1
|
Yin Z, Huang D, Kuhn EMA, Moriarty TF, Li G, Wang X. Unraveling persistent bacteria: Formation, niches, and eradication strategies. Microbiol Res 2025; 297:128189. [PMID: 40311456 DOI: 10.1016/j.micres.2025.128189] [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: 03/12/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
Persistent bacteria (persisters) are phenotypic variants that emerge either randomly or in response to a range of adverse environmental conditions. Persistence represents a state whereby a subpopulation of microorganisms can spontaneously enter a "dormant" state in response to environmental factors, while simultaneously exhibiting elevated tolerance to antimicrobial agents. This review provides the current definition of bacterial persistence and summarizes the mechanisms of persisters formation as well as the various niches of bacterial persistence encountered in clinical practice. Strategies targeting persisters are outlined, including but not limited to direct killing, awakening of persistent bacteria, combined clearance, and inhibition of persistence formation, and we conclude by proposing challenges and solutions for addressing bacterial persistence in current clinical practice.
Collapse
Affiliation(s)
- Zibo Yin
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China
| | - Diandian Huang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China
| | | | | | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China.
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China.
| |
Collapse
|
2
|
Li R, Sun M, Li ZH, Qu Y, Li Y, Ampomah-Wireko M, Li D, Kong H, Wu Y, Hossain AA, Zhang E. Important Role of Triphenylamine in Modulating the Antibacterial Performance Relationships of Antimicrobial Peptide Mimics by Alkyl Chain Engineering. J Med Chem 2025; 68:10299-10313. [PMID: 40270226 DOI: 10.1021/acs.jmedchem.5c00454] [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: 04/25/2025]
Abstract
Multidrug resistance (MDR) bacteria pose a serious threat to human health, and the development of effective antimicrobial drugs is urgent. Herein, we used alkyl chain engineering to design and synthesize two series of antimicrobial peptide mimics with distinct cores: triphenylamine quaternary ammonium derivatives (TPQs) and diphenylethene quaternary ammonium derivatives (BPQs), and we investigated the effect of varying the alkyl chain lengths on antibacterial activity. We found that the introduction of a triphenylamine group significantly enhances the antibacterial activity of short-chain dimethyl quaternary ammonium derivatives while maintaining their excellent biocompatibility. Most notably, TPQ-1 exhibited negligible invasiveness toward living cells and possesses good antimicrobial activities, with good efficacy against biofilms and persisters. Moreover, TPQ-1 exhibited good antimicrobial effects in vivo and significantly accelerated the healing process of methicillin-resistant Staphylococcus aureus-infected wounds. This work promotes the practical application of antimicrobial peptide mimics and triphenylamine derivatives.
Collapse
Affiliation(s)
- Ruirui Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Moran Sun
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Zhi-Hao Li
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Ye Qu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Yuanbo Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Daran Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Hongtao Kong
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Yuequan Wu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Adib Azwad Hossain
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - En Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
- Pingyuan Laboratory (Zhengzhou University), Zhengzhou 450001, China
| |
Collapse
|
3
|
Garcia-Maset R, Chu V, Yuen N, Blumgart D, Yoon J, Murray BO, Joseph AA, Rohn JL. Effect of host microenvironment and bacterial lifestyles on antimicrobial sensitivity and implications for susceptibility testing. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:42. [PMID: 40399473 PMCID: PMC12095824 DOI: 10.1038/s44259-025-00113-3] [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: 02/11/2025] [Accepted: 05/01/2025] [Indexed: 05/23/2025]
Abstract
Bacterial infections remain a major global health issue, with antimicrobial resistance (AMR) worsening the crisis. However, treatment failure can occur even when bacteria show antibiotic susceptibility in diagnostic tests. We explore factors such as phenotypic resilience, bacterial lifestyles such as biofilms, and differences between laboratory tests and real infection sites, highlighting the need for improved platforms to better predict treatment outcomes, and reviewing emerging technologies aimed at improving susceptibility testing.
Collapse
Affiliation(s)
- Ramon Garcia-Maset
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| | - Victoria Chu
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Nicholas Yuen
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Dalia Blumgart
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Jenny Yoon
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Benjamin O Murray
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Amelia A Joseph
- Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Jennifer L Rohn
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
4
|
Chen Y, Chen QW, Fu FS, Yan JH, Su J, Zhang XZ, Yu A. Antibacterial Peptides-Produced Cell Hydrogel Eliminates Intracellular Pathogens and Remodels Immune Microenvironment for Osteomyelitis Therapy. ACS NANO 2025; 19:18433-18449. [PMID: 40336314 DOI: 10.1021/acsnano.5c01600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Featuring bacterial invasion and colonization within cells, chronic infection-induced immune suppression, and inflammatory cell infiltration, osteomyelitis is currently an intractable and recurrent bone disease. In this study, an injectable hydrogel that gels in situ and is loaded with engineered antimicrobial cells (LL37-MSC@OCAHM) is developed. This anti-inflammatory hydrogel not only maintains cell activity in the inflammatory environment but also releases magnesium ions (Mg2+) to promote the differentiation of MSCs into bone-forming cells, contributing to bone mass formation, enhancing bone repair, and accelerating bone healing. The engineered cells continuously produce antimicrobial peptides of LL37, which effectively kill both extracellular and intracellular bacteria at the osteomyelitis site. Additionally, cell hydrogel also modulates the immune response by shifting the osteomyelitis environment from pro-inflammatory to anti-inflammatory, reducing the infiltration of immune cells and myeloid-derived suppressor cells (MDSCs). We also demonstrated its ability to activate immune responses and generate immune memory, thereby preventing the recurrence of secondary infections. This study introduces an engineered cell-based approach that combines active antimicrobial effect, immune modulation, and bone repair, for effectively eliminating osteomyelitis infections and preventing recurrence.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Fang-Sheng Fu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Jian-Hua Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Junwei Su
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| |
Collapse
|
5
|
Nakamura M, Tomoda Y, Kobayashi M, Hanaki H, Kanoh Y. Sulfamethoxazole-trimethoprim plus rifampicin combination therapy for methicillin-resistant Staphylococcus aureus infection: An in vitro study. PLoS One 2025; 20:e0323935. [PMID: 40392929 PMCID: PMC12091750 DOI: 10.1371/journal.pone.0323935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/16/2025] [Indexed: 05/22/2025] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is highly drug-resistant. The current Japanese guidelines (2019 edition) for managing and treating MRSA infections mention alternative anti-MRSA agents, including sulfamethoxazole-trimethoprim (ST) and rifampicin (RFP). Both ST and RFP are oral drugs and are expected to be effective alternatives to anti-MRSA drugs in clinical cases where anti-MRSA drugs are not indicated. Although guidelines for treating MRSA infections describe the efficacy of ST-RFP combination therapy and although it is used in clinical practice, only a limited number of in vitro studies have demonstrated its efficacy in pharmacokinetic/pharmacodynamic models. This study aimed to investigate the efficacy of the combination therapy of ST-RFP against MRSA in some in vitro models, including a pharmacokinetic/pharmacodynamic model. The MRSA strains obtained were subjected to antibiotic susceptibility tests. A checkerboard assay for drug combination synergy of ST-RFP was performed. Furthermore, a chemostat model was used to validate the combination therapy of ST-RFP as an in vitro pharmacokinetic/pharmacodynamic model. Viable cell counts and antibiotic concentrations were measured. The checkerboard assay showed that the ST-RFP combination had an additive effect on all strains (the lowest fractional inhibitory concentration index ranged from 0.63 to 1.00). The in vitro chemostat model demonstrated the usefulness of the combination of ST-RFP against MRSA, especially in ST-resistant strains. Regrowth of MRSA was observed in RFP monotherapy but not in the ST-RFP combination therapy. This study demonstrated the potential effectiveness of the ST-RFP combination against ST-resistant MRSA, providing important foundational data that may support future clinical investigations.
Collapse
Affiliation(s)
- Masaki Nakamura
- Department of Laboratory Medicine, Kitasato University School of Medicine, Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
- Ōmura Satoshi Memorial Institute, Kitasato University, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yoshinori Tomoda
- Laboratory of Clinical Pharmacokinetics, Research and Education Center for Clinical Pharmacy, Kitasato University School of Pharmacy, Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| | - Masahiro Kobayashi
- Laboratory of Clinical Pharmacokinetics, Research and Education Center for Clinical Pharmacy, Kitasato University School of Pharmacy, Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| | - Hideaki Hanaki
- Ōmura Satoshi Memorial Institute, Kitasato University, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yuhsaku Kanoh
- Department of Laboratory Medicine, Kitasato University School of Medicine, Kitazato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| |
Collapse
|
6
|
Ma Q, Liu Y, Cen J, Wang Q, Chen M, Chen S, Zhang Z, Han K, Feng Z, Wu C, Shen J, Jiang H. Disrupting cross-adaptation in high-risk MRSA: Sanguinarine as a multi-effective stress sensitizer for environmental and food safety. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138586. [PMID: 40367773 DOI: 10.1016/j.jhazmat.2025.138586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a significant public health concern owing to its formidable antibiotic resistance and robust capacity for biofilm formation. The cross-adaptation mechanism enables MRSA to develop tolerance to environmental stressors such as antibiotics, acid, heat and osmotic pressure, leading to the persistence infections and environmental contamination. The cross-adaptation mechanism enables MRSA to develop tolerance to environmental stressors, such as antibiotics, acid, heat and osmotic pressure, leading to the persistence infections and environmental contamination. Here, we identified 261 strains of S. aureus and 9 high-risk MRSA from the environment of dairy farms and raw milk. The natural product Sanguinarine (SAN), derived from feed additives, exhibits effective anti-MRSA and anti-biofilm activity. Notably, SAN enhances the sensitivity of MRSA to antibiotics, acid, heat, and osmotic pressure by disrupting the cross-adaptation mechanism. Mechanistic investigations revealed that SAN significantly reduces the transcriptional level of type I (dnaK, groEL, etc.) and type III (clpB, clpP, etc.) heat stress response genes while markedly upregulating type II (σB) gene. Furthermore, SAN upregulates Na+/H+ antiporters activity, F0F1-ATPase activity and purine metabolism, while broadly downregulating DNA damage repair genes and disrupting ribosomal function. Additionally, SAN induces non-synonymous mutations in key stress response factors ClpB/L, leading to a loss of conformational homeostasis. SAN elicits a distinct stress response compared to environmental stressors, weakening MRSA's resilience and demonstrating promising capabilities for MRSA clearance and biofilm inhibition. Overall, SAN provides an effective strategy for the clearance of high-risk MRSA and the assurance of public health security.
Collapse
Affiliation(s)
- Qiang Ma
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yuan Liu
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jianuo Cen
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qiqi Wang
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Meinuo Chen
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shiqi Chen
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhimeng Zhang
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ke Han
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhiyue Feng
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Congming Wu
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haiyang Jiang
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| |
Collapse
|
7
|
Chakraborty A, Bhakta K, Ghosh A, Manna D, Maity AR, Sikder K, Chakraborti S, Basu A. Artesunate Perturbs GTP Binding of the Conserved GTPase Obg Thereby Alleviating Antibiotic Resistance in Methicillin-Resistant Staphylococcus aureus. ACS Infect Dis 2025; 11:1190-1202. [PMID: 40278541 DOI: 10.1021/acsinfecdis.4c01027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is an important nosocomial pathogen that causes various secondary infections among hospital-associated patients. The pathogen is challenging to treat due to its resistance to a wide spectrum of antibiotics, including the last-resort antibiotic vancomycin and newly developed drugs, such as linezolid and daptomycin. While the invention of entirely new drugs to combat MRSA infection seems almost impossible, potentiating the efficacy of conventional antibiotics is critical. Our article explores the novel application of the antimalarial drug artesunate, which enhances the efficacy of vancomycin and cefoxitin in treating MRSA infections. We focused on ObgSa, a conserved GTPase in MRSA, and found that artesunate selectively binds to its GTP-binding pocket. We further evaluated the GTP-binding activity and metal dependence (specifically, Mg2+) of this conserved GTPase. In silico analysis identified several threonine residues essential for GTP binding, which were subsequently mutated to assess their role in GTP binding. As shown in the analysis, these mutations significantly impacted both the GTP binding and hydrolysis functions of ObgSa. Notably, these threonine residues were also crucial for artesunate binding within the GTP-binding domain. When the effect of artesunate was assessed, the drug competitively inhibited GTP binding and hydrolysis of the GTPase. This result was manifested as reduced antibiotic tolerance, disruption of biofilms, and a decrease in persister cells─critical factors in chronic infections. In summary, our research presents an innovative strategy to combat antimicrobial resistance through artesunate, highlighting its potential effectiveness in eradicating infections.
