1
|
Wu Q, Zhu H, Zhang R, Li C, Xiao Q, Jin Y, Liang X, Chen X, Dong X. Closer adult child-parent relations the stronger the vaccine hesitancy: A cross-sectional study of adult Children's attitudes toward pneumococcal vaccination of elderly parents and its determinants in Guangzhou, China. Hum Vaccin Immunother 2025; 21:2440959. [PMID: 39757490 DOI: 10.1080/21645515.2024.2440959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/18/2024] [Accepted: 12/08/2024] [Indexed: 01/07/2025] Open
Abstract
Pneumococcal disease (PD) has a serious effect on older people aged 60 years and above. However, pneumococcal vaccination rates for older people in China remain low. This study aimed to explore adult children's perspectives on the vaccination of their parents and to examine the determinants of vaccine hesitancy. In October 2022, a cross-sectional survey was conducted in Guangzhou, China. The questionnaire assesses data on the sociodemographic characteristics of adult children and their parents, health beliefs about vaccination, and variables measuring adult child-parent relationships. Adult child-parent relationships types were identified by Latent Class Analysis (LCA). Binary logistic regression was employed to examine the factors associated with vaccine hesitancy. A total of 1,597 respondents were enrolled in the study, and 59.8% of the adult children expressed hesitancy about vaccinating their elderly parents. The LCA model identified three distinct types of adult child-parent relations: detached, intimate but distant, and tight-knit. Binary logistic regression analyses revealed that respondents with intimate but distant (OR = 3.04) and tight-knit (OR = 3.01) adult child - parent relationships, high literacy (OR = 2.63), and high perceived barriers of vaccine (OR = 1.18) were more likely to be hesitant. Conversely, those with high income (OR = 0.35) and parents with difficulties in activities of daily living (OR = 0.44) were less likely to exhibit vaccine hesitancy. Close adult child-parent relations were positively associated with vaccine hesitancy. Health education related to the vaccination of older persons should be extended to adult children.
Collapse
Affiliation(s)
- Qiqi Wu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Haiyuan Zhu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Runquan Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Cuizhi Li
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Qin Xiao
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Yuqing Jin
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xiaofeng Liang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xiongfei Chen
- Department of Primary Public Health, Guangzhou Center for Disease Control and Prevention, Guangdong, China
| | - Xiaomei Dong
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Wang X, Wang H, Zhang J, Ma W, Zhang D, Chen Y. Mechanism and different roles of metal-N sites on ZIF-8 for efficient antibacterial. J Environ Sci (China) 2025; 156:68-78. [PMID: 40412966 DOI: 10.1016/j.jes.2024.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 05/27/2025]
Abstract
Bacterial pollution poses a serious threat to human health, making it essential to design and utilize efficient non-antibiotic antibacterial materials. Here, ZIF-8 with different metal-N sites is successfully prepared by introducing divalent metals (Mg2+, Mn2+, Co2+ and Cu2+) directly into the ZIF-8 framework. ZIF-8 with Cu-Nx sites has the best antibacterial activity, with antibacterial rates of 99.8 % and 81.1 % against Escherichia coli and Staphylococcus aureus after 1 h at a concentration of 10 µg/mL, respectively. More importantly, an antibacterial rate of more than 86.7 % can be achieved against multidrug-resistant bacteria MRSA, much higher than Vancomycin. The results show that the introduction of copper could significantly improve the electron transfer, the generation of reactive oxygen species (ROS), the binding affinity with bacteria, and eventually achieve excellent antibacterial activity. DFT calculations show easier oxygen activation at the unsaturated Cu-Nx site. The revealed oxygen activation mechanism sheds light on understanding the high antibacterial activity of the active site of the nanoparticles. Cu-ZIF-8 offers significant advantages in the field of air disinfection.
Collapse
Affiliation(s)
- Xiaoze Wang
- State Key Laboratory of Mesoscience and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Wang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanosystem and Hierarchial Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Jingkun Zhang
- State Key Laboratory of Mesoscience and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjun Ma
- State Key Laboratory of Mesoscience and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Donghai Zhang
- State Key Laboratory of Mesoscience and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yunfa Chen
- State Key Laboratory of Mesoscience and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of the Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Li J, Zhou S, Bai R, Fu Z. A 3D-printed integrated maneuverable device for sensitive colorimetry of Pseudomonas aeruginosa. J Pharm Biomed Anal 2025; 263:116934. [PMID: 40315595 DOI: 10.1016/j.jpba.2025.116934] [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: 02/11/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
Timely and sensitive monitoring of pathogens in clinical specimen is highly demanded for efficient control and precise treatment of infectious diseases. Herein, a 3D-printed maneuverable device integrating incubation, washing, and detection functions was manufactured for colorimetry of Pseudomonas aeruginosa (P. aeruginosa) using transparent resin. The device combined with magnetic beads (MBs) can achieve specific separation and efficient enrichment of P. aeruginosa. Furthermore, it can be directly fixed onto a 96-well plate holder for colorimetry. Specifically, a P. aeruginosa bacteriophage termed as JZ1 acquired from river water was applied as a recognition reagent to functionalize the separation vectors MBs. Then, a nanoconfinement MOFs material termed as PCN-222(Pt) with remarkable peroxidase-like activity was conjugated with polymyxin B to act as a signal tracer. With the formation of target bacterial complexes, the bound PCN-222(Pt) catalyzed the color reaction of 3,3',5,5'-tetramethylbenzidine to enable quantitative colorimetry of P. aeruginosa by the maneuverable device. With this device, P. aeruginosa can be quantified within 40 min, with a dynamic range of 1.9 × 102 ∼ 1.9 × 106 cfu mL-1. The results for colorimetry of P. aeruginosa in diverse sample matrixes demonstrated its satisfactory practicability. This work provides a facile, timely, and cost-effective technique for point-of-care testing of pathogens.
Collapse
Affiliation(s)
- Jizhou Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Shiruoyu Zhou
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ruining Bai
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhifeng Fu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
4
|
Zhou W, Han Y, Li W, Deng A, Li Y, Xu J, Zhu G, Yang Z. Prophage transduction promotes the transmission of phage resistance interfering with adsorption among Chinese foodborne Staphylococcus aureus. Int J Food Microbiol 2025; 440:111271. [PMID: 40403651 DOI: 10.1016/j.ijfoodmicro.2025.111271] [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: 03/28/2025] [Revised: 05/12/2025] [Accepted: 05/14/2025] [Indexed: 05/24/2025]
Abstract
Although bacteriophages have proven to be efficient biocontrol agents for foodborne Staphylococcus aureus, the transmission of phage resistance resulting in the reduced efficacy of phage therapy remains to be explored. In this study, phage resistance and adsorption of 91 Chinese foodborne S. aureus isolates by 18 phages were estimated, and the distribution and transmission of phage resistance genes were investigated. The isolated 91 S. aureus comprised 50 multidrug-resistance isolates, all of which showed sensitivity to more than two phages. However, 9.9 % (9/91) of S. aureus isolates were resistant to all 18 phages, and the majority of phages (83.3 %, 15/18) did not adsorb to all foodborne S. aureus strains. Whole-genome analysis revealed that the 91 isolates comprised 101 phage resistance genes, including 24 genes were found in prophages (intact prophages, 19.8 %, 20/101; incomplete prophages, 16.8 %, 17/101). Notably, a temperate phage SapYZUs631 was successfully induced and exhibited better biological characteristics compared to other isolated S. aureus temperate phages, including higher titre (6.2 × 109 PFU/mL), stronger pH (4-11) and thermal (60 °C for 60 min) stability, and a wider host range (80.2 %, 73/91). The SapYZUs631 genome contained phage resistance gene tarP interfering with adsorption and virulence genes. The lysogeny of SapYZUs631 into S. aureus strains YZUstau27, YZUstau31, and YZUstau35 resulted in increased phage resistance and decreased adsorption. Therefore, our analysis suggests that the interruption of adsorption is the main reason for the phage resistance of foodborne S. aureus in China, which resulted from the transmission of phage resistance by prophage transduction.
Collapse
Affiliation(s)
- Wenyuan Zhou
- College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China; College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Catering Food Processing and Safety Control, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yeling Han
- College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Wenjuan Li
- College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Aiping Deng
- College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Yajie Li
- College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Jiaqian Xu
- College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| | - Zhenquan Yang
- College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Catering Food Processing and Safety Control, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
5
|
Fraiman A, Ziegler LD. Ultra-rapid, quantitative, label-free antibiotic susceptibility testing via optically detected purine metabolites. Talanta 2025; 292:127907. [PMID: 40090249 DOI: 10.1016/j.talanta.2025.127907] [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: 01/16/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
In order to facilitate the best antimicrobial prescribing practices and to help reduce the increasing global threat of antibiotic resistance, there is an urgent need for the development of novel and truly rapid (≤1 h) antibiotic susceptibility testing (AST) platforms. A 785 nm surface enhanced Raman spectroscopy (SERS) based phenotypic methodology is described that results in accurate minimum inhibitory concentration (MIC) determinations for all tested strain/antibiotic pairs. The SERS-AST procedure results in accurate MICs in ∼1 h, including a 30-min incubation period, and is effective for both Gram positive and negative species, and for antibiotics with different initial primary targets of antibiotic activity, and for both bactericidal and bacteriostatic antibiotics. The molecular level mechanism of this methodology is described. Bacterial SERS spectra are due to secreted purine nucleotide degradation products (principally adenine, guanine, xanthine and hypoxanthine) resulting from water washing induced bacterial stringent response and the resulting (p)ppGpp alarmone mediates nucleobase formation from unneeded tRNA and rRNA. The rewiring of metabolic responses resulting from the secondary metabolic effects of antibiotic exposure during the 30-min incubation period accounts for the dose dependence of the SERS spectral intensities which are used to accurately yield the MIC. This is the fastest demonstrated AST method yielding MICs.
