shivangee Rai
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Kaelin Gates
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Olorunsola Praise Kolawole
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Sanchita Kundu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Pragathi Kasani-Akula
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Jagriti Singh
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Jerusha Dasary
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Huimin Zhang
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Fengxiang X. Han
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Paresh Chandra Ray
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| |
Collapse
|
46
|
Zhong L, Sun HJ, Pang JW, Ding J, Zhao L, Xu W, Yuan F, Zhang LY, Ren NQ, Yang SS. Ciprofloxacin affects nutrient removal in manganese ore-based constructed wetlands: Adaptive responses of macrophytes and microbes. JOURNAL OF HAZARDOUS MATERIALS 2024; 473:134579. [PMID: 38761761 DOI: 10.1016/j.jhazmat.2024.134579] [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: 11/22/2023] [Revised: 03/28/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024]
Abstract
Ciprofloxacin (CIP) has received considerable attention in recent decades due to its high ecological risk. However, little is known about the potential response of macrophytes and microbes to varying levels of CIP exposure in constructed wetlands. Therefore, lab-scale manganese ore-based tidal flow constructed wetlands (MO-TFCWs) were operated to evaluate the responses of macrophytes and microbes to CIP over the long term. The results indicated that total nitrogen removal improved from 79.93% to 87.06% as CIP rose from 0 to 4 mg L-1. The chlorophyll content and antioxidant enzyme activities in macrophytes were enhanced under CIP exposure, but plant growth was not inhibited. Importantly, CIP exposure caused a marked evolution of the substrate microbial community, with increased microbial diversity, expanded niche breadth and enhanced cooperation among the top 50 genera, compared to the control (no CIP). Co-occurrence network also indicated that microorganisms may be more inclined to co-operate than compete. The abundance of the keystone bacterium (involved in nitrogen transformation) norank_f__A0839 increased from 0.746% to 3.405%. The null model revealed drift processes (83.33%) dominated the community assembly with no CIP and 4 mg L-1 CIP. Functional predictions indicated that microbial carbon metabolism, electron transfer and ATP metabolism activities were enhanced under prolonged CIP exposure, which may contribute to nitrogen removal. This study provides valuable insights that will help achieve stable nitrogen removal from wastewater containing antibiotic in MO-TFCWs.
Collapse
Affiliation(s)
- Le Zhong
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Han-Jun Sun
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Ji-Wei Pang
- China Energy Conservation and Environmental Protection Group, CECEP Digital Technology Co., Ltd., Beijing 100096, China
| | - Jie Ding
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China.
| | - Lei Zhao
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Wei Xu
- General Water of China Co., Ltd., Beijing 100022, China
| | - Fang Yuan
- General Water of China Co., Ltd., Beijing 100022, China
| | - Lu-Yan Zhang
- School of Environmental Science and Engineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Nan-Qi Ren
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Shan-Shan Yang
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China.