Collapse
Affiliation(s)
- Asmita Chakraborty
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Howrah, West Bengal 711202, India
| | - Koustav Bhakta
- Biological Sciences Unified Academic Campus Bose Institute EN-80, Sector V Bidhan Nagar, Kolkata 700 091, India
| | - Abhrajyoti Ghosh
- Biological Sciences Unified Academic Campus Bose Institute EN-80, Sector V Bidhan Nagar, Kolkata 700 091, India
| | - Dipak Manna
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Howrah, West Bengal 711202, India
| | - Amit Ranjan Maity
- Institute of Biotechnology, Amity University Kolkata Campus, Major Arterial Road, Action Area II, Newtown, Kadampukur, Kolkata, West Bengal 700135, India
| | - Kunal Sikder
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Howrah, West Bengal 711202, India
| | - Soumyananda Chakraborti
- Department of Biological Science, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Hyderabad, Telangana 500078, India
| | - Arnab Basu
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Howrah, West Bengal 711202, India
| |
Collapse
|
8
|
Jiang WH, Zhao XW, Jin XM, Wang WJ, Chen Z. Mixed Infections in the Female Lower Genital Tract: Unlocking the Current Landscape and Future Directions. Curr Med Sci 2025:10.1007/s11596-025-00058-8. [PMID: 40327219 DOI: 10.1007/s11596-025-00058-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
Understanding mixed infections in the female lower genital tract is a critical challenge in modern infection research. The interplay of multiple pathogens complicates disease progression, often resulting in treatment failure, recurrent infections, and significant public health and economic burdens. These infections are further exacerbated by disrupted host immune responses, which hinder the recovery of the vaginal microecosystem. Additionally, microbial biofilms-a fundamental mode of pathogen coexistence-contribute to the persistence and drug resistance of these infections, complicating management strategies. This review examines the pathogenesis, diagnosis, and treatment of mixed infections in the female lower genital tract while exploring potential avenues for future research. These findings emphasize the need for greater focus on these infections and offer insights to enhance further research in this area.
Collapse
Affiliation(s)
- Wen-Hua Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xin-Wei Zhao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xi-Ming Jin
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen-Jia Wang
- Department of Child Healthcare, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhuo Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
9
|
Giorgio RT, Helaine S. Antibiotic-recalcitrant Salmonella during infection. Nat Rev Microbiol 2025; 23:276-287. [PMID: 39558126 DOI: 10.1038/s41579-024-01124-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2024] [Indexed: 11/20/2024]
Abstract
Antibiotic-recalcitrant infections, defined as the prolonged carriage of pathogenic bacteria even in the presence of antibiotics, are often caused by bacteria that are genetically susceptible to the drug. These recalcitrant bacteria fail to proliferate in the presence of antibiotics but remain viable such that they may recolonize their niche following antibiotic withdrawal. Significant progress has been made in our understanding of antibiotic-recalcitrant Salmonella, which are thought to be the source of infection relapse. In recent years, it has been shown that recalcitrant bacteria manipulate host immune defences and could directly contribute to the spread of antimicrobial resistance. In this Review, we provide an overview of what is currently known about the antibiotic recalcitrance of Salmonella during infection and highlight knowledge gaps requiring additional research in the future.
Collapse
Affiliation(s)
- Rachel T Giorgio
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Sophie Helaine
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Tang H, Wang Z, Li C, Yu J, Huang W, Zhou T, Zhang C, Wen B, Wang C, Zhu X, Wang D, Tao J, Lu J, Ni J, Yao YF. Disruption of sulfur transferase complex increases bacterial intramacrophage persistence. PLoS Pathog 2025; 21:e1013136. [PMID: 40367211 PMCID: PMC12077765 DOI: 10.1371/journal.ppat.1013136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Bacterial persisters contribute significantly to clinical treatment failure and relapse. These cells could resist antibiotic treatment via transient phenotypic and gene expression alterations. We conducted a high-throughput screening of Salmonella Typhimurium transposon mutants to identify key genes for intramacrophage antibiotic persistence. The results show that a sulfur transferase complex encoded by yheM, yheL, yheN, trmU and yhhP are involved in bacterial intramacrophage antibiotic persistence. Salmonella could persist in macrophages by downregulating the expression of the sulfur transferase complex during exposure to high concentrations of antibiotics, and even in a persistent infection mouse model. Mechanistically, deletion of yheM increases reactive nitrogen species (RNS) in the exponential phase, which inhibits bacterial respiration and ATP generation. In contrast, absence of yheM promotes persister formation by elevating (p)ppGpp levels in the stationary phase. Taken together, our data demonstrate that bacteria use the sulfur transferase to coordinate intramacrophage replication and persistence for adaptation to various environmental stresses. These findings reveal the role of the sulfur transferase complex in bacterial intramacrophage persistence and provide a promising target for antibacterial infection therapy.
Collapse
Affiliation(s)
- Huang Tang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zuoqiang Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Congcong Li
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingchen Yu
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanqiu Huang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Zhou
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuanzhen Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Bingjie Wen
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengyue Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaocen Zhu
- Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China,
| | - Danni Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Tao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Lu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinjing Ni
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Feng Yao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, China
| |
Collapse
|
11
|
Ge T, Wu R, Yu T, Hasan MSU, Liu J. Halogen anion modulated metal-organic frameworks with enhanced nanozyme activities for bacterial biofilm disruption. NANOSCALE 2025; 17:9963-9973. [PMID: 40145893 DOI: 10.1039/d5nr00131e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
There is an urgent need to develop new nanozymes with enhanced catalytic activities to combat bacterial infections, which have become increasingly challenging due to the misuse of antibiotics and the difficulties of new antibiotic discovery. Here, we employed a new strategy against bacterial biofilms by introducing halide anions to modulate the crystal facets of ZIF-L metal-organic frameworks (MOFs) and then loading chloroquine to form Ch@ZIF-L. The modulation of crystal facets significantly enhanced the oxidase activities of ZIF-L, which can be significantly changed by modulation of its crystal facets, with the hexagonal ZIF-L (ZIF-L-H-Cl) structure showing the highest oxidase activity. At pH 6.0, over 80% of chloroquine was released from Ch@ZIF-L-H-Cl within 8 hours, altering the DNA conformation of bacterial biofilms and disrupting the extracellular polymeric substances (EPSs). The generation of singlet oxygen catalyzed by ZIF-L-H-Cl can effectively kill bacteria at the infected wound site. The composite nanozyme of Ch@ZIF-L-H-Cl, when treated at 100 μg mL-1, exhibited no adverse effects on normal cell growth or hemolysis. Our in vivo experiments demonstrated an 85% reduction of the wound area by day 8 and a rapid recovery of body weight in mice with wounds infected with Staphylococcus aureus (S. aureus) biofilms. Furthermore, substantial reductions in bacterial counts were observed in both wounds and blood samples in the mice, highlighting the great potential of Ch@ZIF-L-H-Cl in combating bacterial biofilm infections.
Collapse
Affiliation(s)
- Tianjin Ge
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| | - Renfei Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| | - Tianrong Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| | - Muhammad Sajjad Ul Hasan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| | - Jian Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| |
Collapse
|
12
|
Han Y, Liu X, Qu S, Duan X, Xiang Y, Jiang N, Yang S, Fang X, Xu L, Wen H, Yu Y, Huang S, Huang J, Zhu K. Tissue geometry spatiotemporally drives bacterial infections. Cell 2025:S0092-8674(25)00394-0. [PMID: 40262607 DOI: 10.1016/j.cell.2025.03.042] [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: 11/15/2023] [Revised: 02/28/2025] [Accepted: 03/25/2025] [Indexed: 04/24/2025]
Abstract
Epithelial tissues serve as the first line of host against bacterial infections. The self-organization of epithelial tissues continuously adapts to the architecture and mechanics of microenvironments, thereby dynamically impacting the initial niche of infections. However, the mechanism by which tissue geometry regulates bacterial infection remains poorly understood. Here, we showed geometry-guided infection patterns of bacteria in epithelial tissues using bioengineering strategies. We discovered that cellular traction forces play a crucial role in the regulation of bacterial invasive sites and marginal infection patterns in epithelial monolayers through triggering co-localization of mechanosensitive ion channel protein Piezo1 with bacteria. Further, we developed precise mechanobiology-based strategies to potentiate the antibacterial efficacy in animal models of wound and intestinal infection. Our findings demonstrate that tissue geometry exerts a key impact on mediating spatiotemporal infections of bacteria, which has important implications for the discovery and development of alternative strategies against bacterial infections.
Collapse
Affiliation(s)
- Yiming Han
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiaoye Liu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing 102206, China
| | - Shaoqi Qu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; Animal-Derived Food Safety Innovation Team, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, China
| | - Xiaocen Duan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Yunqing Xiang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Nan Jiang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Shuyu Yang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xu Fang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Liang Xu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Hui Wen
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Yue Yu
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Shuqiang Huang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China.
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China.
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
13
|
Kavela S, Kakkerla S, Thupurani MK. Broad-spectrum antimicrobial activity and in vivo efficacy of SK1260 against bacterial pathogens. Front Microbiol 2025; 16:1553693. [PMID: 40276222 PMCID: PMC12019989 DOI: 10.3389/fmicb.2025.1553693] [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: 12/31/2024] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Antimicrobial resistance (AMR) is a growing global health concern, necessitating the development of novel therapeutic agents. Antimicrobial peptides (AMPs) have emerged as promising candidates due to their broad-spectrum activity and lower resistance potential. SK1260 is a newly developed AMP evaluated for its efficacy against Gram-positive and Gram-negative pathogens, including multidrug-resistant strains. Methods The antimicrobial activity of SK1260 was assessed through minimum inhibitory concentration (MIC) assays against clinical and reference strains of Escherichia coli, Staphylococcus aureus, Klebsiella pneumoniae, and Pseudomonas aeruginosa. Time-kill kinetics were performed to evaluate bactericidal activity over time. In vivo efficacy was determined using murine infection models, where bacterial burden, tissue pathology, and survival rates were assessed following peptide administration. Results SK1260 exhibited potent antibacterial activity, with MIC values ranging from 3.13 to 12.5 µg/mL. Time-kill studies demonstrated dose- and time-dependent bactericidal effects, achieving complete bacterial clearance at concentrations ≥1× MIC, comparable to ciprofloxacin. In vivo studies revealed significant reductions in bacterial loads in vital organs, reduced histopathological damage, and improved survival in treated mice. Peptide treatment restored normal tissue architecture and showed efficacy equivalent to standard antibiotic therapy. Discussion The study establishes SK1260 as a promising broad-spectrum antimicrobial agent with efficacy against drug-resistant pathogens. Its ability to reduce bacterial burden and protect tissue integrity in vivo highlights its therapeutic potential. Further preclinical development and clinical trials are warranted to explore SK1260 as a viable alternative in the fight against AMR.
Collapse
|
14
|
Post V, Pascoe B, Hitchings MD, Erichsen C, Fischer J, Morgenstern M, Richards RG, Sheppard SK, Moriarty TF. Methicillin-sensitive Staphylococcus aureus lineages contribute towards poor patient outcomes in orthopaedic device-related infections. Microb Genom 2025; 11:001390. [PMID: 40238650 PMCID: PMC12068410 DOI: 10.1099/mgen.0.001390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
Staphylococci are the most common cause of orthopaedic device-related infections (ODRIs), with Staphylococcus aureus responsible for a third or more of cases. This prospective clinical and laboratory study investigated the association of genomic and phenotypic variation with treatment outcomes in ODRI isolates. Eighty-six invasive S. aureus isolates were collected from patients with ODRI, and clinical outcome was assessed after a follow-up examination of 24 months. Each patient was then considered to have been 'cured' or 'not cured' based on predefined clinical criteria. Whole-genome sequencing and molecular characterization identified isolates belonging to globally circulating community- and hospital-acquired lineages. Most isolates were phenotypically susceptible to methicillin and lacked the staphylococcal cassette chromosome mec cassette [methicillin-susceptible S. aureus (MSSA); 94%] but contained several virulence genes, including toxins and biofilm genes. Whilst recognizing the role of the host immune response, we identified genetic variance, which could be associated with the infection severity or clinical outcome. Whilst this and several other studies reinforce the role antibiotic resistance [e.g. methicillin-resistant S. aureus (MRSA) infection] has on treatment failure, it is important not to overlook MSSA that can cause equally destructive infections and lead to poor patient outcomes.