Collapse
Affiliation(s)
- A Fraiman
- Department of Chemistry and the Photonics Center, Boston University, Boston, MA, 02215, USA
| | - L D Ziegler
- Department of Chemistry and the Photonics Center, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
6
|
Song T, Li N, Zuo Q, Huang L, Liu Z, Guo Z. Mucus-penetrating nanomotor system strengthens mucosal immune response to in situ bacterial vaccine against severe bacterial pneumonia. Biomaterials 2025; 320:123236. [PMID: 40054375 DOI: 10.1016/j.biomaterials.2025.123236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/17/2025] [Accepted: 03/02/2025] [Indexed: 04/06/2025]
Abstract
Pathogens causing major infectious diseases primarily invade through mucosal tissues. Promptly killing these pathogens at the mucosal site and constructing mucosal vaccines in situ can prevent further infections and induce robust mucosal immune responses and memory to prevent reinfection. In this study, we utilized chemotherapy, sonodynamic therapy, and gas therapy to eliminate Streptococcus pneumoniae (S. pneumoniae) colonizing the nasal mucosa. Simultaneously, an in situ pneumococcal vaccine was constructed to elicit specific immune responses and memory. Poly-l-arginine (PArg)-modified ZIF-8 metal-organic frameworks (MOFs) loaded with the ultrasonic sensitizer protoporphyrin IX (PpIX) killed S. pneumoniae in the nasal cavity by multiple mechanisms in the presence of ultrasound. When stimulated by ultrasound, PpIX not only generates reactive oxygen species (ROS) for antimicrobial effect, but these ROS also catalyze the release of nitric oxide (NO) from PArg. NO exerts a motor-like effect that facilitates more efficient passage of nanoparticles through the mucus layer of the alveoli. The immunogenic bacterial debris formed a vaccine formulation by complexing with PArg, which adhered electrostatically to the mucosal surface, facilitating in situ vaccination and inducing mucosal immune responses and memory. This cascade-based combination therapy enabled rapid bacterial eradication and long-term immune prevention. It shortens the traditional vaccine development process, eliminates the spatial distance from pathogen invasion to vaccine development, significantly cuts costs, and addresses vaccine failure due to pathogen mutations. This approach offers a groundbreaking strategy for mucosal vaccine development and the prevention of major infectious diseases.
Collapse
Affiliation(s)
- Ting Song
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Nan Li
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qinhua Zuo
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Linghong Huang
- College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Zonghua Liu
- College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Zhong Guo
- Center for Biological Science and Technology & Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China.
| |
Collapse
|
7
|
Zeng X, Li C, Li Z, Tao Z, Li M. Review of research advances in microbial sterilization technologies and applications in the built environment. J Environ Sci (China) 2025; 154:314-348. [PMID: 40049877 DOI: 10.1016/j.jes.2024.09.026] [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: 07/03/2024] [Revised: 09/28/2024] [Accepted: 09/28/2024] [Indexed: 05/13/2025]
Abstract
As globalization accelerates, microbial contamination in the built environment poses a major public health challenge. Especially since Corona Virus Disease 2019 (COVID-19), microbial sterilization technology has become a crucial research area for indoor air pollution control in order to create a hygienic and safe built environment. Based on this, the study reviews sterilization technologies in the built environment, focusing on the principles, efficiency and applicability, revealing advantages and limitations, and summarizing current research advances. Despite the efficacy of single sterilization technologies in specific environments, the corresponding side effects still exist. Thus, this review highlights the efficiency of hybrid sterilization technologies, providing an in-depth understanding of the practical application in the built environment. Also, it presents an outlook on the future direction of sterilization technology, including the development of new methods that are more efficient, energy-saving, and targeted to better address microbial contamination in the complex and changing built environment. Overall, this study provides a clear guide for selecting technologies to handle microbial contamination in different building environments in the future, as well as a scientific basis for developing more effective air quality control strategies.
Collapse
Affiliation(s)
- Xinran Zeng
- School of Mechanical Engineering Department, Tongji University, Shanghai 201804, China
| | - Chunhui Li
- School of Mechanical Engineering Department, Tongji University, Shanghai 201804, China.
| | - Zhenhai Li
- School of Mechanical Engineering Department, Tongji University, Shanghai 201804, China.
| | - Zhizheng Tao
- SWJTU-Leeds Joint School, Southwest Jiaotong University, Chengdu 610097, China
| | - Mingtong Li
- School of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin 132022, China
| |
Collapse
|
8
|
Cifuentes-González C, Le Tong Y, Mejía-Salgado G, Chan R, Walter CFY, Rojas-Carabali W, Putera I, Mobasserian A, Nora RLD, Biswas J, Gangaputra S, Pulido JS, Kempen JH, Nguyen QD, de-la-Torre A, Gupta V, Rosenbaum JT, Agrawal R, REVISE-IUSG study group. Global demographic and etiological variations of retinal vasculitis: A systematic review and meta-analysis: International Uveitis Study Group (IUSG) Retinal Vasculitis Study (ReViSe) Report 1. Surv Ophthalmol 2025; 70:756-770. [PMID: 39921003 DOI: 10.1016/j.survophthal.2025.01.012] [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: 09/28/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 02/10/2025]
Abstract
This systematic review with meta-analysis explores the global demographic and etiological variations of retinal vasculitis (RV), focusing on differences in frequency, age, sex, and etiology across diverse geographic populations. RV is an inflammatory condition that can lead to visual impairment, making understanding its variations essential for targeted screening and management. Systematic searches were conducted in multiple databases up to February, 2023, following PRISMA guidelines. We included studies with at least 10 RV cases, such that a frequency measurement can be estimated, without restrictions on publication date or language. RV was categorized as Idiopathic RV in the absence of additional ocular or systemic disease, Syndromic RV for ocular involvement without systemic disease, and Secondary RV in those asssociated with systemic disease. The risk of bias was evaluated using standardized tools. A total of 95 studies, including 23,180 patients, were analyzed. The overall RV frequency among uveitis cohorts was 17 %, with European populations showing the highest frequency at 25 %. Idiopathic RV accounted for 1 % of uveitis cohorts and 38 % of RV cohorts, with significant differences across continents. Behçet disease had the highest RV frequency at 56 %. The median age of diagnosis was 33.5 years, and RV was more frequent in males (57 %). Our findings underscore the considerable geographic and demographic variability in RV, particularly in Idiopathic RV, tuberculosis-related RV, and Behçet disease, highlighting the need for tailored, region-specific, and gender-specific approaches to RV diagnosis and treatment.
Collapse
Affiliation(s)
- Carlos Cifuentes-González
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore; Programme for Ocular Inflammation & Infection Translational Research, Department of Ophthalmology, National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | - Yong Le Tong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Germán Mejía-Salgado
- Neuroscience (NEUROS) Research Group, Neurovitae Research Center, Institute of Translational Medicine (IMT). Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia; Colombian Visual Science and Translational Eye Research Insitute (CERI), Center of Excellence in Ocular Inflammation, Bogotá, Colombia; Helath Science Faculty, Universidad Autónoma de Bucaramanga UNAB, Bucaramanga, Colombia
| | - Reo Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - William Rojas-Carabali
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore; Programme for Ocular Inflammation & Infection Translational Research, Department of Ophthalmology, National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Ikhwanuliman Putera
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia; Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Azadeh Mobasserian
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Rina La Distia Nora
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia
| | - Jyotirmay Biswas
- Department of Uvea and Ocular Pathology, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Sapna Gangaputra
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jose S Pulido
- Ocular Oncology Service, Wills Eye Hospital, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA, USA; Bower Laboratory for Translational Medicine, Vickie and Jack Farber Vision Research Center at Wills Eye Hospital, Wills Eye Hospital, Philadelphia, PA, USA
| | - John H Kempen
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology and Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA; Sight for Souls, Bellevue, WA, USA; MyungSung Christian Medical Center (MCM) Comprehensive Specialized Hospital and MyungSung Medical College, Addis Ababa, Ethiopia; MCM Eye Unit, Addis Ababa, Ethiopia; Department of Ophthalmology, Addis Ababa University School of Medicine, Addis Ababa, Ethiopia
| | - Quan Dong Nguyen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Alejandra de-la-Torre
- Neuroscience (NEUROS) Research Group, Neurovitae Research Center, Institute of Translational Medicine (IMT). Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Vishali Gupta
- Advanced Eye Centre, Post, graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | - Rupesh Agrawal
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore; Programme for Ocular Inflammation & Infection Translational Research, Department of Ophthalmology, National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Singapore Eye Research Institute, Singapore; Duke NUS Medical School, Singapore.
| | | |
Collapse
|
9
|
Shrestha S, Malla B, Haramoto E. 6-plex Crystal Digital PCR® for comprehensive surveillance of respiratory and foodborne bacterial pathogens in wastewater. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 375:126298. [PMID: 40274213 DOI: 10.1016/j.envpol.2025.126298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
Bacterial wastewater surveillance (WS) is less explored area compared to viral WS despite high burden of bacterial respiratory and gastrointestinal infections worldwide. This study established a 6-plex Crystal Digital PCR® (cdPCR) system, to comprehensively monitor an acute respiratory pathogen - Group A Streptococcus (GAS) pyogenes, foodborne disease (FBD) pathogens - Clostridium perfringens, Salmonella spp., Campylobacter jejuni, and Campylobacter coli, and an indicator bacterium, Escherichia coli in wastewater. Fifty-two grab influent samples collected weekly from a wastewater treatment plant in Yamanashi Prefecture, Japan, between June 2023 and May 2024 were centrifuged, followed by DNA extraction and cdPCR. cdPCR was performed using the naica® system (Stilla Technologies). The 6-plex cdPCR assays showed strong performance. Among the 52 samples, 100 % of samples were positive for C. perfringens, 98 % for Salmonella spp., 56 % for C. jejuni, 25 % for C. coli, and 63 % for S. pyogenes, with concentrations ranging between 4.2 ± 0.3 to 7.5 ± 0.2 log10 copies/L. The concentration of C. perfringens was significantly higher than that of other pathogens (p < 0.05), indicating its dominance. Salmonella spp. had high detection rate, implying increased Salmonella infection in the population. Seasonal variation was not observed in any of FBD pathogens, except for the detection rate of C. coli. S. pyogenes concentrations were significantly higher in spring than in other seasons, agreeing with the trend of GAS pharyngitis cases in the catchment. In conclusion, the 6-plex cdPCR system is a valuable tool for comprehensive WS, offering significant implications for public health monitoring.