| |
Collapse
|
47
|
Li L, Xie Y, Wang J, Sun Q, Gao M, Li C. Biofilm microenvironment-activated multimodal therapy nanoplatform for effective anti-bacterial treatment and wound healing. Acta Biomater 2024; 183:221-234. [PMID: 38849021 DOI: 10.1016/j.actbio.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
Antimicrobial drug development faces challenges from bacterial resistance, biofilms, and excessive inflammation. Here, we design an intelligent nanoplatform utilizing mesoporous silica nanoparticles doped with copper ions for loading copper sulfide (DM/Cu2+-CuS). The mesoporous silica doped with tetrasulfide bonds responds to the biofilm microenvironment (BME), releasing Cu2+ions, CuS along with hydrogen sulfide (H2S) gas. The release of hydrogen sulfide within 72 h reached 793.5 µM, significantly higher than that observed with conventional small molecule donors. H2S induces macrophages polarization towards the M2 phenotype, reducing inflammation and synergistically accelerating endothelial cell proliferation and migration with Cu2+ions. In addition, H2S disrupts extracellular DNA within biofilms, synergistically photothermal enhanced peroxidase-like activity of CuS to effectively eradicate biofilms. Remarkably, DM-mediated consumption of endogenous glutathione enhances the anti-biofilm activity of H2S and improves oxygen species (ROS) destruction efficiency. The combination of photothermal therapy (PTT), chemodynamic therapy (CDT), and gas treatment achieves sterilization rates of 99.3 % and 99.6 % against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli), respectively, in vitro under 808 nm laser irradiation. Additionally, in vivo experiments demonstrate a significant biosafety and antibacterial potential. In summary, the H2S donor developed in this study exhibits enhanced biocompatibility and controlled release properties. By integrating BME-responsive gas therapy with antibacterial ions, PTT and CDT, a synergistic multimodal strategy is proposed to offer new therapeutic approaches for wound healing. STATEMENT OF SIGNIFICANCE: The advanced DMOS/Cu2+-CuS (DMCC) multimodal therapeutic nanoplatform has been developed for the treatment of drug-resistant bacterial wound infections and has exhibited enhanced therapeutic efficacy through the synergistic effects of photothermal therapy, chemodynamic therapy, Cu2+ions, and H2S. The DMCC exhibited exceptional biocompatibility and could release CuS, Cu2+, and H2S in response to elevated concentrations of glutathione within the biofilm microenvironment. H2S effectively disrupted the biofilm structure. Meanwhile, peroxidase activity of CuS combined with GSH-mediated reduction of Cu2+ to Cu+ generated abundant hydroxyl radicals under acidic conditions, leading to efficient eradication of pathogenic bacteria. Furthermore, both H2S and Cu2+ could modulate M2 macrophages polarization and regulate immune microenvironment dynamics. These strategies collectively provided a novel approach for developing antibacterial nanomedical platforms.
Collapse
Affiliation(s)
- Lei Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China
| | - Yulin Xie
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China
| | - Junrong Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China
| | - Qianqian Sun
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China
| | - Minghong Gao
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China.
| | - Chunxia Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, 266237, PR China.
| |
Collapse
|
48
|
Su W, Li P, Zhong L, Liang W, Li T, Liu J, Ruan T, Jiang G. Occurrence and Distribution of Antibacterial Quaternary Ammonium Compounds in Chinese Estuaries Revealed by Machine Learning-Assisted Mass Spectrometric Analysis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:11707-11717. [PMID: 38871667 DOI: 10.1021/acs.est.4c02380] [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: 06/15/2024]
Abstract
Antimicrobial resistance (AMR) undermines the United Nations Sustainable Development Goals of good health and well-being. Antibiotics are known to exacerbate AMR, but nonantibiotic antimicrobials, such as quaternary ammonium compounds (QACs), are now emerging as another significant driver of AMR. However, assessing the AMR risks of QACs in complex environmental matrices remains challenging due to the ambiguity in their chemical structures and antibacterial activity. By machine learning prediction and high-resolution mass spectrometric analysis, a list of antibacterial QACs (n = 856) from industrial chemical inventories is compiled, and it leads to the identification of 50 structurally diverse antibacterial QACs in sediments, including traditional hydrocarbon-based compounds and new subclasses that bear additional functional groups, such as choline, ester, betaine, aryl ether, and pyridine. Urban wastewater, aquaculture, and hospital discharges are the main factors influencing QAC distribution patterns in estuarine sediments. Toxic unit calculations and metagenomic analysis revealed that these QACs can influence antibiotic resistance genes (particularly sulfonamide resistance genes) through cross- and coresistances. The potential to influence the AMR is related to their environmental persistence. These results suggest that controlling the source, preventing the co-use of QACs and sulfonamides, and prioritizing control of highly persistent molecules will lead to global stewardship and sustainable use of QACs.