Collapse
Affiliation(s)
| | - Ben Pascoe
- Ineos Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, USA
| | | | | | - Julian Fischer
- Centrum of Orthopedic Isartal, Pullach im Isartal, Germany
| | - Mario Morgenstern
- Department of Orthopedic and Trauma Surgery, University Hospital, Basel, Switzerland
| | | | - Samuel K. Sheppard
- Ineos Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
| | - T. Fintan Moriarty
- AO Research Institute Davos, Davos, Switzerland
- Department of Orthopedic and Trauma Surgery, University Hospital, Basel, Switzerland
| |
Collapse
|
15
|
Maalaoui A, Trimeche A, Marnet PG. Alternative approaches to antibiotics in the control of mastitis in dairy cows: a review. Vet Res Commun 2025; 49:150. [PMID: 40126814 DOI: 10.1007/s11259-025-10720-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/15/2025] [Indexed: 03/26/2025]
Abstract
Bovine mastitis is the most widespread and economically burdensome condition affecting dairy herds worldwide, causing substantial financial losses in the livestock and dairy sectors. The main approach to treating mastitis in dairy cows is based on the administration of antibiotics. However, their widespread use has led to the emergence of antibiotic-resistant pathogens, and thus to numerous food safety problems. Consequently, a growing body of scientific research has been directed towards exploring new and effective therapeutic alternatives for the management of bovine mastitis, which could replace conventional antibiotic therapy. This review surveys the various alternative strategies employed in the prevention and treatment of mastitis in dairy cattle. These strategies include nanoparticle therapy, bacteriophage therapy, vaccination, phytotherapy, the use of animal proteins, probiotics and bacteriocins. In addition, the potential synergistic effects resulting from the combination of these treatments has shown real benefits that will be highlighted.
Collapse
Affiliation(s)
- Abir Maalaoui
- Higher Institute of Biotechnology of Beja, University of Jendouba, Beja, 9000, Tunisia.
- Laboratoire de recherche gestion de la santé et de la qualité des productions animales, Ecole Nationale de Médecine Vétérinaire de Sidi Thabet, Université de La Manouba, Ariana, 2020, Tunisia.
| | - Abdesselem Trimeche
- Laboratoire de recherche gestion de la santé et de la qualité des productions animales, Ecole Nationale de Médecine Vétérinaire de Sidi Thabet, Université de La Manouba, Ariana, 2020, Tunisia
| | - Pierre Guy Marnet
- Département Productions animales, agroalimentaire, nutrition, service des sciences et productions animales, Institut Agro Rennes-Angers, 65 rue de St Brieuc, Rennes, 35000, France
- Laboratoire SELMET (Systèmes d'élevage méditerranéens et tropicaux), CIRAD/Inrae/Institut Agro, Campus international de Baillarguet, Montpellier Cedex 5, 34398, France
| |
Collapse
|
16
|
Mahieu G, Elens L, Prebonnaud N, Chauzy A, Van Bambeke F. A hollow fiber infection model to study intracellular and extracellular antibiotic activity against Staphylococcus aureus. iScience 2025; 28:112076. [PMID: 40124509 PMCID: PMC11930174 DOI: 10.1016/j.isci.2025.112076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/24/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Antibiotic activity against intracellular pathogens is commonly evaluated in static models that do not reproduce plasma concentration fluctuations. However, efficacy is influenced by exposure conditions, related to drug pharmacokinetic profile. This study developed and validated an intracellular pharmacodynamic model using the hollow fiber system, the gold standard for evaluating extracellular antibiotic activity. The activity of fluoroquinolones, i.e., bactericidal antibiotics with intracellular tropism, was studied against intracellular Staphylococcus aureus, involved in persistence/recurrence of infections. In this model, moxifloxacin was more effective than in static conditions (0.87 log10 killing gain), while ciprofloxacin kill rate was slower (18 vs. 12 h to achieve 1 log10 killing). These differences were linked to the Cmax/MIC ratio, which was 2.5-fold higher for moxifloxacin but 3.4-fold lower for ciprofloxacin in dynamic vs. static conditions. This model could be applied to other drugs, cell types, or pathogens, offering a tool for optimizing dosing schemes and considering intracellular reservoirs.
Collapse
Affiliation(s)
- Gwenaëlle Mahieu
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute (LDRI), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Integrated PharmacoMetrics, PharmacoGenomics and Pharmacokinetics (PMGK) Research Group, Louvain Drug Research Institute (LDRI), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Laure Elens
- Integrated PharmacoMetrics, PharmacoGenomics and Pharmacokinetics (PMGK) Research Group, Louvain Drug Research Institute (LDRI), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche expérimentale et clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Noémie Prebonnaud
- Université de Poitiers, INSERM U1070, PHAR2, 86000 Poitiers, France
- Laboratoire de Toxicologie-Pharmacocinétique, CHU de Poitiers, Poitiers, France
| | - Alexia Chauzy
- Université de Poitiers, INSERM U1070, PHAR2, 86000 Poitiers, France
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute (LDRI), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
17
|
Dong F, Wang G, Feng X, Gong C, Zheng Z, Chen Y, Huo Y, Zhang Y, Wang H. Investigation and Analysis of Staphylococcus aureus Contamination in Food in Yantai City, China: Based on a 14-Year Continuous Monitoring. Foodborne Pathog Dis 2025. [PMID: 39982751 DOI: 10.1089/fpd.2024.0175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025] Open
Abstract
Staphylococcus aureus is a foodborne zoonotic pathogen that threatens food safety and public health. However, few people have conducted long-term and systematic studies on S. aureus contamination in food in Yantai City. To investigate the contamination situation of S. aureus in food and improve the ability of early warning and control of foodborne diseases, a total of 2384 samples from 17 categories were collected from 13 monitoring points in Yantai City, from 2010 to 2023. Forty-four samples were positively detected for S. aureus, with a detection rate of 1.85% (44/2384). The detection rate of S. aureus was highest in Zhifu District (4.12%), followed by Penglai District (2.45%), Zhaoyuan District (2.37%), Kaifa District (2.19%), and Longkou District (1.98%). Positive detection rates were higher in frozen rice and flour products at 8.82% (6/68), quick-frozen dishes at 5.56% (1/18), aquatic products at 4.05% (3/74), and meat and meat products at 3.55% (27/760). Positive detection rates in samples from the first, second, third, and fourth quarters were 0% (0/44), 2.21% (20/906), 2.13% (22/1033), and 0.50% (2/401), respectively. Positive detection rates in bulk and prepackaged samples were 2.33% (36/1546) and 0.95% (8/838), respectively, with statistically significant differences (χ2 = 5.66, p < 0.05). Positive detection rates were significantly different for samples collected from different sampling stages, of which at production and processing stages was 7.78% (20/257), catering stages 1.38% (10/727), and distribution stages 1% (14/1400) (χ2 = 56.41, p < 0.05). Frozen rice and flour products, quick-frozen dishes, aquatic products, and meat and meat products are the main food products contaminated with S. aureus, and the resulting secondary contamination is a hidden danger for the occurrence of foodborne diseases, which should be given sufficient attention.
Collapse
Affiliation(s)
- Fengguang Dong
- Department of Nutrition and Food Hygiene, Yantai Center for Disease Control and Prevention, Yantai, China
| | - Guiqiang Wang
- Office of General Administration (Department of Audit), Yantai Center for Disease Control and Prevention, Yantai, China
| | - Xueying Feng
- Department of Nutrition and Food Hygiene, Yantai Center for Disease Control and Prevention, Yantai, China
| | - Chunbo Gong
- Department of Nutrition and Food Hygiene, Yantai Center for Disease Control and Prevention, Yantai, China
| | - Zhong Zheng
- Office of General Administration (Department of Audit), Yantai Center for Disease Control and Prevention, Yantai, China
| | - Youxia Chen
- Department of Nutrition and Food Hygiene, Yantai Center for Disease Control and Prevention, Yantai, China
| | - Yapeng Huo
- Department of Nutrition and Food Hygiene, Yantai Center for Disease Control and Prevention, Yantai, China
| | - Yiyi Zhang
- Department of Nutrition and Food Hygiene, Yantai Center for Disease Control and Prevention, Yantai, China
| | - Hongtao Wang
- Office of General Administration (Department of Audit), Yantai Center for Disease Control and Prevention, Yantai, China
| |
Collapse
|
18
|
Wang G, Wen J, Tian Z, Zhou H, Peng X, Zhang P, Li Z. Piceatannol and its analogues alleviate Staphylococcus aureus pathogenesis by targeting β-lactamase biofilms and α-hemolysin. Sci Rep 2025; 15:5551. [PMID: 39952994 PMCID: PMC11828952 DOI: 10.1038/s41598-025-89654-1] [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: 10/15/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
β-Lactamases, biofilms and toxins pose challenges for combating S. aureus infection. Thus, identifying inhibitors that can restore bacterial sensitivity to antibiotics, destroy biofilms, and antitoxins is a promising way to develop alternative agents. In this study, we found that piceatannol (pit), along with its analogues resveratrol (ret) and pterostilbene (pts) bind with β-lactamase to inhibit its activity, and 96TYR, 58ILE and 66LYS were identified as the critical binding residues. Pit and pts reduced the ampicillin (Amp) and gentamicin (Gm) MICs against S. aureus and enhanced the bactericidal ability of Amp. Pit and its analogues inhibited the formation of S. aureus USA300. In addition, the pit analogues bound with α-hemolysin and suppressed the hemolysis activity of the bacterial culture supernatant. The mechanism analysis revealed that pit exhibited multiple potential binding modes with α-hemolysin. Pit significantly decreased the cytotoxicity and the adherence effect mediated by S. aureus and increased the survival rate of Galleria mellonella that infected with S. aureus, the pathological tissue damage of Galleria mellonella was alleviated by treatment with pit alone or in combination with Amp. Taken together, our findings identify promising compounds for the development of S. aureus infection inhibitors.
Collapse
Affiliation(s)
- Guizhen Wang
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Jingyao Wen
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Zizeng Tian
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Hanbing Zhou
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Xinli Peng
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Peigang Zhang
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Zhandong Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China.
- Jilin Engineering Normal University, Changchun, 130052, China.
| |
Collapse
|
19
|
Lv H, Yang M, Yang Y, Tang Z, Guo Y, Zhou J, Gui Y, Huang R, Cai J, Yu B, Yang J, Bao Y, Zhang Z, Zhang D, Hou T. Metal Ion and Antibiotic Co-loaded Nanoparticles for Combating Methicillin-Rresistant Staphylococcus aureus-Induced Osteomyelitis. ACS NANO 2025; 19:5253-5268. [PMID: 39886847 DOI: 10.1021/acsnano.4c11956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) causes osteomyelitis (OM), which seriously threatens public health due to its antimicrobial resistance. To increase the sensitivity of antibiotics and eradicate intracellular bacteria, a Zn2+ and vancomycin (Van) codelivered nanotherapeutic (named Man-Zn2+/Van NPs) was fabricated and characterized via mannose (Man) modification. Man-Zn2+/Van NPs exhibit significant inhibitory activity against extra- and intracellular MRSA and obviously decrease the minimum inhibitory concentration of Van. Man-Zn2+/Van NPs can be easily internalized by MRSA-infected macrophages and significantly accumulated in infected bone via Man-mediated targeting. In vivo experiments in a mouse OM model verified that Man-Zn2+/Van NPs significantly reduce the extra- and intracellular MRSA burden, improve gait patterns, increase bone mass, and decrease inflammatory cytokine expression. The antibacterial mechanism of Man-Zn2+/Van NPs includes destruction of the MRSA membrane, degeneration of intracellular proteins and DNA, inhibition of MRSA glycolysis, and intervention in the energy metabolism of bacteria. Overall, this metal-antibiotic nanotherapeutics strategy provides new insight for combating extra- and intracellular infections caused by MRSA-induced OM.
Collapse
Affiliation(s)
- Hui Lv
- Department of Emergency and trauma orthopedics, the 958th Hospital of Chinese People's Liberation Army, Army Medical University (Third Military Medical University), Chongqing 400023, China
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ming Yang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yusheng Yang
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhenzhen Tang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuan Guo
- Department of Emergency and trauma orthopedics, the 958th Hospital of Chinese People's Liberation Army, Army Medical University (Third Military Medical University), Chongqing 400023, China
| | - Jiangling Zhou
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yingtao Gui
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Rong Huang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Juan Cai
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Bo Yu
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jing Yang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ying Bao
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhongrong Zhang
- Department of Emergency and trauma orthopedics, the 958th Hospital of Chinese People's Liberation Army, Army Medical University (Third Military Medical University), Chongqing 400023, China
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Tianyong Hou
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
20
|
Almatroudi A. Biofilm Resilience: Molecular Mechanisms Driving Antibiotic Resistance in Clinical Contexts. BIOLOGY 2025; 14:165. [PMID: 40001933 PMCID: PMC11852148 DOI: 10.3390/biology14020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025]
Abstract
Healthcare-associated infections pose a significant global health challenge, negatively impacting patient outcomes and burdening healthcare systems. A major contributing factor to healthcare-associated infections is the formation of biofilms, structured microbial communities encased in a self-produced extracellular polymeric substance matrix. Biofilms are critical in disease etiology and antibiotic resistance, complicating treatment and infection control efforts. Their inherent resistance mechanisms enable them to withstand antibiotic therapies, leading to recurrent infections and increased morbidity. This review explores the development of biofilms and their dual roles in health and disease. It highlights the structural and protective functions of the EPS matrix, which shields microbial populations from immune responses and antimicrobial agents. Key molecular mechanisms of biofilm resistance, including restricted antibiotic penetration, persister cell dormancy, and genetic adaptations, are identified as significant barriers to effective management. Biofilms are implicated in various clinical contexts, including chronic wounds, medical device-associated infections, oral health complications, and surgical site infections. Their prevalence in hospital environments exacerbates infection control challenges and underscores the urgent need for innovative preventive and therapeutic strategies. This review evaluates cutting-edge approaches such as DNase-mediated biofilm disruption, RNAIII-inhibiting peptides, DNABII proteins, bacteriophage therapies, antimicrobial peptides, nanoparticle-based solutions, antimicrobial coatings, and antimicrobial lock therapies. It also examines critical challenges associated with biofilm-related healthcare-associated infections, including diagnostic difficulties, disinfectant resistance, and economic implications. This review emphasizes the need for a multidisciplinary approach and underscores the importance of understanding biofilm dynamics, their role in disease pathogenesis, and the advancements in therapeutic strategies to combat biofilm-associated infections effectively in clinical settings. These insights aim to enhance treatment outcomes and reduce the burden of biofilm-related diseases.