Collapse
Affiliation(s)
- Sadhana Shrestha
- Interdisciplinary Center for River Basin Environment, University of Yamanashi, 4-3-11 Takeda, Kofu, Yamanashi, 400-8511, Japan
| | - Bikash Malla
- Interdisciplinary Center for River Basin Environment, University of Yamanashi, 4-3-11 Takeda, Kofu, Yamanashi, 400-8511, Japan
| | - Eiji Haramoto
- Interdisciplinary Center for River Basin Environment, University of Yamanashi, 4-3-11 Takeda, Kofu, Yamanashi, 400-8511, Japan.
| |
Collapse
|
10
|
Lv P, Pei Y, Jiang Y, Wang Q, Liu Y, Qu M, Xu X, Chen M, Wang Y. Genomic insights into antibiotic-resistant non-typhoidal Salmonella isolates from outpatients in Minhang District in Shanghai. COMMUNICATIONS MEDICINE 2025; 5:228. [PMID: 40506473 PMCID: PMC12163078 DOI: 10.1038/s43856-025-00950-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 06/03/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Salmonella enterica (S. enterica) causes tens of thousands of cases of diarrheal disease worldwide each year. However, our understanding of the genome and transmission dynamics of S. enterica in Minhang District in Shanghai, China is still insufficient. This study is aimed to better understand the population structure, antibiotic resistance patterns, and evolution dynamics of local strains. METHODS We sequenced 458 S. enterica strains from outpatients at Minhang District Central Hospital in Shanghai, China, from 2012 to 2021. Bioinformatics analyses on antibiotic resistance genes, virulence factors, mobile genetic elements, pathogenic islands, and phylogenetic relationships were performed. RESULTS Here we show that two dominant serovars are S. Enteritidis and S. Typhimurium isolated from outpatients in Minhang District in Shanghai, China. A total of 40 serovars and 53 sequence types (STs) are identified, two S. Montevideo strains isolated in 2013 belong to a newly identified ST10844, which is firstly identified in Minhang District in Shanghai, China. More than half of the isolates show resistance to fluoroquinolones and beta-lactams. Notably, 259 (56.6%) of the 458 isolates exhibit a multidrug-resistant pattern. Third-generation cephalosporin resistance gene blaCTX-M-55 is identified in 15 (3.3%) isolates, and fluoroquinolone resistance gene qnrS1 is identified in 42 (9.2%) isolates, both of which are strongly correlated with IS26. Mutations of T57S in ParC and D87Y in GyrA are observed in 149 (32.5%) and 133 (29.0%) isolates, respectively. In addition, phylogenetic analysis confirms the presence of outbreaks caused by S. Enteritidis and S. Typhimurium, respectively. CONCLUSIONS These results suggest local expansion and evolution in Salmonella occurred in Shanghai, China, and the underlying emergence of the undefined multidrug-resistant clone. Our findings enlarge the knowledge of local epidemics of Salmonella, especially S. Enteritidis and S. Typhimurium in Shanghai, and provide a piece of useful baseline information for future whole-genome sequencing surveillance.
Collapse
Affiliation(s)
- Panpan Lv
- Research and Translational Laboratory of Acute Injury and Secondary Infection, and Department of Laboratory Medicine, Minhang Hospital, Fudan University, Shanghai, China
| | - Yuhang Pei
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yue Jiang
- Research and Translational Laboratory of Acute Injury and Secondary Infection, and Department of Laboratory Medicine, Minhang Hospital, Fudan University, Shanghai, China
| | - Qiang Wang
- Research and Translational Laboratory of Acute Injury and Secondary Infection, and Department of Laboratory Medicine, Minhang Hospital, Fudan University, Shanghai, China
| | - Yue Liu
- Department of Microbiology, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Mengqi Qu
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Xuebin Xu
- Department of Microbiology, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China.
| | - Mingliang Chen
- Research and Translational Laboratory of Acute Injury and Secondary Infection, and Department of Laboratory Medicine, Minhang Hospital, Fudan University, Shanghai, China.
| | - Yanan Wang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- Longhu Laboratory, Zhengzhou, Henan, China.
- Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
| |
Collapse
|
11
|
Weng J, Hu Y, Wang K, Chen Y, Yang Y, Xing B, Ma L, Wei Z. Minibinders targeting Staphylococcus aureus α-toxin exert protective effects in vivo. Int J Biol Macromol 2025:145215. [PMID: 40513733 DOI: 10.1016/j.ijbiomac.2025.145215] [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: 02/27/2025] [Revised: 06/09/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Staphylococcus aureus is a leading bacterial pathogen worldwide. It secrets α-toxin, a pore-forming cytotoxin that significantly contributes to the pathogenesis of pneumonia, skin necrosis, and lethal infections. Targeting α-toxin has emerged as a promising strategy for developing therapeutics against S. aureus infections. In this study, we employed a de novo computational design pipeline to generate minibinder proteins specifically targeting to α-toxin. Three top-ranked designs were expressed in E. coli, purified, and characterized. The minibinders exhibited thermostable α-helical structures and specific binding to α-toxin, with Binder2 showing the highest affinity (KD = 40.2 ± 3.1 μM). All designs effectively inhibited α-toxin's hemolytic and cytotoxic activities in vitro. Notably, Binder2 attenuated α-toxin-induced cytokine expression in A549 and THP-1 cells. Mechanistically, binders neutralized α-toxin cytolytic activity by blocking its cell binding and heptamer formation. In a murine model, co-injection of minibinders with α-toxin significantly extended the survival times of mice challenged with α-toxin, and Binder2 exhibited the most potent protective effects. These findings highlight Binder2 as a promising lead molecule for further development as an α-toxin antagonist. Our study demonstrates the potential of computationally designed minibinders as a novel therapeutic approach against S. aureus infections.
Collapse
Affiliation(s)
- Jun Weng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yang Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Keke Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Yuhua Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Yi Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Banbin Xing
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Lixin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Zigong Wei
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China.
| |
Collapse
|
12
|
Aggarwal SD, Toussaint J, Lees JA, Weiser JN. Colonization dynamics of Streptococcus pneumoniae are determined by polymorphisms in the BlpAB transporter. Infect Immun 2025; 93:e0006125. [PMID: 40387421 DOI: 10.1128/iai.00061-25] [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: 03/07/2025] [Accepted: 03/31/2025] [Indexed: 05/20/2025] Open
Abstract
Colonization of the human airways, the first step in the pathogenesis of Streptococcus pneumoniae (Spn), is the determining factor in the ecological spread of the bacterium. Since co-colonization by multiple strains is common, within-host bacterial competition contributes to the success of Spn strains. Competition both between and within strains is mediated by bacteriocin gene clusters, notably the quorum sensing-regulated bacteriocin-like peptide (blp) locus. A key component of this system is the BlpAB transporter that exports pheromones and bacteriocins expressed by the blp locus. However, ~75% of Spn strains lack a functional BlpAB transporter and instead rely on the paralogous ComAB transporter for this export, raising questions about the evolutionary persistence of BlpAB(+) strains. Using molecular barcoding, we demonstrate that BlpAB(+) and BlpAB(-) strains show major differences in population dynamics during colonization modeled in mice. The BlpAB(+) strains exhibit slower loss of clonal diversity as a consequence of intrastrain competition relative to their isogenic BlpAB(-). The contribution of a functional BlpAB transporter was then examined in an association study of >2,000 human carriage isolates from a highly colonized population. The median carriage duration was ~177 days longer for BlpAB(+) relative to BlpAB(-) strains. This increased duration of natural carriage correlates with a competitive advantage for BlpAB(+) strains when tested in the murine model. Thus, our work provides insight into how differences in the population dynamics of Spn mediated by bacterial competition impact host colonization.IMPORTANCESpn is a frequent colonizer of the human upper respiratory tract. Success during colonization is dictated by the arsenal of weapons these bacteria possess, which provides them with an advantage over their competitors. A key example includes the blp bacteriocins that are exported by the cell through both BlpAB and ComAB transporters. While most Spn strains lack a functional BlpAB, a subset of the strains retains it. Given this redundancy in export systems, our study questioned the evolutionary advantage of retaining BlpAB. Herein, we show that a functional BlpAB transporter causes a slower loss of clonal diversity in vivo. This correlates with longer Spn carriage duration in the human population and a competitive advantage during experimental co-colonization. Our work highlights the reasons behind the persistence of Spn with a functional BlpAB. These findings reveal how genetic variability in the blp locus shapes Spn colonization and evolutionary success.