Collapse
Affiliation(s)
- Wenyuan Su
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pengyang Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Laijin Zhong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenqing Liang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingyu Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiyan Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Ruan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
49
|
Li Q, Wu J, Chen Z, Wang J, Gong Y, Yin X. Prevalence of self-medication with antibiotics and its related factors among the general public and health professionals during the COVID-19 pandemic: A cross-sectional study in China. Am J Infect Control 2024; 52:759-764. [PMID: 38401644 DOI: 10.1016/j.ajic.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Self-medication with antibiotics (SMA) is a common public health concern. This study aimed to assess the prevalence of SMA in the general public and health professionals during the COVID-19 pandemic and identify the associated factors. METHODS A cross-sectional study was conducted from October 28, 2022, to November 6, 2022. Logistics regression analysis was used to examine the associated factors. RESULTS The rate of SMA was 10.25% in the general public and 12.69% in health professionals. For the public, those who perceived themselves as average or good health, had moderate antibiotic knowledge, and had easy access to nearby health facilities were less likely to SMA; while those who live in rural areas, found it easy to purchase antibiotics without prescriptions, and those who frequently encountered antibiotics recommended by pharmacy staff were more likely to SMA. For health professionals, those who were female, perceived themselves as good health, had moderate or high antibiotic knowledge, and had easy access to health facilities were less likely to SMA; while those who found it easy to purchase antibiotics without prescriptions were more likely to SMA. CONCLUSIONS SMA is prevalent in both the general public and health professionals. Promoting the rational use of antibiotics requires joint participation and effort.
Collapse
Affiliation(s)
- Qinnan Li
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianxiong Wu
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhenyuan Chen
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Wang
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanhong Gong
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoxv Yin
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
50
|
Li W, Zhang X, Hao X, Xin R, Zhang Y, Ma Y, Niu Z. Fish skin mucosal surface becomes a barrier of antibiotic resistance genes under apramycin exposure. ENVIRONMENTAL RESEARCH 2024; 252:118930. [PMID: 38615788 DOI: 10.1016/j.envres.2024.118930] [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: 12/26/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Antibiotic resistance genes (ARGs) are a kind of emerging environmental contamination, and are commonly found in antibiotic application situations, attracting wide attention. Fish skin mucosal surface (SMS), as the contact interface between fish and water, is the first line of defense against external pollutant invasion. Antibiotics are widely used in aquaculture, and SMS may be exposed to antibiotics. However, what happens to SMS when antibiotics are applied, and whether ARGs are enriched in SMS are not clear. In this study, Zebrafish (Danio rerio) were exposed to antibiotic and antibiotic resistant bacteria in the laboratory to simulate the aquaculture situation, and the effects of SMS on the spread of ARGs were explored. The results showed that SMS maintained the stability of the bacterial abundance and diversity under apramycin (APR) and bacterial exposure effectively. Until 11 days after stopping APR exposure, the abundance of ARGs in SMS (mean value was 3.32 × 10-3 copies/16S rRNA copies) still did not recover to the initial stage before exposure, which means that enriched ARGs in SMS were persistently remained. Moreover, non-specific immunity played an important role in resisting infection of external contamination. Besides, among antioxidant proteins, superoxide dismutase showed the highest activity. Consequently, it showed that SMS became a barrier of antibiotic resistance genes under APR exposure, and ARGs in SMS were difficult to remove once colonized. This study provided a reference for understanding the transmission, enrichment process, and ecological impact of antibiotics and ARGs in aquatic environments.
Collapse
Affiliation(s)
- Wenpeng Li
- School of Marine Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Xiaohan Zhang
- School of Environmental Science and Engineering, Tianjin University, Tianjin, 300350, China
| | - Xiaohan Hao
- School of Marine Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Rui Xin
- School of Marine Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Ying Zhang
- School of Environmental Science and Engineering, Tianjin University, Tianjin, 300350, China
| | - Yongzheng Ma
- School of Marine Science and Technology, Tianjin University, Tianjin, 300072, China.
| | - Zhiguang Niu
- School of Marine Science and Technology, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|