Collapse
Affiliation(s)
- Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
21
|
Yang Y, Zhou H, Li F, Zhang Y, Yang J, Shen Y, Hu N, Zou Q, Qin L, Zeng H, Huang W. Staphylococcus aureus induces mitophagy via the HDAC11/IL10 pathway to sustain intracellular survival. J Transl Med 2025; 23:156. [PMID: 39905391 PMCID: PMC11796158 DOI: 10.1186/s12967-025-06161-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND The immune evasion and prolonged survival of Staphylococcus aureus (S. aureus) within macrophages are key factors contributing to the difficulty in curing osteomyelitis. Although macrophages play a vital role as innate immune cells, the mechanisms by which S. aureus survives within them and suppresses host immune functions remain incompletely understood. METHODS This study employed confocal microscopy, flow cytometry, ELISA, and siRNA technology to assess the survival capacity of S. aureus within macrophages and the impact of inflammatory cytokines on its persistence. Proteomics was used to investigate the potential mechanisms and differential proteins involved in S. aureus intracellular survival. Additionally, confocal microscopy, flow cytometry, Mdivi-1 intervention, and Western blot were utilized to validate the role of mitophagy in supporting S. aureus survival. The study further explored how the HDAC11/IL10 axis enhances mitophagy to promote intracellular S. aureus survival by using HDAC11 overexpression, siRNA, and rapamycin intervention combined with confocal microscopy and flow cytometry. RESULTS The findings demonstrated that IL10 promotes mitophagy to clear mitochondrial reactive oxygen species (mtROS), thereby enhancing the intracellular survival of S. aureus within macrophages. Additionally, we discovered that the transcriptional repressor of IL10, HDAC11, was significantly downregulated during S. aureus infection. Overexpression of HDAC11 and the use of the autophagy activator rapamycin further validated that the HDAC11/IL10 axis regulates mitophagy via the mTOR pathway, which is essential for supporting S. aureus intracellular survival. CONCLUSION This study reveals that S. aureus enhances IL10 production by inhibiting HDAC11, thereby promoting mitophagy and mtROS clearance, which supports its survival within macrophages. These findings offer new insights into the intracellular survival mechanisms of S. aureus and provide potential therapeutic approaches for the clinical management of osteomyelitis.
Collapse
Affiliation(s)
- Yaji Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Haotian Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Feilong Li
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
- Department of Orthopaedics, The People's Hospital of Dazu, Chongqing, 402360, China
| | - Yanhao Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Jianye Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Yidong Shen
- Department of Orthopaedics, The First people's Hospital of Yancheng, Yancheng, Jiangsu, 224006, China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Leilei Qin
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China.
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
22
|
Yang M, Chao H, Hou Z, Wang L, Xu W, Zhao X. Antimicrobial activity of octyl gallate nanoemulsion combined with photodynamic technology and its effect on food preservation. Int J Food Microbiol 2025; 429:111023. [PMID: 39693859 DOI: 10.1016/j.ijfoodmicro.2024.111023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
Photodynamic inactivation, as a safe and effective antimicrobial technology that does not damage the organoleptic properties of the food itself, decreases the use of preservatives and is gradually gaining attention in the food industry. This study selected octyl gallate (OG) as an antimicrobial photosensitizer with eucalyptus oil as the oil phase and prepared it as an octyl gallate nanoemulsion (OG-NE) to ensure the delivery of the photosensitizer. Escherichia coli and Staphylococcus aureus inactivation with the OG-NE combined with photodynamic technology, as well as the effect on the quality of food products, was investigated. The results showed the successful preparation and homogeneous distribution of the OG-NE with an encapsulation rate of 85.18 %. The OG-NE's ability to produce single oxygen (1O2) was significantly higher, as shown by 1O2 production. The OG-NE combined photodynamic technique confirmed the effectiveness of microbial removal, demonstrating a significant increase in reactive oxygen species (ROS) and the permeability of the cell membrane. The effect of the OG-NE combined photodynamic technology on perch (microbiology, pH, whiteness, water holding capacity, TVB-N and TBA) and litchi (weight loss, titratable acid and sugar content) preservation was assessed. Food preservation experiments revealed that the OG-NE combined photodynamic technology exhibited a positive effect on food quality. The results indicated that the combination of the OG-NE and photodynamic technology provided a new alternative strategy for the food industry in antimicrobial and preservation.
Collapse
Affiliation(s)
- Ming Yang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huijing Chao
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zihan Hou
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lingling Wang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Weizhuo Xu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xu Zhao
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
23
|
Ip TKY, Wang Y, Wang S, Pu K, Wang R, Han B, Gao P, Xie Y, Kao RY, Ho PL, Li H, Sun H. Hinokitiol potentiates antimicrobial activity of bismuth drugs: a combination therapy for overcoming antimicrobial resistance. RSC Med Chem 2025:d4md00860j. [PMID: 40027343 PMCID: PMC11865920 DOI: 10.1039/d4md00860j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025] Open
Abstract
Antimicrobial resistance (AMR) poses a significant global health threat, rendering many infections untreatable. To combat AMR, repurposing approved drugs has emerged as a cost-effective strategy. Bismuth drugs, when combined with antibiotics, have been proven to be effective against Helicobacter pylori, including antibiotic-resistant strains. However, bismuth drugs alone exhibit limited antimicrobial activity against a narrow spectrum of pathogens. Therefore, a novel approach to enhance the efficacy and broaden the antimicrobial spectrum of bismuth drugs is highly desirable. Herein, we show that a naturally occurring monoterpenoid, hinokitiol, could potentiate the antimicrobial activity of bismuth drugs. We demonstrate a strong synergy between hinokitiol and colloidal bismuth subcitrate (CBS) against various Gram-positive and Gram-negative bacterial strains, including methicillin-resistant Staphylococcus aureus (MRSA). Moreover, the combination of hinokitiol and CBS exhibits anti-biofilm activity by preventing biofilm formation and eliminating S. aureus persister cells. Importantly, the combination therapy demonstrates promising antimicrobial efficacy in murine infection models including skin wound, gastrointestinal and blood infections. Mechanistic studies reveal that hinokitiol enhances bismuth ion (Bi(iii)) accumulation and reduces intracellular iron levels. By using thermal proteome profiling combined with dynamic quantitative proteomics analysis, we demonstrate that the bismuth-hinokitiol combination propagated the bismuth binding and interfered with ribosome synthesis, the glycolysis process, impaired bacterial cell wall synthesis and pathogenesis in MRSA. Our finding highlights the potential of combinatorial hinokitiol and bismuth drugs in the fight against AMR.
Collapse
Affiliation(s)
- Tiffany Ka-Yan Ip
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Yuchuan Wang
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
- School of Chemistry, Sun Yat-Sen University Guangzhou P. R. China
| | - Suyu Wang
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Keyuan Pu
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Runming Wang
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Bingjie Han
- School of Chemistry, Sun Yat-Sen University Guangzhou P. R. China
| | - Peng Gao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong Pokfulam Hong Kong SAR P. R. China
| | - Yanxuan Xie
- School of Chemistry, Sun Yat-Sen University Guangzhou P. R. China
| | - Richard Y Kao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong Pokfulam Hong Kong SAR P. R. China
| | - Pak-Leung Ho
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong Pokfulam Hong Kong SAR P. R. China
| | - Hongyan Li
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Hongzhe Sun
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| |
Collapse
|
24
|
Song L, Schwinn LS, Barthel J, Ketter V, Lechler P, Linne U, Rastan AJ, Vogt S, Ruchholtz S, Paletta JRJ, Günther M. Implant-Derived S. aureus Isolates Drive Strain-Specific Invasion Dynamics and Bioenergetic Alterations in Osteoblasts. Antibiotics (Basel) 2025; 14:119. [PMID: 40001363 PMCID: PMC11852183 DOI: 10.3390/antibiotics14020119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Implants are integral to modern orthopedic surgery. The outcomes are good, but infections remain a serious issue. Staphylococcus aureus (S. aureus), along with Staphylococcus epidermidis, are predominant pathogens responsible for implant-associated infections, as conventional antibiotic treatments often fail due to biofilm formation or the pathogens' ability to invade cells and to persist intracellularly. Objectives: This study therefore focused on interactions of S. aureus isolates from infected implants with MG63 and SaOS2 osteoblasts by investigating the adhesion, invasion, and the impact on the bioenergetics of osteoblasts. Methods and Results: We found that the ability of S. aureus to adhere to osteoblasts depends on the isolate and was not associated with a single gene or expression pattern of characteristic adhesion proteins, and further, was not correlated with invasion. However, analysis of invasion capabilities identified better invasion conditions for S. aureus isolates with the SaOS2 osteoblastic cells. Interestingly, metabolic activity of osteoblasts remained unaffected by S. aureus infection, indicating cell survival. In contrast, respiration assays revealed an altered mitochondrial bioenergetic turnover in infected cells. While basal as well as maximal respiration in MG63 osteoblasts were not influenced statistically by S. aureus infections, we found increased non-mitochondrial respiration and enhanced glycolytic activity in the osteoblasts, which was again, more pronounced in the SaOS2 osteoblastic cells. Conclusions: Our findings highlight the complexity of S. aureus-host interactions, where both the pathogen and the host cell contribute to intracellular persistence and survival, representing a major factor for therapeutic failures.
Collapse
Affiliation(s)
- Lei Song
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Lea-Sophie Schwinn
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Juliane Barthel
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Vanessa Ketter
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Philipp Lechler
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Uwe Linne
- Faculty of Chemistry, Philipps-University Marburg, 35032 Marburg, Germany
| | - Ardawan J. Rastan
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Sebastian Vogt
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Steffen Ruchholtz
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Jürgen R. J. Paletta
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Madeline Günther
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| |
Collapse
|
25
|
Gong JY, Li Y, Wang P, Peng X, Xie R, Wang W, Liu Z, Pan DW, Ju XJ, Chu LY. K +-Responsive Nanoparticles with Charge Reversal and Gating Synergistic Effects for Targeted Intracellular Bacteria Eradication. ACS NANO 2025; 19:2345-2361. [PMID: 39772435 DOI: 10.1021/acsnano.4c12542] [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: 01/11/2025]
Abstract
Intracellular bacteria can evade the attack of the immune system and the bactericidal effects of most antibiotics due to the protective effect of the host cells. Herein, inspired by the stimuli-responsive behaviors of biological ion channels, a kind of synergistic cascade potassium ion (K+)-responsive nanoparticles gated with K+-responsive polymers is ingeniously designed to target intracellular bacteria and then control drug release. Due to the cooperative interaction of host-guest complexation and conformational transition of K+-responsive polymers, the grafted gates based on these polymers could recognize high K+ concentration to reverse the negatively charged nanoparticles into positively charged ones for targeting bacteria and subsequently inducing a switch from the hydrophobic shrinking "off" state to the hydrophilic stretching "on" state for drug release. The K+-responsive nanoparticles can effectively load antibiotics and be endocytosed into the infected cells, and K+-responsive gates can be actuated by a high intracellular K+ concentration. With the efficient synergistic cascade strategy, these K+-responsive nanocarriers can deliver antibiotics to the subcellular region where intracellular bacteria reside and show superior elimination efficiencies in vitro and in vivo than the free drug in delivering vancomycin. The K+-responsive nanocarriers are expected to improve the bioavailability of drugs and enhance their antibacterial efficacy.