Collapse
Affiliation(s)
- Surya D Aggarwal
- Department of Microbiology, New York University School of Medicine, New York, New York, USA
| | - Jacqueline Toussaint
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, United Kingdom
| | - John A Lees
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, United Kingdom
| | - Jeffrey N Weiser
- Department of Microbiology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
13
|
Vickers TJ, Buckley DP, Khatoon N, Sheikh A, Setu B, Berndsen ZT, Fleckenstein JM. Parenteral vaccination with recombinant EtpA glycoprotein impairs enterotoxigenic E. coli colonization. Infect Immun 2025; 93:e0060124. [PMID: 40310293 DOI: 10.1128/iai.00601-24] [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: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 05/02/2025] Open
Abstract
Enterotoxigenic E. coli (ETEC) causes hundreds of millions of cases of acute diarrheal illness in low- and middle-income regions, disproportionately in young children. To date, there is no licensed, broadly protective vaccine against these common but antigenically heterogeneous pathogens. One of the more highly conserved antigens of ETEC, EtpA, is an extracellular glycoprotein adhesin that preferentially binds to A blood group glycans on intestinal epithelia. EtpA contributes to increased severity of illness in A blood group individuals, elicits robust serologic and fecal antibody responses following infection, and has been associated with protection against subsequent infection. However, its utility as a protective antigen needs further examination. In the present studies, we examined whether parenteral vaccination with recombinant EtpA (rEtpA) could afford protection against intestinal colonization in a murine model of ETEC infection. Here, we demonstrate that intramuscular vaccination with rEtpA, adjuvanted with double mutant LT (dmLT), primes IgG predominant mucosal antibody responses to ETEC challenge. Notably, however, both antibody levels and avidity, as well as protection, were dependent on the vaccination schedule. Likewise, through electron microscopy polyclonal epitope mapping (EMPEM), we observed a different repertoire of epitopes targeted by antibodies after a more protracted vaccination schedule. Next, we explored the utility of IM immunization with alum-adjuvanted rEtpA. This elicited strong serologic and fecal IgG responses. Although accompanied by negligible IgA mucosal responses, EtpA alum-adjuvanted IM vaccination nevertheless protected against ETEC intestinal colonization. Collectively, these data suggest that EtpA could expand the portfolio of antigens targeted in ETEC subunit vaccine development.
Collapse
Affiliation(s)
- Tim J Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David P Buckley
- Department of Biochemistry, University of Missouri Columbia, Columbia, Missouri, USA
| | - Nazia Khatoon
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bipul Setu
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zachary T Berndsen
- Department of Biochemistry, University of Missouri Columbia, Columbia, Missouri, USA
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Infectious Diseases, Medicine Service, Veterans Affairs Saint Louis Health Care System, St. Louis, Missouri, USA
| |
Collapse
|
14
|
Jing H, Wang X, Zhao Z, Tang Y, Yuan Y, Liao J, Wei H, Chen Z, Gou Q, Zou Q, Hu R, Zhang X, Zhang J. Metallo-β-lactamase NDM-1 serves as a universal vaccine candidate for combatting antimicrobial resistance. Int J Biol Macromol 2025; 318:145158. [PMID: 40505900 DOI: 10.1016/j.ijbiomac.2025.145158] [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: 01/03/2025] [Revised: 05/21/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
The rapid emergence and spread of antimicrobial resistance have become critical global health issues, leading to significant morbidity and mortality worldwide. With the increase in resistance to multiple drugs, especially frontline clinical antibiotics, there is an urgent need for novel and effective alternative strategies. Herein, we developed a vaccine targeting the antimicrobial resistance enzyme NDM-1, which was first identified in Klebsiella pneumoniae and has quickly spread to other gram-negative bacteria. Our results demonstrate that NDM-1 primarily triggers a humoral immune response and effectively protects mice from lethal Klebsiella pneumoniae infection, as evidenced by increased survival rates, reduced bacterial loads, and decreased lung inflammation in mice. The specific antibodies generated were able to inhibit the enzymatic activity of NDM-1, bacterial growth, and exhibit opsonophagocytic activity against Klebsiella pneumoniae in vitro. Both active and passive immunization with NDM-1 showed an additive effect when combined with meropenem therapy. Furthermore, NDM-1 immunization induced cross-reactivity with NDM-1 variants, potentially providing broad protection against bacteria carrying different NDM genes. Additionally, heptamerization of NDM-1 improved its immunogenicity and protective efficacy in mice. These results highlight the potential of vaccine development based on antibiotic resistance candidates for broadly combatting antimicrobial resistance.
Collapse
Affiliation(s)
- Haiming Jing
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Xiaoqiong Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China; School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Yuanda Tang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Yue Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Jingwen Liao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Hongyu Wei
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Zhifu Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Qiang Gou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Renjian Hu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| | - Xiaoli Zhang
- Department of Clinical Hematology, College of Pharmacy, Army Medical University, Chongqing 400038, PR China.
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China.
| |
Collapse
|
15
|
Kuriu A, Ishikawa K, Tsuchiya K, Furuta K, Kaito C. Xenopus laevis as an infection model for human pathogenic bacteria. Infect Immun 2025; 93:e0012625. [PMID: 40310291 DOI: 10.1128/iai.00126-25] [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: 03/07/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
Animal infection models are essential for understanding bacterial pathogenicity and corresponding host immune responses. In this study, we investigated whether juvenile Xenopus laevis could be used as an infection model for human pathogenic bacteria. Xenopus frogs succumbed to intraperitoneal injection containing the human pathogenic bacteria Staphylococcus aureus, Pseudomonas aeruginosa, and Listeria monocytogenes. In contrast, non-pathogenic bacteria Bacillus subtilis and Escherichia coli did not induce mortality in Xenopus frogs. The administration of appropriate antibiotics suppressed mortality caused by S. aureus and P. aeruginosa. Strains lacking the agr locus, cvfA (rny) gene, or hemolysin genes in S. aureus, LIPI-1-deleted mutant of L. monocytogenes, which attenuate virulence within mammals, exhibited reduced virulence in Xenopus frogs compared with their respective wild-type counterparts. Bacterial distribution analysis revealed that S. aureus persisted in the blood, liver, heart, and muscles of Xenopus frogs until death. These results suggested that intraperitoneal injection of human pathogenic bacteria induces sepsis-like symptoms in Xenopus frogs, supporting their use as a valuable animal model for evaluating antimicrobial efficacy and identifying virulence genes in various human pathogenic bacteria.
Collapse
Affiliation(s)
- Ayano Kuriu
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazuya Ishikawa
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Kazuyuki Furuta
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chikara Kaito
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
16
|
Nonne F, Molfetta M, Belciug GF, Carducci M, Cianchi V, Zakroff C, Durante S, Zellmer C, Baker S, Stanton TD, Holt KE, Wyres K, Ravenscroft N, Gasperini G, Rossi O, Giannelli C, Berlanda Scorza F, Micoli F. The characterization of Klebsiella pneumoniae associated with neonatal sepsis in low- and middle-income countries to inform vaccine design. Commun Biol 2025; 8:898. [PMID: 40490475 DOI: 10.1038/s42003-025-08258-7] [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: 12/31/2024] [Accepted: 05/19/2025] [Indexed: 06/11/2025] Open
Abstract
Klebsiella pneumoniae is the leading cause of neonatal sepsis, strongly associated to antimicrobial resistance, with no vaccine available. K-antigens (KAg) have been identified as potential targets, but their diversity makes vaccine development challenging. Alternatively, the use of subcapsular O-antigens (OAg) raises questions about antibodies accessibility. We characterized clinical isolates from the BARNARDS study, designed to identify the burden of neonatal sepsis in low-middle income countries. Genomic prediction was verified through structural analysis of polysaccharides. Antibodies generated against common KAg and OAg bound all homologous organisms, regardless of specific polysaccharide structural features. Interestingly, anti-KAg antibodies exhibited bactericidal activity against a comparable number of isolates as anti-OAg antibodies. There was no association between polysaccharide characteristics and K. pneumoniae susceptibility to killing. Antibody cross-reactivity among different KAg was observed, together with extensive cross-reactivity among OAg antibodies. This study aids in defining an optimal vaccine composition to prevent neonatal sepsis caused by K. pneumoniae.
Collapse
Affiliation(s)
- Francesca Nonne
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | | | | | - Virginia Cianchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | | | | | - Caroline Zellmer
- The Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stephen Baker
- A*STAR Infectious Diseases Labs (A*STAR IDL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Thomas D Stanton
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Centre to Impact AMR, Monash University, Clayton, VIC, Australia
| | - Kathryn E Holt
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Kelly Wyres
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Centre to Impact AMR, Monash University, Clayton, VIC, Australia
| | - Neil Ravenscroft
- Department of Chemistry, University of Cape Town, Cape Town, South Africa
| | | | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - Carlo Giannelli
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | | |
Collapse
|
17
|
Kumar R, Kumar A, Kumar S. Sepsis in liver failure patients: Diagnostic challenges and recent advancements. World J Crit Care Med 2025; 14:101587. [PMID: 40491883 PMCID: PMC11891854 DOI: 10.5492/wjccm.v14.i2.101587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/19/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Acute liver failure (ALF) and acute-on-chronic LF (ACLF) are prevalent hepatic emergencies characterized by an increased susceptibility to bacterial infections (BI), despite significant systemic inflammation. Literature indicates that 30%-80% of ALF patients and 55%-81% of ACLF patients develop BI, attributed to immunological dysregulation. Bacterial sepsis in these patients is associated with adverse clinical outcomes, including prolonged hospitalization and increased mortality. Early detection of bacterial sepsis is critical; however, distinguishing between sterile systemic inflammation and sepsis poses a significant challenge due to the overlapping clinical presentations of LF and sepsis. Conventional sepsis biomarkers, such as procalcitonin and C-reactive protein, have shown limited utility in LF patients due to inconsistent results. In contrast, novel biomarkers like presepsin and sTREM-1 have demonstrated promising discriminatory performance in this population, pending further validation. Moreover, emerging research highlights the potential of machine learning-based approaches to enhance sepsis detection and characterization. Although preliminary findings are encouraging, further studies are necessary to validate these results across diverse patient cohorts, including those with LF. This article provides a comprehensive review of the magnitude, impact, and diagnostic challenges associated with BI in LF patients, focusing on novel advancements in early sepsis detection and characterization.