Collapse
Affiliation(s)
- Jue-Ying Gong
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yao Li
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Po Wang
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xu Peng
- Experimental and Research Animal Institute, Chengdu, Sichuan 610065, China
| | - Rui Xie
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
- State Key Laboratory of Polymer Materials Engineering, Chengdu, Sichuan 610065, China
| | - Wei Wang
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
- State Key Laboratory of Polymer Materials Engineering, Chengdu, Sichuan 610065, China
| | - Zhuang Liu
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
- State Key Laboratory of Polymer Materials Engineering, Chengdu, Sichuan 610065, China
| | - Da-Wei Pan
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiao-Jie Ju
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
- State Key Laboratory of Polymer Materials Engineering, Chengdu, Sichuan 610065, China
| | - Liang-Yin Chu
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
- State Key Laboratory of Polymer Materials Engineering, Chengdu, Sichuan 610065, China
| |
Collapse
|
26
|
Yang X, Tang X, Yi S, Guo T, Liao Y, Wang Y, Zhang X. Maltodextrin-derived nanoparticles resensitize intracellular dormant Staphylococcus aureus to rifampicin. Carbohydr Polym 2025; 348:122843. [PMID: 39562116 DOI: 10.1016/j.carbpol.2024.122843] [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: 07/17/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 11/21/2024]
Abstract
Intracellular bacteria are recognized as a crucial factor in the persistence and recurrence of infections. The efficacy of current antibiotic treatments faces substantial challenges due to the dormant state formation of intracellular bacteria. In this study, we devised a strategy aimed at reverting intracellular dormant bacteria to a metabolically active state, thereby increasing their vulnerability to antibiotics. We found that oligosaccharides, especially maltodextrin (MD), can be absorbed by dormant S. aureus, leading to their revival and restoration of sensitivity to rifampicin (Rif). We then synthesized a reactive oxygen species (ROS)-responsive MD-prodrug by covalently binding MD with 4-(hydroxymethyl) phenylboronic acid pinacol ester (MD-PBAP) and prepared a ROS-responsive nanoparticles (MDNP) using a nanoprecipitation and self-assembly method. Once internalized by host cells, MDNP was degraded to MD, reactivating dormant S. aureus, and enhancing their susceptibility to Rif. More importantly, MDNP treatment restored the sensitivity of intracellular persistent S. aureus to Rif in both a reservoir transfer model and whole-body infection model. Additionally, MDNP have demonstrated excellent biocompatibility in both in vitro and in vivo settings. These results offer a promising therapeutic avenue for managing persistent intracellular bacterial infections by reviving and resensitizing intracellular dormant bacteria to conventional antibiotics.
Collapse
Affiliation(s)
- Xiaodi Yang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Xiyu Tang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Sisi Yi
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Tao Guo
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Yue Liao
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Yan Wang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Xiangjun Zhang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China.
| |
Collapse
|
27
|
Szymczak K, Rychłowski M, Zhang L, Nakonieczna J. Harnessing light-activated gallium porphyrins to combat intracellular Staphylococcus aureus using an in vitro keratinocyte infection model. Sci Rep 2025; 15:1295. [PMID: 39779728 PMCID: PMC11711192 DOI: 10.1038/s41598-024-84312-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Staphylococcus aureus (S. aureus) can survive inside nonprofessional phagocytes such as keratinocytes, enabling it to evade antibiotics and cause recurrent infections once treatment stops. New antibacterial strategies to eliminate intracellular, multidrug-resistant bacteria are needed. This study used a keratinocyte model infected with methicillin-resistant S. aureus (MRSA) to test light-activated compounds, specifically heme-mimetic gallium (III) porphyrin (Ga3+CHP) and visible light, known as antimicrobial photodynamic inactivation (aPDI), for eliminating intracellular MRSA. Ga3+CHP was found to accumulate more in infected cells, particularly within lysosomal structures where MRSA resides. Flow cytometry and fluorescence microscopy revealed significant colocalization of MRSA and Ga3+CHP. Under aPDI, MRSA showed reduced adhesion to host cells and a 70% reduction in the GFP signal from intracellular bacteria. Additionally, light-activated Ga3+CHP significantly decreased the number of extracellular bacteria, reducing the potential for further infection. This study is the first to analyze aPDI toxicity in real time within an infection model, demonstrating that this method is neither cytotoxic nor phototoxic.
Collapse
Affiliation(s)
- Klaudia Szymczak
- Laboratory of Photobiology and Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland
| | - Michał Rychłowski
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland
| | - Lei Zhang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin, China
| | - Joanna Nakonieczna
- Laboratory of Photobiology and Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland.
| |
Collapse
|
28
|
Sargen MR, Helaine S. A prophage competition element protects Salmonella from lysis. Cell Host Microbe 2024; 32:2063-2079.e8. [PMID: 39515326 PMCID: PMC11840918 DOI: 10.1016/j.chom.2024.10.012] [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: 06/27/2024] [Revised: 09/11/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Most bacteria are polylysogens that carry multiple prophages integrated into the chromosome. These prophages confer advantages to their bacterial host, yet also pose a lethal threat as they can reactivate and enter a lytic cycle. DNA damage of the bacterial host is a common trigger of prophage lytic cycles. Because DNA damage is frequently experienced by bacterial pathogens exposed to host immune defenses, prophage activation may be common during infection. Investigating the consequences of prophage induction in Salmonella, we discover a prophage competition element in the Gifsy-1 prophage that we name ribonuclease effector module with ATPase, inhibitor, and nuclease (RemAIN) because it blocks the lytic cycles and release of viral particles of co-resident prophages. Intramacrophage Salmonella persisters, a subpopulation that incurs DNA damage, experience prophage reactivation and subsequent RemAIN activation, which influences Salmonella persisters and macrophage response to infection. Our findings reveal a multi-layered host-pathogen arms race in which prophage-prophage competition influences bacterial persistence and the mammalian immune response.
Collapse
Affiliation(s)
- Molly R Sargen
- Department of Microbiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Sophie Helaine
- Department of Microbiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
29
|
Batchelder JI, Taylor AJ, Mok WWK. Metabolites augment oxidative stress to sensitize antibiotic-tolerant Staphylococcus aureus to fluoroquinolones. mBio 2024; 15:e0271424. [PMID: 39475229 PMCID: PMC11633220 DOI: 10.1128/mbio.02714-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/08/2024] [Indexed: 12/12/2024] Open
Abstract
If left unchecked, infections involving antibiotic-refractory bacteria are expected to cause millions of deaths per year in the coming decades. Beyond genetically resistant bacteria, persisters, which are genetically susceptible cells that survive antibiotic doses that kill the rest of the clonal population, can potentially contribute to treatment failure and infection relapse. Stationary-phase bacterial cultures are enriched with persisters, and it has been shown that stimulating these populations with exogenous nutrients can reduce persistence to different classes of antibiotics, including topoisomerase-targeting fluoroquinolones (FQs). In this study, we show that adding glucose and amino acids to nutrient-starved Staphylococcus aureus cultures enhanced their sensitivity to FQs, including delafloxacin (Dela)-a drug that was recently approved for treating staphylococcal infections. We found that while the added nutrients increased nucleic acid synthesis, this increase was not required to sensitize S. aureus to FQs. We further demonstrate that addition of these nutrients increases membrane potential and the ability to generate harmful reactive oxygen species (ROS) during FQ treatment. Chelating iron, scavenging hydroxyl radicals, and limiting oxygenation during FQ treatment and during recovery following FQ treatment rescued nutrient-stimulated S. aureus. In all, our data suggest that while nutrient stimulation increases the activity of FQ targets in stationary-phase S. aureus, the resulting generation of ROS, presumably made possible through metabolic upregulation, is the primary driver of increased sensitivity to these drugs.IMPORTANCEStaphylococcus aureus causes many chronic and relapsing infections because of its ability to endure host immunity and antibiotic therapy. While several studies have focused on the nutrient requirements for the formation and maintenance of staphylococcal infections, the effects of the nutrient environment on bacterial responses to antibiotic treatment remain understudied. Here, we show that adding nutrients to starved S. aureus activates biosynthetic processes, including DNA synthesis, but it is the generation of harmful reactive oxidants that sensitizes S. aureus to DNA topoisomerase-targeting FQs. Our results suggest that the development of approaches aimed at perturbing metabolism and increasing oxidative stress can potentiate the bactericidal activity of FQs against antibiotic-tolerant S. aureus.
Collapse
Affiliation(s)
- Jonathan I. Batchelder
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, Connecticut, USA
| | - Andrew J. Taylor
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Wendy W. K. Mok
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
30
|
Bakhti S, Ahmadi MH, Owlia P. Inducing programmed cell death trough MazEF system to combat Staphylococcus aureus: A non-antibiotic treatment candidate. PLoS One 2024; 19:e0314867. [PMID: 39637174 PMCID: PMC11620692 DOI: 10.1371/journal.pone.0314867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
The aim of the present study was to evaluate the role of extracellular death factor (EDF) derived from Escherichia coli in the induction of programmed cell death (PCD) in methicillin-resistant and -susceptible Staphylococcus aureus (MRSA and MSSA). The confirmation of bacterial strains as well as the minimum inhibitory concentration (MIC) test were performed according to CLSI, 2022. The extraction and efficacy determination of EDF as well as the CFU assessment were done. The expression of mazE and mazF gens in different conditions was evaluated by Real-time PCR. The likely formation of persister cells from MRSA and MSSA, and the possible synthesis of EDF in old cultures of these pathogens was evaluated, as well. The combination of EDF of two E. coli strains and sub-MIC rifampin reduced the CFUs of MRSA and MSSA strains in mid-logarithmic growth phase while increased the expression of mazF several times more than mazE gene. The expression of these genes in different conditions were unlike. EDF was produced in the old cultures of MRSA and MSSA. The supernatant of E. coli 25922 was more powerful than the clinical strain ones to decrease the CFUs of the MRSA and MSSA. The EDF derived from E. coli in combination with sub-MIC rifampin could induce PCD in MRSA and MSSA through activation of the MazEF system. This phenomenon could be exploited as a non-antibiotic treatment candidate to combat the infections caused by the antibiotic-resistant pathogens. However, more studies should be performed in this regard.
Collapse
Affiliation(s)
- Shahriar Bakhti
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | | | - Parviz Owlia
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
31
|
Zhang L, Zhang Y, Tian L, Shen Q, Ma X. Doxifluridine effectively kills antibiotic-resistant Staphylococcus aureus in chronic obstructive pulmonary disease. Microbiol Spectr 2024; 12:e0180524. [PMID: 39530670 PMCID: PMC11619241 DOI: 10.1128/spectrum.01805-24] [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: 07/19/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading cause of morbidity and mortality globally, often exacerbated by infections such as methicillin-resistant Staphylococcus aureus (MRSA). The rise in antibiotic-resistant strains complicates treatment and underscores the need for novel therapeutic drugs. In this paper, we further investigated the antimicrobial potential of a fluoropyrimidine anticancer drug doxifluridine against multidrug-resistant S. aureus. Determination of minimum inhibitory concentration (MIC) or minimum bactericidal concentration (MBC), monitoring of growth curve, time-kill assays, biofilm bactericidal assays, and chequerboard studies were conducted to evaluate the antibacterial efficacy of doxifluridine. Safety was assessed via hemolysis and cytotoxicity assays, and an in vivo Galleria mellonella larvae model was employed to test protective effects. Doxifluridine demonstrated significant antibacterial activity against clinical multidrug resistance (MDR) S. aureus isolates, with MIC and MBC values ranging from 0.5 to 2 µg/mL and 1 to 4 µg/mL, respectively. The results revealed doxifluridine's potent bactericidal effects within 8 hours. Moreover, doxifluridine-treated bacteria showed a substantial reduction in biofilm mass and viability. Furthermore, chequerboard assays indicated synergistic interactions between doxifluridine and other antibiotics, reducing MIC values by two- to eightfold. More importantly, safety evaluations confirmed that doxifluridine did not exhibit hemolytic toxicity or cytotoxicity. Finally, doxifluridine significantly increased the survival rate of MRSA-infected G. mellonella larvae in vivo. In brief, doxifluridine exhibited promising in vitro and in vivo antibacterial activity against MRSA, suggesting its potential as a repurposed drug for treating resistant bacterial infections in COPD patients.IMPORTANCEThe study provides robust evidence for the antibacterial efficacy of doxifluridine against Methicillin-resistant Staphylococcus aureus in chronic obstructive pulmonary disease (COPD) patients. Its rapid action, ability to disrupt biofilms, and synergistic effects with other antibiotics, combined with a favorable safety profile, highlight its potential as a repurposed therapeutic agent. Future clinical trials will be essential to confirm these findings and pave the way for its integration into clinical practice. This work not only provides candidate for tackling the management of bacterial infections in COPD but also exemplifies the potential of drug repurposing in combating antibiotic-resistant infections.
Collapse
Affiliation(s)
- Lianshen Zhang
- Respiratory and Critical Care Medicine Department, Tongxiang Second People’s Hospital, Tongxiang, Zhejiang, China
| | - Yingzhang Zhang
- Respiratory and Critical Care Medicine Department, Tongxiang Second People’s Hospital, Tongxiang, Zhejiang, China
| | - Lijie Tian
- General Clinic, Chongfu Town Community Health Service Center, Tongxiang, Zhejiang, China
| | - Qiang Shen
- Respiratory and Critical Care Medicine Department, Tongxiang Second People’s Hospital, Tongxiang, Zhejiang, China
| | - Xiaolong Ma
- Department of Respiratory, The Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
32
|
Tooke AK, Hodges RE, Pyrah JF, Bayles KW, Renshaw SA, Foster SJ. Tetracycline and Oxacillin Act Synergistically on Biofilms and Display Increased Efficacy In Vivo Against Staphylococcus aureus. Curr Microbiol 2024; 81:447. [PMID: 39505760 PMCID: PMC11541413 DOI: 10.1007/s00284-024-03959-4] [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: 07/04/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024]
Abstract
Oxacillin (bactericidal) and tetracycline (bacteriostatic) are clinically relevant antibiotics that are routinely prescribed to treat Staphylococcus aureus infections but not conventionally used in combination. There is an urgent need for treatment regimens that can act upon biofilms during infection, associated with chronic infections on indwelling devices, as well as acute planktonic (systemic) infection. Here we show that in an in vitro model oxacillin and tetracycline act synergistically against S. aureus UAMS-1 biofilms, reducing the concentration of both antibiotics necessary to eradicate an established biofilm. Using an in vivo zebrafish larval infection model with S. aureus NewHG, they display improved bacterial clearance compared to each drug alone and can counteract a loss of host phagocytes, an important innate defence against S. aureus. In these cases, the bacteriostatic nature of tetracycline enhances rather than dampens the bactericidal action of oxacillin, although an exact mechanism remains to be elucidated. We suggest a dual therapy could be of clinical use against biofilm-forming S. aureus and has a potential use in patients with a compromised immune system.
Collapse
Affiliation(s)
- Amy K Tooke
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK
- Microbes in Health and Disease Theme, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Rebecca E Hodges
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Josie F Pyrah
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK
| | - Kenneth W Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Stephen A Renshaw
- The Bateson Centre, University of Sheffield, Western Bank, Sheffield, S10 2TH, UK
- School of Medicine and Population Health, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Simon J Foster
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK.
- Florey Institute, University of Sheffield, Sheffield, S10 2TH, UK.
| |
Collapse
|
33
|
Xiong Y, Wang R, Zheng J, Fang D, He P, Liu S, Lin Z, Chen X, Chen C, Shang Y, Yu Z, Liu X, Han S. Discovery of novel dihydropyrrolidone-thiadiazole compound crosstalk between the YycG/F two-component regulatory pathway and cell membrane homeostasis to combat methicillin-resistant Staphylococcus aureus. Eur J Med Chem 2024; 277:116770. [PMID: 39208742 DOI: 10.1016/j.ejmech.2024.116770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/30/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024]
Abstract
The rapid emergence and spread of multidrug-resistant (MDR) Gram-positive pathogens present a significant challenge to global healthcare. Methicillin-resistant Staphylococcus aureus (MRSA) is a particular concern because of its high resistance to most antibiotics. Based on our previously reported chemical structure of compound 62, a series of novel derivatives were synthesized and evaluated for their antibacterial activities. We found that some of these derivatives displayed effective antibacterial activity against Gram-positive pathogens, with minimal cytotoxicity (CC50>100 μM) and hemolytic activity (HC50>200 μM). Among these derivatives, the minimum inhibitory concentration (MIC) of 62-7c against Gram-positive bacterial isolates ranged from 6.25 to 25 μM. This derivative also exhibited significant synergistic antibacterial effects with daptomycin both in vitro and in vivo, with an ability to eradicate planktonic and persister cells of MRSA. Additionally, 62-7c inhibited biofilm formation and eradicated mature biofilms of MRSA. Mechanistic studies revealed that 62-7c inhibited the YycG kinase activity and disrupted the cell membrane by binding to cardiolipin (CL), leading to cell death. Importantly, no development of drug resistance was observed even after 20 serial passages. Furthermore, 62-7c exhibited high biosafety and potent effectiveness in combating infections in both mouse pneumonia and mouse wound models infected with MRSA. Thus, our study revealed that 62-7c has the potential to serve as a novel antibacterial agent for treating MRSA infections.
Collapse
Affiliation(s)
- Yanpeng Xiong
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China; Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Ruian Wang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Jiaoyang Zheng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Di Fang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Peikun He
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Shanghong Liu
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Zhiwei Lin
- Laboratory of Respiratory Disease, People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Xuecheng Chen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Chengchun Chen
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Yongpeng Shang
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Zhijian Yu
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China.
| | - Xiaoju Liu
- Department of Infectious Diseases, Shenzhen Nanshan People's Hospital, Shenzhen University Medical School, Shenzhen 518052, China.
| | - Shiqing Han
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
34
|
Wan Y, Zheng J, Chan EW, Chen S. Proton motive force and antibiotic tolerance in bacteria. Microb Biotechnol 2024; 17:e70042. [PMID: 39487809 PMCID: PMC11531170 DOI: 10.1111/1751-7915.70042] [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: 04/24/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
Bacterial antibiotic tolerance is a decades-old phenomenon in which a bacterial sub-population, commonly known as persisters, does not respond to antibiotics and remains viable upon prolonged antimicrobial treatment. Persisters are detectable in populations of bacterial strains that are not antibiotic-resistant and are known to be responsible for treatment failure and the occurrence of chronic and recurrent infection. The clinical significance of antibiotic tolerance is increasingly being recognized and comparable to antibiotic resistance. To eradicate persisters, it is necessary to understand the cellular mechanisms underlying tolerance development. Previous works showed that bacterial antibiotic tolerance was attributed to the reduction in metabolic activities and activation of the stringent response, SOS response and the toxin-antitoxin system which down-regulates transcription functions. The latest research findings, however, showed that decreased metabolic activities alone do not confer a long-lasting tolerance phenotype in persisters, and that active defence mechanisms such as efflux and DNA repair are required for the long-term maintenance of phenotypic tolerance. As such active tolerance-maintenance mechanisms are energy-demanding, persisters need to generate and maintain the transmembrane proton motive force (PMF) for oxidative phosphorylation. This minireview summarizes the current understanding of cellular mechanisms essential for prolonged expression of phenotypic antibiotic tolerance in bacteria, with an emphasis on the importance of generation and maintenance of PMF in enabling proper functioning of the active tolerance mechanisms in persisters. How such mechanisms can be utilized as targets for the development of anti-persister strategies will be discussed.
Collapse
Affiliation(s)
- Yingkun Wan
- State Key Lab of Chemical Biology and Drug Discovery and the Department of Food Science and NutritionThe Hong Kong Polytechnic UniversityKowloonHong Kong
- Shenzhen Key Lab of Food Microbial Safety ControlThe Hong Kong Polytechnic University Shenzhen Research InstituteShenzhenChina
| | - Jiaqi Zheng
- State Key Lab of Chemical Biology and Drug Discovery and the Department of Food Science and NutritionThe Hong Kong Polytechnic UniversityKowloonHong Kong
| | - Edward Wai‐Chi Chan
- State Key Lab of Chemical Biology and Drug Discovery and the Department of Food Science and NutritionThe Hong Kong Polytechnic UniversityKowloonHong Kong
| | - Sheng Chen
- State Key Lab of Chemical Biology and Drug Discovery and the Department of Food Science and NutritionThe Hong Kong Polytechnic UniversityKowloonHong Kong
- Shenzhen Key Lab of Food Microbial Safety ControlThe Hong Kong Polytechnic University Shenzhen Research InstituteShenzhenChina
| |
Collapse
|
35
|
Sherry J, Rego EH. Phenotypic Heterogeneity in Pathogens. Annu Rev Genet 2024; 58:183-209. [PMID: 39083846 DOI: 10.1146/annurev-genet-111523-102459] [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: 08/02/2024]
Abstract
Pathogen diversity within an infected organism has traditionally been explored through the lens of genetic heterogeneity. Hallmark studies have characterized how genetic diversity within pathogen subpopulations contributes to treatment escape and infectious disease progression. However, recent studies have begun to reveal the mechanisms by which phenotypic heterogeneity is established within genetically identical populations of invading pathogens. Furthermore, exciting new work highlights how these phenotypically heterogeneous subpopulations contribute to a pathogen population better equipped to handle the complex and fluctuating environment of a host organism. In this review, we focus on how bacterial pathogens, including Staphylococcus aureus, Salmonella typhimurium, Pseudomonas aeruginosa, and Mycobacterium tuberculosis, establish and maintain phenotypic heterogeneity, and we explore recent work demonstrating causative links between this heterogeneity and infection outcome.
Collapse
Affiliation(s)
- Jessica Sherry
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA; ,
| | - E Hesper Rego
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA; ,
| |
Collapse
|
36
|
Qin L, Hu N, Zhang Y, Yang J, Zhao L, Zhang X, Yang Y, Zhang J, Zou Y, Wei K, Zhao C, Li Y, Zeng H, Huang W, Zou Q. Antibody-antibiotic conjugate targeted therapy for orthopedic implant-associated intracellular S. aureus infections. J Adv Res 2024; 65:239-255. [PMID: 38048846 PMCID: PMC11519013 DOI: 10.1016/j.jare.2023.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/07/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023] Open
Abstract
INTRODUCTION Treating orthopedic implant-associated infections, especially those caused by Staphylococcus aureus (S. aureus), remains a significant challenge. S. aureus has the ability to invade host cells, enabling it to evade both antibiotics and immune responses during infection, which may result in clinical treatment failures. Therefore, it is critical to identify the host cell type of implant-associated intracellular S. aureus infections and to develop a strategy for highly targeted delivery of antibiotics to the host cells. OBJECTIVES Introduced an antibody-antibiotic conjugate (AAC) for the targeted elimination of intracellular S. aureus. METHODS The AAC comprises of a human monoclonal antibody (M0662) directly recognizes the surface antigen of S. aureus, Staphylococcus protein A, which is conjugated with vancomycin through cathepsin-sensitive linkers that are cleavable in the proteolytic environment of the intracellular phagolysosome. AAC, vancomycin and vancomycin combined with AAC were used in vitro intracellular infection and mice implant infection models. We then tested the effect of AAC in vivo and in vivo by fluorescence imaging, in vivo imaging, bacterial quantitative analysis and bacterial biofilm imaging. RESULTS In vitro, it was observed that AAC captured extracellular S. aureus and co-entered the cells, and subsequently released vancomycin to induce rapid elimination of intracellular S. aureus. In the implant infection model, AAC significantly improved the bactericidal effect of vancomycin. Scanning electron microscopy showed that the application of AAC effectively blocked the formation of bacterial biofilm. Further histochemical and micro-CT analysis showed AAC significantly reduced the level of bone marrow density (BMD) and bone volume fraction (BV/TV) reduction caused by bacterial infection in the distal femur of mice compared to vancomycin treatment alone. CONCLUSIONS The application of AAC in an implant infection model showed that it significantly improved the bactericidal effects of vancomycin and effectively blocked the formation of bacterial biofilms, without apparent toxicity to the host.
Collapse
Affiliation(s)
- Leilei Qin
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China.
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China
| | - Yanhao Zhang
- National Engineering Research Center of Immunological, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China.
| | - Jianye Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China.
| | - Liqun Zhao
- National Engineering Research Center of Immunological, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China.
| | - Xiaokai Zhang
- National Engineering Research Center of Immunological, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China.
| | - Yun Yang
- National Engineering Research Center of Immunological, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China.
| | - Jinyong Zhang
- National Engineering Research Center of Immunological, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China.
| | - Yinshuang Zou
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China.
| | - Keyu Wei
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China.
| | - Chen Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China.
| | - Yujian Li
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China.
| | - Hao Zeng
- National Engineering Research Center of Immunological, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China; State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, China.
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China.
| | - Quanming Zou
- National Engineering Research Center of Immunological, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China.
| |
Collapse
|
37
|
Ge M, Ruan Z, Zhu YX, Wu W, Yang C, Lin H, Shi J. A natural killer cell mimic against intracellular pathogen infections. SCIENCE ADVANCES 2024; 10:eadp3976. [PMID: 39475620 PMCID: PMC11524181 DOI: 10.1126/sciadv.adp3976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024]
Abstract
In the competition between the pathogen infection and the host defense, infectious microorganisms may enter the host cells by evading host defense mechanisms and use the intracellular biomolecules as replication nutrient. Among them, intracellular Staphylococcus aureus relies on the host cells to protect itself from the attacks by antibiotics or immune system to achieve long-term colonization in the host, and the consequent clinical therapeutic failures and relapses after antibiotic treatment. Here, we demonstrate that intracellular S. aureus surviving well even in the presence of vancomycin can be effectively eliminated using an emerging cell-mimicking therapeutic strategy. These cell mimics with natural killer cell-like activity (NKMs) are composed of a redox-responsive degradable carrier, and perforin and granzyme B within the carrier. NKMs perform far more effectivly than clinical antibiotics in treating intracellular bacterial infections, providing a direct evidence of the NK cell-mimicking immune mechanism in the treatment of intracellular S. aureus.