Collapse
Affiliation(s)
- Ramesh Kumar
- Department of Gastroenterology, All India Institute of Medical Sciences, Patna 801507, Bihar, India
| | - Abhishek Kumar
- Department of Gastroenterology, All India Institute of Medical Sciences, Patna 801507, Bihar, India
| | - Sudhir Kumar
- Department of Gastroenterology, All India Institute of Medical Sciences, Patna 801507, Bihar, India
| |
Collapse
|
18
|
Li Y, Liu Y, Yu X, Zhang F, Ma R, Wang R, Wang Z. Bactericidal effect of plasma-activated water generated by a novel super-potable plasma device as a novel antibacterial method: an in vitro study. BMC Oral Health 2025; 25:934. [PMID: 40481450 PMCID: PMC12144717 DOI: 10.1186/s12903-025-06217-6] [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: 10/11/2024] [Accepted: 05/20/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND A continuous risk from bacterial infection poses a major environmental and public health challenge. As an emerging strategy for inhibiting bacterial infections, plasma-activated water (PAW) has proved highly effective, environment-friendly, and non-drug resistant to many bacteria. However, the plasma device is too big and requires specialists to operate, which inevitably limits its real-life application. It has been confirmed that the bactericidal effect of traditional PAW would be significantly reduced in the organic-rich environments. To better resolve the limitations, a novel super-portable plasma device was applied, and the low concentration of H2O2 was added to enhance the bactericidal effect of PAW in the organ-rich environments. METHODS A super-portable plasma device, weighing only 1 kg, was applied to generate PAW. Staphylococcus aureus (S. aureus), Escherichia coli (E. coli) and Porphyromonas gingivalis (P. gingivalis) were selected as the test bacteria. Colony-forming units (CFU) counting and bactericidal rate were employed to evaluate the bactericidal effect of PAW. Optical emission spectroscopy (OES) was used to identify the major reactive species in PAW. Different concentration of H2O2 was added to PAW (H2O2/PAW), and the pH, oxidation-reduction potential (ORP), conductivity, H2O2 and NO content of HP/PAW were measured to explore the bactericidal mechanisms. CCK-8 assay was used to assess the cytotoxicity of 0.1% H2O2/PAW on human periodontal ligament fibroblasts (hPDLFs). RESULTS The 20 mL of PAW generated by the novel super-portable plasma device achieved the bactericidal rate of 98.94% against S. aureus (t = 10 min) and 99.66% (t = 20 min). When the PAW was prepared with a volume of 20 mL, activation times of 6 min and 8 min, and the treatment time was 30 min, the bactericidal rates all exceeded 98% against P. gingivalis. The addition of 0.1% H2O2 significantly enhanced the antibacterial effect of PAW in the presence of Bovine Serum Albumin (BSA) (p<0.05). The addition of 0.1% H2O2 lowered the pH, increased the ORP and conductivity, and raised the content of H2O2 and NO in the solution. Cytotoxicity evaluation indicated that 0.1% H2O2/PAW had moderate cytotoxicity only after 20 min of preparation and 48 h of treatment, while other preparation and treatment times showed mild or no cytotoxicity. CONCLUSIONS The PAW generated by the novel super-portable plasma device exhibited an excellent bactericidal effect. The addition of low concentrations of H2O2 to PAW, specifically 0.1% H2O2/PAW, maintained an effective antibacterial effect in complex environments in the presence of BSA with non-cytotoxicity.
Collapse
Affiliation(s)
- Yinglong Li
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Yu Liu
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Xin Yu
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Fang Zhang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
- Department of Stomatology, Beijing First Hospital of Integrated Chinese and Western Medicine, Beijing, 100026, China
| | - Ruonan Ma
- Zhengzhou Research Base, National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - RuiXue Wang
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Zuomin Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongti South Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
19
|
Rossato Viana A, Aytar EC, Pippi N, Santos D, Rhoden CRB, Stefanello Vizzotto B, Flores EMM, Passaglia Schuch A, Krause LMF. Chemical composition, in vitro and in silico activity of the methanolic extract derived from Vassobia breviflora against clinically relevant bacteria. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:463-478. [PMID: 39849319 DOI: 10.1080/15287394.2025.2453858] [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: 01/25/2025]
Abstract
This study aimed to identify chemical compounds derived from Vassobia breviflora methanolic extract using ESI-ToF-MS and their antioxidant potential activity utilizing the following methods: total phenols, DPPH, and ABTS•+. The MTT assay measured cytotoxic activity, while DCFH-DA and nitric oxide assays were employed to determine reactive oxygen species (ROS) and reactive nitrogen species (RNS) levels using African green monkey kidney (VERO) and human keratinocyte (HaCat) cell lines. The minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) were assessed in seven clinical isolates and nine ATCC strains. Biofilm inhibition was tested against four biofilm-forming strains. The antioxidant properties of the methanolic extract were identified as follows: 35.74 mg GAE/g (gallic acid equivalents)/g for total phenols, 10.5 µg/ml for DPPH, and 50.68 µmol trolox/µg for ABTS•+. The mean inhibitory concentration (IC50) values were 622.86 µg/ml (VERO) and 784.33 µg/ml (HaCat). These concentrations did not markedly alter levels of ROS and RNS. Conversely, Bacillus cereus β-hemolytic displayed higher sensitivity to the extract, with MIC of 64 µg/ml and MBC of 128 µg/ml. Enterococcus faecium exhibited the lowest biofilm formation among the tested bacteria. The studied plant exhibited activity against all bacterial strains at concentrations lower than the IC50 VERO and HaCat cells, suggesting potential for future studies. Data present a comprehensive molecular docking analysis against the HlyIIR protein (PDB ID: 2FX0) and determined antimicrobial and endocrine-modulating potentials. Notably, lancifodilactone I and nicandrin B demonstrated the strongest binding affinities, with binding energies of -9.8 kcal/mol and -8.3 kcal/mol, respectively, and demonstrated significant antimicrobial effects against B. cereus. In addition, several compounds showed potential interactions with nuclear receptors, indicating potential endocrine-modulating effects. These findings provide insights into developing target-specific antimicrobial therapies and endocrine-modulating agents.
Collapse
Affiliation(s)
- Altevir Rossato Viana
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Brazil
| | - Erdi Can Aytar
- Faculty of Agriculture, Department of Horticulture Uşak, Usak University, Türkiye
| | - Nickolas Pippi
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Brazil
| | - Daniel Santos
- Department of Chemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | | | | | | | - André Passaglia Schuch
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Brazil
| | | |
Collapse
|
20
|
Luo D, Xie W, Ma S, Wang L, Zhu J, Wang Z. A new perspective on the antimicrobial mechanism of linezolid against Staphylococcus aureus revealed by proteomics and metabolomics analysis. Int J Antimicrob Agents 2025; 65:107470. [PMID: 40049374 DOI: 10.1016/j.ijantimicag.2025.107470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 01/01/2025] [Accepted: 02/14/2025] [Indexed: 03/28/2025]
Abstract
Understanding bacterial responses to antimicrobials is crucial for identifying tolerance mechanisms and for developing new therapies. Using mass spectrometry-based metabolomics and proteomics, this study examines the response of Staphylococcus aureus to linezolid (LZD) treatment. Under LZD stress, significant fluctuations were observed in key metabolic pathways such as amino acid biosynthesis and the TCA cycle, alongside a general increase in ribosomal protein complexes. Additionally, LZD disrupted nucleotide metabolism, particularly affecting pyrimidine pathways. Combining LZD with the pyrimidine synthesis inhibitor leflunomide enhanced bactericidal effects both in vitro and in vivo, highlighting the importance of targeting pyrimidine biosynthesis to amplify the antimicrobial efficacy of protein inhibitors. These results underscore downstream metabolic processes as viable targets for synergistic drug combinations, suggesting a strategy to potentially improve the clinical effectiveness of LZD.
Collapse
Affiliation(s)
- Dan Luo
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Weile Xie
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Shiwei Ma
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Longlong Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Jianguo Zhu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhe Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China..
| |
Collapse
|
21
|
Liao Y, Zeng T, Guo X, Li X. Ferritin's role in infectious diseases: Exploring pathogenic mechanisms and clinical implications. New Microbes New Infect 2025; 65:101582. [PMID: 40230813 PMCID: PMC11995792 DOI: 10.1016/j.nmni.2025.101582] [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: 09/05/2024] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Ferritin, an iron storage protein, is crucial for maintaining iron metabolism balance throughout the body and serves as a key biomarker for evaluating the body's iron reserves. Reduced ferritin levels typically indicate iron deficiency, whereas elevated ferritin levels indicate an acute inflammatory response in infectious diseases. Recent research has established a significant link between elevated ferritin levels and disease severity and prognosis. The concept of hyperferritinemic syndrome has underscored ferritin's role as a pathogenic mediator. During infections, ferritin not only serves as a biomarker of inflammation but also exerts pro-inflammatory functions, which is a key factor in perpetuating the vicious pathogenic cycle. This review offers a comprehensive exploration of ferritin, covering its structural characteristics, regulatory mechanisms, and how diverse pathogens modulate ferritin. Understanding its pivotal role in infectious diseases is essential for identifying novel therapeutic prospects and enhancing disease management and prevention.