Collapse
Affiliation(s)
- Min Ge
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai 200050, P. R. China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Zesong Ruan
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P. R. China
| | - Ya-Xuan Zhu
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai 200050, P. R. China
| | - Wencheng Wu
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai 200050, P. R. China
| | - Chuang Yang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P. R. China
| | - Han Lin
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai 200050, P. R. China
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
| | - Jianlin Shi
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai 200050, P. R. China
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, P. R. China
| |
Collapse
|
38
|
Lu KY, Yang X, Eldridge MJG, Wagner NJ, Hardy B, Axtman M, Rowe SE, Wang X, Fowler VG, Helaine S, Pearce KH, Conlon BP. A host-directed adjuvant resuscitates and sensitizes intracellular bacterial persisters to antibiotics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.30.615828. [PMID: 39554024 PMCID: PMC11565766 DOI: 10.1101/2024.09.30.615828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
There are two major problems in the field of antimicrobial chemotherapy-antibiotic resistance and antibiotic tolerance. In the case of antibiotic tolerance, antibiotics fail to kill the bacteria as their phenotypic state affords them protection from the bactericidal activity of the antibiotic. Antibiotic tolerance can affect an entire bacterial population, or a subset of cells known as persister cells. Interaction with the host induces the formation of persister cells in numerous pathogens, with reactive oxygen and nitrogen species production being heavily implicated in the collapse of bacterial energy levels and entrance into an antibiotic tolerant state. Here, we developed a high-throughput screen to identify energy modulators for intracellular Staphylococcus aureus . The identified compound, KL1 , increases intracellular bacterial energy and sensitizes the persister population to antibiotics, without causing cytotoxicity or bacterial outgrowth. We demonstrate that KL1 exhibits adjuvant activity in a murine model of S. aureus bacteremia and intracellular infection of Salmonella Typhimurium . Transcriptomic analysis and further studies on its mechanism of action reveal that KL1 modulates host immune response genes and suppresses the production of reactive species in host macrophages, alleviating one of the major stressors that induce antibiotic tolerance. Our findings highlight the potential to target intracellular persister cells by stimulating their metabolism and encourage larger efforts to address antibiotic tolerance at the host-pathogen interface, particularly within the intracellular milieu.
Collapse
|
39
|
Rycroft JA, Giorgio RT, Sargen M, Helaine S. Tracking the progeny of bacterial persisters using a CRISPR-based genomic recorder. Proc Natl Acad Sci U S A 2024; 121:e2405983121. [PMID: 39374386 PMCID: PMC11494289 DOI: 10.1073/pnas.2405983121] [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: 03/22/2024] [Accepted: 08/11/2024] [Indexed: 10/09/2024] Open
Abstract
The rise of antimicrobial failure is a global emergency, and causes beyond typical genetic resistance must be determined. One probable factor is the existence of subpopulations of transiently growth-arrested bacteria, persisters, that endure antibiotic treatment despite genetic susceptibility to the drug. The presence of persisters in infected hosts has been successfully established, notably through the development of fluorescent reporters. It is proposed that infection relapse is caused by persisters resuming growth after cessation of the antibiotic treatment, but to date, there is no direct evidence for this. This is because no tool or reporter currently exists to track the extent to which infection relapse is initiated by regrowth of persisters in the host. Indeed, once they have transitioned out of the persister state, the progeny of persisters are genetically and phenotypically identical to susceptible bacteria in the population, making it virtually impossible to ascertain the source of relapse. We designed pSCRATCH (plasmid for Selective CRISPR Array expansion To Check Heritage), a molecular tool that functions to record the state of antibiotic persistence in the genome of Salmonella persisters. We show that pSCRATCH successfully marks persisters by adding spacers in their CRISPR arrays and the genomic label is stable in persister progeny after exit from persistence. We further show that in a Salmonella infection model the system enables the discrimination of treatment failure originating from persistence versus resistance. Thus, pSCRATCH provides proof of principle for stable marking of persisters and a prototype for applications to more complex infection models and other pathogens.
Collapse
Affiliation(s)
| | | | - Molly Sargen
- Department of Microbiology, Harvard Medical School, Boston, MA02115
| | - Sophie Helaine
- Department of Microbiology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
40
|
Dadole I, Blaha D, Personnic N. The macrophage-bacterium mismatch in persister formation. Trends Microbiol 2024; 32:944-956. [PMID: 38443279 DOI: 10.1016/j.tim.2024.02.009] [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: 07/21/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/07/2024]
Abstract
Many pathogens are hard to eradicate, even in the absence of genetically detectable antimicrobial resistance mechanisms and despite proven antibiotic susceptibility. The fraction of clonal bacteria that temporarily elude effective antibiotic treatments is commonly known as 'antibiotic persisters.' Over the past decade, there has been a growing body of research highlighting the pivotal role played by the cellular host in the development of persisters. In parallel, this research has also sought to elucidate the molecular mechanisms underlying the formation of intracellular antibiotic persisters and has demonstrated a prominent role for the bacterial stress response. However, questions remain regarding the conditions leading to the formation of stress-induced persisters among a clonal population of intracellular bacteria and despite an ostensibly uniform environment. In this opinion, following a brief review of the current state of knowledge regarding intracellular antibiotic persisters, we explore the ways in which macrophage functional heterogeneity and bacterial phenotypic heterogeneity may contribute to the emergence of these persisters. We propose that the degree of mismatch between the macrophage permissiveness and the bacterial preparedness to invade and thrive intracellularly may explain the formation of stress-induced nonreplicating intracellular persisters.
Collapse
Affiliation(s)
- Iris Dadole
- CIRI - Centre International de Recherche en Infectiologie, CNRS, INSERM, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France; Group Persistence and single-cell dynamics of respiratory pathogens, CIRI, Lyon, France
| | - Didier Blaha
- CIRI - Centre International de Recherche en Infectiologie, CNRS, INSERM, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France; Group Persistence and single-cell dynamics of respiratory pathogens, CIRI, Lyon, France
| | - Nicolas Personnic
- CIRI - Centre International de Recherche en Infectiologie, CNRS, INSERM, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France; Group Persistence and single-cell dynamics of respiratory pathogens, CIRI, Lyon, France.
| |
Collapse
|
41
|
Hira J, Singh B, Halder T, Mahmutovic A, Ajayi C, Sekh AA, Hegstad K, Johannessen M, Lentz CS. Single-cell phenotypic profiling and backtracing exposes and predicts clinically relevant subpopulations in isogenic Staphylococcus aureus communities. Commun Biol 2024; 7:1228. [PMID: 39354092 PMCID: PMC11445386 DOI: 10.1038/s42003-024-06894-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
Isogenic bacterial cell populations are phenotypically heterogenous and may include subpopulations of antibiotic tolerant or heteroresistant cells. The reversibility of these phenotypes and lack of biomarkers to differentiate functionally different, but morphologically identical cells is a challenge for research and clinical detection. To overcome this, we present ´Cellular Phenotypic Profiling and backTracing (CPPT)´, a fluorescence-activated cell sorting platform that uses fluorescent probes to visualize and quantify cellular traits and connects this phenotypic profile with a cell´s experimentally determined fate in single cell-derived growth and antibiotic susceptibility analysis. By applying CPPT on Staphylococcus aureus we phenotypically characterized dormant cells, exposed bimodal growth patterns in colony-derived cells and revealed different culturability of single cells on solid compared to liquid media. We demonstrate that a fluorescent vancomycin conjugate marks cellular subpopulations of vancomycin-intermediate S. aureus with increased likelihood to survive antibiotic exposure, showcasing the value of CPPT for discovery of clinically relevant biomarkers.
Collapse
Affiliation(s)
- Jonathan Hira
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Bhupender Singh
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Tirthankar Halder
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Anel Mahmutovic
- Early Biometrics & Statistical Innovation Data Science & AI AstraZeneca, Biopharmaceuticals RD AstraZeneca, Mölndal, Sweden
| | - Clement Ajayi
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | | | - Kristin Hegstad
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Mona Johannessen
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Christian S Lentz
- Centre for New Antibacterial Strategies (CANS) and Research Group for Host-Microbe Interactions, Department of Medical Biology, UiT - The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
42
|
Jeong GJ, Khan F, Tabassum N, Cho KJ, Kim YM. Marine-derived bioactive materials as antibiofilm and antivirulence agents. Trends Biotechnol 2024; 42:1288-1304. [PMID: 38637243 DOI: 10.1016/j.tibtech.2024.03.009] [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: 01/05/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024]
Abstract
Microbial infections are major human health issues, and, recently, the mortality rate owing to bacterial and fungal infections has been increasing. In addition to intrinsic and extrinsic antimicrobial resistance mechanisms, biofilm formation is a key adaptive resistance mechanism. Several bioactive compounds from marine organisms have been identified for use in biofilm therapy owing to their structural complexity, biocompatibility, and economic viability. In this review, we discuss recent trends in the application of marine natural compounds, marine-bioinspired nanomaterials, and marine polymer conjugates as possible therapeutic agents for controlling biofilms and virulence factors. We also comprehensively discuss the mechanisms underlying biofilm formation and inhibition of virulence factors by marine-derived materials and propose possible applications of novel and effective antibiofilm and antivirulence agents.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea.
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Kyung-Jin Cho
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
43
|
Zhao R, Xu L, Chen J, Yang Y, Guo X, Dai M, Tian GB, Qin LN. Itaconate induces tolerance of Staphylococcus aureus to aminoglycoside antibiotics. Front Microbiol 2024; 15:1450085. [PMID: 39403084 PMCID: PMC11471559 DOI: 10.3389/fmicb.2024.1450085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/09/2024] [Indexed: 01/04/2025] Open
Abstract
INTRODUCTION Staphylococcus aureus is one of the chief pathogens that cause chronic and recurrent infections. Failure of the antibiotics to curb the infections contributes to relapse and is an important reason for the high mortality rate. Treatment failure may also be due to antibiotic tolerance. Accumulating evidence suggests that t the host immune environment plays an important role in inducing antibiotic tolerance of S. aureus, but research in this area has been limited. METHODS In this study,the minimum inhibitory concentration (MIC) of the antibiotics against S. aureus was determined using the standard broth microdilution method.The study evaluated whether itaconate induces antibiotic tolerance in S. aureus through an antibiotic bactericidal activity assay.The effect of itaconate on the growth of S. aureus was evaluated by monitoring the growth of S. aureus in medium supplemented with itaconate. Additionally, RNA sequencing and metabolomics analyses were used to determine transcriptional and metabolic changes in S. aureus when exposed to itaconate. RESULTS AND DISCUSSION According to the study,we found that the immune metabolite itaconate can induce tolerance in both methicillin-resistant and -susceptible S. aureus to aminoglycosides. When S. aureus was exposed to itaconate, its growth slowed down and transcriptomic and metabolomic alterations associated with decreased energy metabolism, including the tricarboxylate cycle, glycolysis, pyruvate metabolism, and arginine biosynthesis, were observed. These changes are associated with aminoglycoside tolerance. This study highlights the role of immune signaling metabolites in bacterial antibiotic tolerance and suggests new strategies to improve antibiotic treatment by modulating the host immune response and stimulating the metabolism of bacteria.
Collapse
Affiliation(s)
- Runping Zhao
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Lei Xu
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Jieyun Chen
- Zhongshan School of Medicine, Advanced Medical Technology Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Key Laboratory of Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Yanxian Yang
- Zhongshan School of Medicine, Advanced Medical Technology Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Key Laboratory of Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Xilong Guo
- Zhongshan School of Medicine, Advanced Medical Technology Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Key Laboratory of Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Min Dai
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Guo-Bao Tian
- Zhongshan School of Medicine, Advanced Medical Technology Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Key Laboratory of Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Li-Na Qin
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
44
|
Piechowicz L, Kosznik-Kwaśnicka K, Jarzembowski T, Daca A, Necel A, Bonawenturczak A, Werbowy O, Stasiłojć M, Pałubicka A. Staphylococcus aureus Co-Infection in COVID-19 Patients: Virulence Genes and Their Influence on Respiratory Epithelial Cells in Light of Risk of Severe Secondary Infection. Int J Mol Sci 2024; 25:10050. [PMID: 39337536 PMCID: PMC11431965 DOI: 10.3390/ijms251810050] [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: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Pandemics from viral respiratory tract infections in the 20th and early 21st centuries were associated with high mortality, which was not always caused by a primary viral infection. It has been observed that severe course of infection, complications and mortality were often the result of co-infection with other pathogens, especially Staphylococcus aureus. During the COVID-19 pandemic, it was also noticed that patients infected with S. aureus had a significantly higher mortality rate (61.7%) compared to patients infected with SARS-CoV-2 alone. Our previous studies have shown that S. aureus strains isolated from patients with COVID-19 had a different protein profile than the strains in non-COVID-19 patients. Therefore, this study aims to analyze S. aureus strains isolated from COVID-19 patients in terms of their pathogenicity by analyzing their virulence genes, adhesion, cytotoxicity and penetration to the human pulmonary epithelial cell line A549. We have observed that half of the tested S. aureus strains isolated from patients with COVID-19 had a necrotizing effect on the A549 cells. The strains also showed greater variability in terms of their adhesion to the human cells than their non-COVID-19 counterparts.