Collapse
Affiliation(s)
| | | | - Xiaoyan Guo
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xinhua Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| |
Collapse
|
22
|
Wu C, You Y, Yu D, Zhu YX, Lin H, Shi J. Selenide-Driven Reactive Oxygen Species Activation and Fe(II) Regeneration for Enhanced Nanocatalytic Antibacterial Therapeutics. Adv Healthc Mater 2025; 14:e2501021. [PMID: 40344503 DOI: 10.1002/adhm.202501021] [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: 03/05/2025] [Revised: 04/13/2025] [Indexed: 05/11/2025]
Abstract
Fenton-based nanocatalytic therapy has attracted widespread attention for its high efficiency and safety. Nevertheless, Fe2+ regeneration, as the rate-limiting step of Fenton reaction, hinders the ROS-induced oxidative killing. Herein, a Fe2+ auto-regeneration strategy is exemplified by 2D FeSe2 nanosheets to break the rate limitation of Fenton reaction and subsequently enhances the antibacterial oxidative damage via dual ROS generation pathways. To be specific, the Se species accelerate the Fe3+ reduction to maintain high ·OH productivity of Fe2+-mediated Fenton reaction, which is accompanied by the production of H2Se in the presence of H+. The H2Se further converts O2 into O2 ·- and synergistically breaks the oxidative threshold of bacteria, leading to irreversible bacterial death with glutathione depletion, lipid peroxidation, and membrane destruction. In summary, the FeSe2-mediated Fe2+ auto-regeneration and ROS self-production pathways largely elevate its oxidative killing capability, providing a potential ROS enhancement strategy for broad-spectrum nonantibiotic bacterial disinfection.
Collapse
Affiliation(s)
- Chenyao Wu
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, P. R. China
| | - Yanling You
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, P. R. China
| | - Dehong Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Ya-Xuan Zhu
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Han Lin
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, P. R. China
| | - Jianlin Shi
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
23
|
Chen H, Song S, Cui R, Feng YW, Ge P. Global trends in staphylococcus aureus-related lower respiratory infections from 1990 to 2021: findings from the 2021 global burden of disease report. Eur J Clin Microbiol Infect Dis 2025; 44:1455-1469. [PMID: 40186828 DOI: 10.1007/s10096-025-05111-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/17/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Lower respiratory infections (LRIs) represent a significant global health issue, especially affecting low- and middle-income countries. In this study, we explored the mortality and disability-adjusted life years (DALYs) associated with Staphylococcus aureus-related LRIs from 1990 to 2021, highlighting trends by age, sex, and Socio-Demographic Index (SDI). METHODS Data were derived from the 2021 Global Burden of Disease (GBD) database. Temporal trends in age-standardized mortality rates (ASMR) and disability-adjusted life years (DALYs) rates (ASDR) for S. aureus-related LRIs were analyzed based on the average annual percent change (AAPC), in terms of sex, 20-age groups, 21 regions, 204 countries, and 5 SDI quintiles. RESULTS In 2021, S. aureus-related LRIs contributed to 423,837 deaths (95% UI: 382,183-458,926), a 67.56% increase since 1990. In comparison, the global ASMR was 5.43 per 100,000 (95% UI: 4.89-5.90), and the ASDR was 156.80 per 100,000 (95% UI: 139.44-176.08), both exhibiting a declining trend compared to 1990. Rates were higher in low SDI regions, with Central Sub-Saharan Africa reporting the highest ASMR, while Eastern Europe had the lowest. Among the 204 countries analyzed, Zimbabwe recorded high ASMR and ASDR, at 24.84 (95% UI: 19.44-30.16) and 754.34 (95% UI: 591.05-923.06), respectively. CONCLUSIONS Although the global ASMR and ASDR decreased in 2021, the number of deaths from S. aureus-related LRIs significantly increased driven by the growing population and proportion of aged individuals. Additionally, the emergence of multidrug-resistant strains has made treatment more complex, particularly in low SDI regions, highlighting the urgent need for more targeted strategies, therapies, and vaccines.
Collapse
Affiliation(s)
- Hong Chen
- Department of Clinical Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Shuang Song
- Department of Clinical Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Rui Cui
- Department of Clinical Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Yong-Wang Feng
- Department of Clinical Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin, China.
- National Clinical Research Center for Cancer, Tianjin, China.
| | - Peng Ge
- Department of Clinical Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin, China.
- National Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
24
|
Li H, Yang Q, Liu W, Li R, Zhang D, Zhang G, Xu Y. Plant secretions and volatiles contribute to the evolution of bacterial antibiotic resistance in soil-crop system. J Environ Sci (China) 2025; 152:516-526. [PMID: 39617572 DOI: 10.1016/j.jes.2024.05.039] [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: 03/04/2024] [Revised: 05/05/2024] [Accepted: 05/23/2024] [Indexed: 12/10/2024]
Abstract
The exponential growth of antibiotic-resistant bacteria and antibiotic-resistant genes (ARGs) in soil-crop systems in recent years has posed a great challenge to ecological security and human health. While many studies have documented the residues of ARGs in soils and crops, but little is known about who drives the proliferation of ARGs in farming systems and what their underlying mechanisms are. Herein, we explored the occurrence and proliferating behavior of ARGs in soil-crop environments in terms of root secretions and plant volatiles. This review highlighted that plant root secretions and volatile organic compounds (VOCs) served as key substances mediating the development of antibiotic resistance in the soil-crop system. Still, there is controversy here as to plant root secretions promote the ARGs proliferation or inhibit. Some studies indicated that root secretions can suppress the colonization of ARGs, mainly attributed by the production of blunted metabolic enzymes and blocking of cellular exocytosis systems. Whereas the others have evidenced that root secretions can promote ARGs proliferation, primarily by altering the structure of microbial communities to influence species interactions and thus indirectly affect the proliferation of ARGs. Also, VOCs can act as molecular signals to convey antibiotic resistance information to their neighbors, which in turn drive the up-regulation of ARGs expression. Even so, the mechanism by which VOC-driven antibiotic resistance acquisition and proliferation need to be further probed. Overall, this review contributed to the development of products and technologies to impede the ARGs proliferation in agricultural environment.
Collapse
Affiliation(s)
- Houyu Li
- Agro-Environmental Protection Institute, Ministry of Agricultural and Rural Affairs, Tianjin 300191, China
| | - Qifan Yang
- Agro-Environmental Protection Institute, Ministry of Agricultural and Rural Affairs, Tianjin 300191, China
| | - Wei Liu
- Department F.A. Forel for Environmental and Aquatic Sciences, Faculty of Sciences, University of Geneva, Bvd. Carl-Vogt 66, 1211 Geneva, Switzerland
| | - Ruolan Li
- Agro-Environmental Protection Institute, Ministry of Agricultural and Rural Affairs, Tianjin 300191, China
| | - Dandan Zhang
- Agro-Environmental Protection Institute, Ministry of Agricultural and Rural Affairs, Tianjin 300191, China
| | - Guilong Zhang
- Agro-Environmental Protection Institute, Ministry of Agricultural and Rural Affairs, Tianjin 300191, China
| | - Yan Xu
- Agro-Environmental Protection Institute, Ministry of Agricultural and Rural Affairs, Tianjin 300191, China.
| |
Collapse
|
25
|
Frigoli M, Lowdon JW, Cleij TJ, Diliën H, Eersels K, van Grinsven B. Detection of antibiotic sulfamethoxazole residues in milk using a molecularly imprinted polymer-based thermal biosensor. Food Chem 2025; 476:143525. [PMID: 39999504 DOI: 10.1016/j.foodchem.2025.143525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025]
Abstract
Antibiotic resistance is a growing concern, partly due to inadequate inspections in the food safety chain. The accumulation of antibiotics like sulfamethoxazole (SMX) in animal products contributes to the rise of resistant microorganisms, posing a global health challenge. This work focuses on developing a thermal sensor to quickly and affordably detect SMX residues in milk samples. Molecularly imprinted polymers (MIPs) were synthesized and immobilized on an aluminum chip to measure thermal changes using the heat-transfer method (HTM). The sensor's detection limit in calcium chloride solutions was 261 ± 12 pmol L-1, well below regulatory limits for sulfonamides in dairy. The sensor also showed good selectivity when tested against antibiotics from different classes, and good performances in spiked milk samples. These results indicate that the thermal sensor provides a sensitive, low-cost alternative for detecting sulfamethoxazole traces in dairy products, contributing to improved food safety.
Collapse
Affiliation(s)
- Margaux Frigoli
- Sensor Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands.
| | - Joseph W Lowdon
- Sensor Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Thomas J Cleij
- Sensor Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Hanne Diliën
- Sensor Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Kasper Eersels
- Sensor Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| | - Bart van Grinsven
- Sensor Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, 6200, MD, Maastricht, the Netherlands
| |
Collapse
|
26
|
Rivera-Olivero IA, Kyriakidis NC. Latin America's Battle Against Antibiotic Resistance and Cancer: A Call to Action. Am J Med 2025; 138:928-929. [PMID: 39900232 DOI: 10.1016/j.amjmed.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/05/2025]
Affiliation(s)
- Ismar A Rivera-Olivero
- One Health Research Group. Facultad de Medicina. Universidad de Las Américas. Quito, Ecuador.
| | - Nikolaos C Kyriakidis
- Cancer Research Group, Facultad de Medicina, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
27
|
Diop M, Guitoula C, Tamouh AG, Youbong T, Daffé SMM, Ndoye M, Gueye MW, Wone F, Ngom M, Seck M, Youm N, Bassoum O, Lakhe NA, Ba PS, Faye A, Gning SB. Prevalence of bacterial infections and factors associated with death related to these infections in two medical departments of a tertiary hospital in Dakar, Senegal. IJID REGIONS 2025; 15:100623. [PMID: 40230497 PMCID: PMC11994947 DOI: 10.1016/j.ijregi.2025.100623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/16/2025]
Abstract
Objectives This study aimed to estimate the prevalence of bacterial infections and identify the factors associated with death related to these infections in the internal medicine (Brévié) and infectious diseases departments (Boufflers) of Dakar Principal Hospital. Methods This cross-sectional study was carried out from January 1 to December 31, 2023, including patients with bacterial infections hospitalized in Boufflers and Brévié. Multivariate logistic regression was used to identify factors associated with death. Results Out of 1,085 hospitalized patients, 181 (16.7%) had bacterial infections. The mean age was 60±18 years, with a sex ratio of 1.08. Urinary tract infection (37%) was the most represented clinical presentation. Bacteria were isolated in 123 patients (68%), with Escherichia coli (39%), Staphylococcus aureus (12%), and Klebsiella pneumoniae (12%) as the most represented species. Of the 140 identified bacteria, 78 (55.7%) were multidrug-resistant. The death rate was 15%. Multivariate analysis showed that age ≥ 65 years (odds ratio [OR] = 3.2; 95% confidence interval [CI] 1.2-9.5), prior hospitalization (OR = 2.9; 95% CI 1.1-8.5), and hemoglobin levels between 3.4 g/dl and 7 g/dl (OR = 11.7; 95% CI 2.5-60) or between 8 g/dl and 11 g/dl (OR = 4.9; 95% CI 1.6-18.4) compared with levels ≥11 g/dl were associated with death. Conclusions This study showed a high prevalence and mortality rate of bacterial infections in the internal medicine and infectious diseases departments, particularly among older adult patients, those with a history of hospitalization, or those with low hemoglobin levels.
Collapse
Affiliation(s)
- Moustapha Diop
- Department of Infectious and Tropical Diseases, Dakar Principal Hospital, Dakar, Senegal
| | - Chancia Guitoula
- Private Institute of Medical Training and Research, Dakar, Senegal
| | | | - Tracie Youbong
- Department of Infectious and Tropical Diseases, Dakar Principal Hospital, Dakar, Senegal
| | | | - Maguette Ndoye
- Laboratories Federation, Dakar Principal Hospital, Dakar, Senegal
| | | | - Fatimata Wone
- Department of Infectious and Tropical Diseases, Dalal Jamm Hospital, Dakar, Senegal
| | - Mor Ngom
- Laboratories Federation, Dakar Principal Hospital, Dakar, Senegal
| | - Mamadou Seck
- Department of Infectious and Tropical Diseases, FANN Teaching Hospital, Dakar, Senegal
| | - Nogaye Youm
- Department of Infectious and Tropical Diseases, Dakar Principal Hospital, Dakar, Senegal
| | - Oumar Bassoum
- Health and Development Institute, Cheikh Anta DIOP University, Dakar, Senegal
| | - Ndèye Aissatou Lakhe
- Department of Infectious and Tropical Diseases, FANN Teaching Hospital, Dakar, Senegal
| | - Papa Samba Ba
- Department of Infectious and Tropical Diseases, Dakar Principal Hospital, Dakar, Senegal
| | - Adama Faye
- Health and Development Institute, Cheikh Anta DIOP University, Dakar, Senegal
| | | |
Collapse
|
28
|
Heidari M, Tabatabaei-Malazy O, Jahani Z, Amini MR, Dastjerdi MV. A review of advancements in antiseptics for wound care in diabetic and non-diabetic patients. J Diabetes Metab Disord 2025; 24:101. [PMID: 40224527 PMCID: PMC11985748 DOI: 10.1007/s40200-025-01607-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/09/2025] [Indexed: 04/15/2025]
Abstract
Wounds affect many people and require a considerable annual cost to manage. Wound infections significantly delay the healing process, particularly in individuals with diabetes mellitus, due to impaired immunity and microvascular complications. The use of antiseptics is considered a way to reduce this problem. The study aims to assess the different antiseptic categories frequently employed in wound management, focusing on identifying and understanding their unique features. A comprehensive review of PubMed, Scopus, and EMBASE databases identified key antiseptics, including isopropyl alcohol, chlorhexidine, polyhexanide, octenidine, povidone-iodine, hypochlorous acid, silver-based products, hydrogen peroxide, triclosan, and benzalkonium chloride. These antiseptics exhibit varying efficacies and cytotoxicity profiles, necessitating tailored usage to optimize healing while preventing antimicrobial resistance. The primary indication for antiseptics is the prevention of Surgical Site Infections (SSIs), as recommended by guidelines. For diabetic foot ulcers, the strongest evidence supports the use of hypochlorous acid. There are no universal recommendations for antiseptic use; their application depends on specific circumstances. This review highlights the need for evidence-based, condition-specific antiseptic strategies to address unique patient needs effectively. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-025-01607-7.
Collapse
Affiliation(s)
- Mohammadreza Heidari
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ozra Tabatabaei-Malazy
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Jahani
- Department of Infectious Disease, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Amini
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Institute, No.10, Jalal Al-e-Ahmad Ave., North Kargar Ave, Tehran, Iran
| | - Marzieh Vahid Dastjerdi
- Department of Obstetrics and Gynecology, School of Medicine, Arash Women’s Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Melby MK, Mylabathula S, Azad MB, Turner S, Geva-Zatorsky N, Tropini C, Manus MB, Blaser M, Nichter M. Public Health Microbiome Curriculum: Looking Below the Tip of the Iceberg for Approaches to Population Health. Microb Biotechnol 2025; 18:e70160. [PMID: 40515726 DOI: 10.1111/1751-7915.70160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 06/16/2025] Open
Abstract
We discuss the opportunity for public health microbiome curricula to bridge the gaps in knowledge that exist between microbiome researchers and the lay public. We propose equipping public health professionals, important facilitators of public outreach and behaviour change, with three public health curriculum modules focused on breastfeeding, antibiotics and diet. These modules shift the focus from microbes as pathogens to potential partners in promoting health across the life course. Current public health messages cover only the 'tip of the iceberg' in exploring mechanisms, and this microbiome curriculum dives below the surface to provide fresh perspectives. These microbiome insights allow us to move beyond a focus on microbes as pathogens to understand the numerous collaborative roles played by the microbiome in producing health, and the upstream factors influencing the microbiome, thereby offering mechanistic insights that can be harnessed for public health education.
Collapse
Affiliation(s)
- Melissa K Melby
- CIFAR Humans & the Microbiome Program, Toronto, Canada
- Department of Anthropology, University of Delaware, Newark, Delaware, USA
| | - Swapna Mylabathula
- CIFAR Humans & the Microbiome Program, Toronto, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Meghan B Azad
- CIFAR Humans & the Microbiome Program, Toronto, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sarah Turner
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Naama Geva-Zatorsky
- CIFAR Humans & the Microbiome Program, Toronto, Canada
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Rappaport Technion Integrated Cancer Center (RTICC), Technion-Israel Institute of Technology, Haifa, Israel
| | - Carolina Tropini
- CIFAR Humans & the Microbiome Program, Toronto, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Melissa B Manus
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- Department of Anthropology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Martin Blaser
- CIFAR Humans & the Microbiome Program, Toronto, Canada
- Departments of Medicine and Pathology & Laboratory Medicine RBHS, Robert Wood Johnson Medical School, Rutgers University, Rutgers, New Jersey, USA
| | - Mark Nichter
- CIFAR Humans & the Microbiome Program, Toronto, Canada
- School of Anthropology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
30
|
Vilela FP, Rodrigues DDP, Allard MW, Falcão JP. First characterization of the resistome, virulome and genomic diversity of Salmonella enterica serovar Inganda: a rare, clinically-related and drug susceptible serovar. Curr Genet 2025; 71:13. [PMID: 40448871 DOI: 10.1007/s00294-025-01317-w] [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: 02/26/2025] [Revised: 05/06/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025]
Abstract
Non-typhoid Salmonella are among the main causes of foodborne diseases worldwide. However, information on rare serovars is scarce, limiting the understanding of their prevalence, distribution and pathogenesis. Salmonella enterica serovar Inganda (S. Inganda) is a rare non-typhoid serovar. Considering the few existing reports, and the current use of genomics, this study characterized for the first time the antimicrobial resistance, pathogenic potential and diversity of S. Inganda genomes worldwide. A S. Inganda strain from human feces in 2018 in Brazil (SI264) had its resistance determined against 18 antimicrobials by disk-diffusion and had its genome sequenced. S. Inganda publicly available genomes (n = 12) were analyzed for genotypic resistance, stress and virulence genes, plasmids, pathogenicity islands, prophages, Multi-Locus Sequence Typing (MLST), core-genome MLST (cgMLST), and single-nucleotide polymorphisms (SNPs). SI264 showed no phenotypic resistance. All 12 S. Inganda genomes harbored genes or mutations for aminoglycoside (aac(6')-Iaa), quinolone (parC Thr57→Ser), and acid (asr) resistance, multi-drug efflux systems (mdsAB), and gold tolerance (golST). One genome from US harbored pKPC-CAV1321 plasmid. Nine pathogenicity islands, 174 Salmonella virulence genes, and 17 prophages were found in different frequencies. Although a great genomic diversity was noticed, S. Inganda genomes from US and UK were closely related. In conclusion, genomic analyses were able to characterize the current available genomes of S. Inganda strains mostly as genetically diverse, susceptible to antimicrobials, and potentially acid and heavy metal resistant. The presence of numerous virulence features also suggested their pathogenic potential, especially among clinical strains, and reinforced the importance to better characterize rare non-typhoid serovars.