Collapse
Affiliation(s)
- Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Katarzyna Kosznik-Kwaśnicka
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Tomasz Jarzembowski
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Agnieszka Daca
- Department of Physiopathology, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Agnieszka Necel
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Ada Bonawenturczak
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Olesia Werbowy
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Małgorzata Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland
| | - Anna Pałubicka
- Specialist Hospital in Koscierzyna Sp. z o.o., Department of Laboratory and Microbiological Diagnostics, Koscierzyna, Alojzego Piechowskiego 36, 83-400 Koscierzyna, Poland
| |
Collapse
|
45
|
Liu X, Liu Y, Song M, Zhu K, Shen J. A Rhein-Based Derivative Targets Staphylococcus aureus. Antibiotics (Basel) 2024; 13:882. [PMID: 39335055 PMCID: PMC11428220 DOI: 10.3390/antibiotics13090882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
The rise in antibiotic-resistant bacteria highlights the need for novel antimicrobial agents. This study presents the design and synthesis of a series of rhein (RH)-derived compounds with improved antimicrobial properties. The lead compound, RH17, exhibited a potent antibacterial activity against Staphylococcus aureus (S. aureus) isolates, with minimum inhibitory concentrations (MICs) ranging from 8 to 16 μg/mL. RH17 disrupted bacterial membrane stability, hindered metabolic processes, and led to an increase in reactive oxygen species (ROS) production. These mechanisms were confirmed through bacterial growth inhibition assays, membrane function assessments, and ROS detection. Notably, RH17 outperformed the parent compound RH and demonstrated bactericidal effects in S. aureus. The findings suggest that RH17 is a promising candidate for further development as an antimicrobial agent against Gram-positive pathogens, addressing the urgent need for new therapies.
Collapse
Affiliation(s)
- Xiaojia Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Yuan Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Meirong Song
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| |
Collapse
|
46
|
Ahmed F, Mirani ZA, Urooj S, Noor Ul Hudda H, Janees Imdad M, Zhao Y, Malakar PK. A rare biofilm dispersion strategy demonstrated by Staphylococcus aureus under oxacillin stress. Microb Pathog 2024; 194:106838. [PMID: 39111368 DOI: 10.1016/j.micpath.2024.106838] [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: 02/17/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
Staphylococcus aureus (S. aureus), a versatile Gram-positive bacterium, is implicated in a spectrum of infections, and its resilience is often attributed to biofilm formation. This study investigates the effect of sub-inhibitory doses of oxacillin on biofilm formation by methicillin-resistant S. aureus (MRSA). Specifically, it examines how these doses influence biofilms' development, maturation, and dispersal. The biofilm's zenith reached 48 h of incubation, followed by a noteworthy decline at 96 h and a distinctive clearance zone around biofilm-positive cells exposed to oxacillin. Scanning electron micrographs unveiled an intriguing active biofilm dispersal mechanism, a rarity in this species. Among 180 isolates, only three carrying the elusive icaD gene exhibited this phenomenon. icaD gene was absent in their counterparts. Notably, the icaD gene emerges as a distinctive marker, crucial in regulating biofilm dispersion and setting these isolates apart. The captivating interplay of oxacillin, biofilm dynamics, and genetic signatures disintegrate novel dimensions in understanding MRSA's adaptive strategies and underscores the importance of the icaD gene in engineering biofilm resilience.
Collapse
Affiliation(s)
- Faraz Ahmed
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; International Research Centre for Food and Health, Shanghai Ocean University, Shanghai, China.
| | - Zulfiqar Ali Mirani
- Microbiology Section, FMRRC, PCSIR Laboratories Complex Karachi, Sindh, Pakistan
| | - Shaista Urooj
- Aquatic Diagnostic and Research Center Bahria University, Karachi, Sindh, Pakistan
| | | | - Muhammad Janees Imdad
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; International Research Centre for Food and Health, Shanghai Ocean University, Shanghai, China
| | - Yong Zhao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; International Research Centre for Food and Health, Shanghai Ocean University, Shanghai, China; Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai, China; Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai, China
| | - Pradeep K Malakar
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; International Research Centre for Food and Health, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
47
|
Cheng K, Sun Y, Yu H, Hu Y, He Y, Shen Y. Staphylococcus aureus SOS response: Activation, impact, and drug targets. MLIFE 2024; 3:343-366. [PMID: 39359682 PMCID: PMC11442139 DOI: 10.1002/mlf2.12137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/17/2024] [Accepted: 04/10/2024] [Indexed: 10/04/2024]
Abstract
Staphylococcus aureus is a common cause of diverse infections, ranging from superficial to invasive, affecting both humans and animals. The widespread use of antibiotics in clinical treatments has led to the emergence of antibiotic-resistant strains and small colony variants. This surge presents a significant challenge in eliminating infections and undermines the efficacy of available treatments. The bacterial Save Our Souls (SOS) response, triggered by genotoxic stressors, encompasses host immune defenses and antibiotics, playing a crucial role in bacterial survival, invasiveness, virulence, and drug resistance. Accumulating evidence underscores the pivotal role of the SOS response system in the pathogenicity of S. aureus. Inhibiting this system offers a promising approach for effective bactericidal treatments and curbing the evolution of antimicrobial resistance. Here, we provide a comprehensive review of the activation, impact, and key proteins associated with the SOS response in S. aureus. Additionally, perspectives on therapeutic strategies targeting the SOS response for S. aureus, both individually and in combination with traditional antibiotics are proposed.
Collapse
Affiliation(s)
- Kaiying Cheng
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Yukang Sun
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Huan Yu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yingxuan Hu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yini He
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yuanyuan Shen
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| |
Collapse
|
48
|
Gerile S, Shen Q, Kang J, Liu W, Dong A. Current advances in black phosphorus-based antibacterial nanoplatform for infection therpy. Colloids Surf B Biointerfaces 2024; 241:114037. [PMID: 38878660 DOI: 10.1016/j.colsurfb.2024.114037] [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: 04/19/2024] [Revised: 06/01/2024] [Accepted: 06/12/2024] [Indexed: 07/29/2024]
Abstract
Black phosphorus (BP) has attracted much attention due to its excellent physiochemical properties. However, due to its biodegradability and simple antibacterial mechanism, using only BP nanomaterials to combat bacterial infections caused by drug-resistant pathogens remains a significant challenge. In order to improve the antibacterial efficiency and avoid the emergence of drug resistance, BP nanomaterials have been combined with other functional materials to form black phosphorus-based antibacterial nanoplatform (BPANP), which provides unprecedented opportunities for the treatment of drug-resistant infections. This article reviews the performance of BPANP and its multiple antibacterial mechanisms while emphatically introducing its design direction and latest application progress in antibacterial fields. Moreover, this paper additionally summarizes and discusses the current challenges and inadequacies of BPANP that need to be improved in future research. We believe that this review will provide researchers with an up-to-date and multifaceted reference, and provide new ideas for designing effective strategies against drug-resistant bacteria.
Collapse
Affiliation(s)
- Saren Gerile
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, PR China
| | - Qiudi Shen
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, PR China
| | - Jing Kang
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, PR China.
| | - Wenxin Liu
- College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao 028000, PR China.
| | - Alideertu Dong
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, Inner Mongolia University, Hohhot 010021, PR China.
| |
Collapse
|
49
|
Chapartegui-González I, Stockton JL, Bowser S, Badten AJ, Torres AG. Unraveling the role of toxin-antitoxin systems in Burkholderia pseudomallei: exploring bacterial pathogenesis and interactions within the HigBA families. Microbiol Spectr 2024; 12:e0074824. [PMID: 38916327 PMCID: PMC11302019 DOI: 10.1128/spectrum.00748-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/28/2024] [Indexed: 06/26/2024] Open
Abstract
Burkholderia pseudomallei (Bpm) is a Gram-negative intracellular pathogen that causes melioidosis in humans, a neglected, underreported, and lethal disease that can reach a fatal outcome in over 50% of the cases. It can produce both acute and chronic infections, the latter being particularly challenging to eliminate because of the intracellular life cycle of the bacteria and its ability to generate a "persister" dormant state. The molecular mechanism that allows the switch between growing and persister phenotypes is not well understood but it is hypothesized to be due at least in part to the participation of toxin-antitoxin (TA) systems. We have previously studied the link between one of those systems (defined as HigBA) with specific expression patterns associated with levofloxacin antibiotic exposure. Through in silico methods, we predicted the presence of another three pairs of genes encoding for additional putative HigBA systems. Therefore, our main goal was to establish which mechanisms are conserved as well as which pathways are specific among different Bpm TA systems from the same family. We hypothesize that the high prevalence, and sometimes even redundancy of these systems in the Bpm chromosomes indicates that they can interact with each other and not function as only individual systems, as it was traditionally thought, and might be playing an undefined role in Bpm lifecycle. Here, we show that both the toxin and the antitoxin of the different systems contribute to bacterial survival and that toxins from the same family can have a cumulative effect under environmental stressful conditions. IMPORTANCE Toxin-antitoxin (TA) systems play a significant role in bacterial persistence, a phenomenon where bacterial cells enter a dormant or slow-growing state to survive adverse conditions such as nutrient deprivation, antibiotic exposure, or host immune responses. By studying TA systems in Burkholderia pseudomallei, we can gain insights into how this pathogen survives and persists in the host environment, contributing to its virulence and ability to cause melioidosis chronic infections.
Collapse
Affiliation(s)
| | - Jacob L. Stockton
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Sarah Bowser
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Alexander J. Badten
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Alfredo G. Torres
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
50
|
Chaudhary K, Agrahari B, Biswas B, Chatterjee N, Chaudhary A, Kumar A, Sonker H, Dewan S, Saxena D, Akhir A, Malhotra N, Chopra S, Misra S, Matheswaran S, Singh RG. Pyridine-2,6-Dicarboxamide Proligands and their Cu(II)/Zn(II) Complexes Targeting Staphylococcus Aureus for the Attenuation of In Vivo Dental Biofilm. Adv Healthc Mater 2024; 13:e2400378. [PMID: 38621382 DOI: 10.1002/adhm.202400378] [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: 02/10/2024] [Revised: 04/07/2024] [Indexed: 04/17/2024]
Abstract
In the pursuit to combat stubborn bacterial infections, particularly those stemming from gram-positive bacteria, this study is an attempt to craft a precision-driven platform characterized by unparalleled selectivity, specificity, and synergistic antimicrobial mechanisms. Leveraging remarkable potential of metalloantibiotics in antimicrobial applications, herein, this work rationally designs, synthesizes, and characterizes a new library of Pyridine-2,6-dicarboxamide ligands and their corresponding transition metal Cu(II)/Zn(II) complexes. The lead compound L11 demonstrates robust antibacterial properties against Staphylococcus aureus (Minimum Inhibitory Concentration (MIC) = 2-16 µg mL-1), methicillin and vancomycin-resistant S. aureus (MIC = 2-4 µg mL-1) and exhibit superior antibacterial activity when compared to FDA-approved vancomycin, the drug of last resort. Additionally, the compound exhibits notable antimicrobial efficacy against resistant enterococcus strains (MIC = 2-8 µg mL-1). To unravel mechanistic profile, advanced imaging techniques including SEM and AFM are harnessed, collectively suggesting a mechanistic pathway involving cell wall disruption. Live/dead fluorescence studies further confirm efficacy of L11 and its complexes against S. aureus membranes. This translational exploration extends to a rat model, indicating promising in vivo therapeutic potential. Thus, this comprehensive research initiative has capabilities to transcends the confines of this laboratory, heralding a pivotal step toward combatting antibiotic-resistant pathogens and advancing the frontiers of metalloantibiotics-based therapy with a profound clinical implication.
Collapse
Affiliation(s)
| | | | - Bhumika Biswas
- Department of Biological Sciences and Bioengineering, IIT, Kanpur, 208016, India
| | - Niranjan Chatterjee
- Department of Biological Sciences and Bioengineering, IIT, Kanpur, 208016, India
| | | | | | | | - Sayari Dewan
- Department of Chemistry, IIT, Kanpur, 208016, India
| | - Deepanshi Saxena
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Abdul Akhir
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Nidhi Malhotra
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence, Gautam Budh Nagar, 201314, India
| | - Sidharth Chopra
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Santosh Misra
- Department of Biological Sciences and Bioengineering, IIT, Kanpur, 208016, India
| | | | | |
Collapse
|