Collapse
Affiliation(s)
- Felipe Pinheiro Vilela
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto - USP, Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Av. do Café, s/n. Bloco S - Sala 41, Ribeirão Preto, SP, 14040-903, Brazil
| | | | - Marc William Allard
- Division of Microbiology, Office of Regulatory Science, Human Foods Program, U.S. Food and Drug Administration, College Park, MD, USA
| | - Juliana Pfrimer Falcão
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto - USP, Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Av. do Café, s/n. Bloco S - Sala 41, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
31
|
Ramay BM, Caudell MA, Castillo C, Grajeda L, Santos LF, Romero JC, Lopez MR, Omulo S, Ning MF, Palmer GH, Smith RM, Herzig CTA, Styczynski A, Cordon-Rosales C, Call DR. Risk factors associated with community colonization of extended-spectrum cephalosporin-resistant Enterobacterales from an antibiotic resistance in communities and hospitals (ARCH) study, Guatemala. Sci Rep 2025; 15:18925. [PMID: 40442174 PMCID: PMC12122697 DOI: 10.1038/s41598-025-03379-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
Colonization with extended-spectrum cephalosporin-resistant Enterobacterales (ESCrE) in communities may contribute to proliferation of resistance genes and drug-resistant community and hospital infections. Previous work in the Western Highlands of Guatemala found that approximately 46% of the population is colonized with these bacteria, setting the stage to identify factors that are associated with increased odds of ESCrE colonization. Stool samples and questionnaire data were collected from randomly selected participants in the catchment area of the third largest tertiary hospital in Guatemala. Logistic regression path analysis was used for this cross-sectional study to identify potential direct and indirect risk factors for colonization with ESCrE. Participants (N = 951) had a higher odds of ESCrE colonization if they had exposure to a healthcare facility within 30 days of enrollment (OR: 2.12, 95% CI = 1.19-3.77), if they resided in urban areas (OR: 1.93, 95% CI 1.09-3.42), if they did not have a service to remove household trash (OR: 1.99, 95% CI 1.11-3.58), and if the household reported drinking water from non-bottled sources (OR:1.53, 95% CI 1.0-2.33). Antibiotic self-medication was not significantly associated with the risk of colonization (OR: 1.16, 95% CI 0.65-2.06). Multiple transmission-related factors were associated with increased likelihood of ESCrE colonization, but the cross-sectional nature of this study does not distinguish factors that are correlated with an individual's risk for colonization whence exposed. Assessing risk factors associated with colonization with antibiotic resistant bacteria may be useful for identifying mitigation strategies and evaluating the effectiveness of interventions against antibiotic resistance in community settings.
Collapse
Affiliation(s)
- Brooke M Ramay
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA.
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala.
| | - Mark A Caudell
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Carmen Castillo
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Laura Grajeda
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Lucas F Santos
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Juan Carlos Romero
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Maria Renee Lopez
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Sylvia Omulo
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
- Washington State University Global Health-Kenya, Nairobi, Kenya
| | - Mariangeli Freitas Ning
- Central America Regional Office, U.S. Centers for Disease Control and Prevention, Guatemala City, Guatemala
| | - Guy H Palmer
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Rachel M Smith
- Division of Healthcare Quality Promotion, U.S. Centers for Disease Control and Prevention, Atlanta, USA
| | - Carolyn T A Herzig
- Division of Healthcare Quality Promotion, U.S. Centers for Disease Control and Prevention, Atlanta, USA
| | - Ashley Styczynski
- Division of Healthcare Quality Promotion, U.S. Centers for Disease Control and Prevention, Atlanta, USA
| | - Celia Cordon-Rosales
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Douglas R Call
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| |
Collapse
|
32
|
Liu L, Shi J, Wang H, Du H, Yang J, Wei K, Zhou Z, Li M, Huang S, Zhan L, Li G, Lv Y, Shen H, Cai W. The characteristics of tissue microbiota in different anatomical locations and different tissue types of the colorectum in patients with colorectal cancer. mSystems 2025:e0019825. [PMID: 40422085 DOI: 10.1128/msystems.00198-25] [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: 02/10/2025] [Accepted: 04/27/2025] [Indexed: 05/28/2025] Open
Abstract
The gut microbiota is intricately associated with the onset and progression of colorectal cancer (CRC), leading to significant interest in developing prevention and treatment strategies that leverage gut microbiota. In this study, we collected 57 samples from 19 CRC patients, comprising cancerous tissue, paracancerous tissue, and normal mucosa. Utilizing metagenomic sequencing and bioinformatics analysis, we identified differences in the microbiomes and their functional characteristics across the various tissue types. The results indicated that species such as Alistipes putredinis were predominantly found in normal tissues, while Pseudomonas putida was enriched in paracancerous tissue, and Malassezia restricta was prevalent in cancerous tissues. Furthermore, the microbial functions exhibited variability among the different tissue types. Random forest analysis suggested that Moraxella osloensis may be implicated in the onset and progression of colorectal cancer. We also classified the patients into three subgroups based on the anatomical location of the colorectum: right-sided colon, left-sided colon, and rectum. The subgroup analysis revealed that the microbiota enriched in normal mucosa and paracancerous tissue varied across different anatomical sites. These findings not only elucidate the characteristics of the microbiomes in the normal mucosa, paracancerous tissue, and cancerous tissues of CRC patients, thereby providing new potential targets for clinical diagnosis and treatment, but also contribute to the existing microbiome data pertinent to CRC research.IMPORTANCEThis study provides crucial insights into the relationship between gut microbiota and colorectal cancer (CRC) by analyzing microbial communities in different tissue types and anatomical locations of CRC patients. We identified distinct microbial signatures, such as Alistipes putredinis in normal tissues and Malassezia restricta in cancerous tissues, indicating location-specific microbiomes with unique functional attributes. These findings suggest potential new biomarkers or therapeutic targets for CRC. The observed microbiota variations among right-sided colon, left-sided colon, and rectum cancers underscore the heterogeneity of CRC, pointing toward more personalized treatment strategies. By enhancing our understanding of the microbiome's role in CRC, this research paves the way for innovative diagnostic tools and targeted therapies tailored to individual patient profiles. This work is essential for advancing clinical approaches to CRC management.
Collapse
Affiliation(s)
- Lei Liu
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Jianguo Shi
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Hui Wang
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Hansong Du
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Jia Yang
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Wei
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Zhuohui Zhou
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Moli Li
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Shuai Huang
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lifang Zhan
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| | - Guolong Li
- School of Life Sciences, Hubei University, Wuhan, Hubei, China
| | - Yongling Lv
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
- School of Life Sciences, Hubei University, Wuhan, Hubei, China
| | - Hexiao Shen
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
- School of Life Sciences, Hubei University, Wuhan, Hubei, China
| | - Wei Cai
- Department of Gastrointestinal Surgery and Intestinal Microenvironment Treatment Center, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Provincial Engineering Research Center of Intestinal Microecological Diagnostics, Therapeutics, and Clinical Translation, Wuhan, Hubei, China
| |
Collapse
|
33
|
Fredes-García D, Jiménez-Rodríguez J, Piña-Iturbe A, Caballero-Díaz P, González-Villarroel T, Dueñas F, Wozniak A, Adell AD, Moreno-Switt AI, García P. Development of a robust FT-IR typing system for Salmonella enterica, enhancing performance through hierarchical classification. Microbiol Spectr 2025:e0015925. [PMID: 40422737 DOI: 10.1128/spectrum.00159-25] [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: 01/15/2025] [Accepted: 03/22/2025] [Indexed: 05/28/2025] Open
Abstract
Salmonella enterica is a leading cause of foodborne illnesses globally, with significant mortality rates, especially among vulnerable populations. Traditional serotyping methods for Salmonella are accurate but expensive, resource-intensive, and time-consuming, necessitating faster and more reliable alternatives. This study evaluates the IR Biotyper, a Fourier-transform infrared spectroscopy system, in differentiating Salmonella serovars. We assessed 458 isolates of nine Salmonella serovars (Infantis, Enteritidis, Typhimurium, I,4,[5],12:i:-, Montevideo, Agona, Thompson, Panama, and Abony) from diverse sources. The IR Biotyper was used to acquire spectra from these isolates. Machine learning algorithms, including support vector machines, were trained to classify the isolates. The accuracy of classifiers was validated using a validation set to determine sensitivity, specificity, positive predictive value, and negative predictive value. Initial classifiers showed high accuracy for Abony, Agona, Enteritidis, and Infantis serovars, with sensitivities close to 100%. However, classifiers for S. Typhimurium, S. Panama, and S. Montevideo exhibited lower performance. Implementing a hierarchical classification system enhanced the accuracy of serogroup O:4 serovars, demonstrating that this approach offers a robust framework for Salmonella serovar identification. The hierarchical system enables progressive refinement of classification, minimizing misclassifications by focusing on serogroup-specific features, making it adaptable to complex data sets and diverse serovars. The IR Biotyper demonstrates high potential for rapid and accurate Salmonella serovar identification. This study supports its implementation as a cost-effective, high-throughput tool for pathogen typing, enhancing real-time epidemiological surveillance, and guiding treatment strategies for salmonellosis. This method establishes a robust and scalable framework for advancing Salmonella serotyping practices across clinical, industrial, and public health domains by leveraging hierarchical classification.IMPORTANCEEarly and accurate identification of Salmonella serovars is extremely important for epidemiological surveillance, public health, and food safety. Traditional serotyping is very successful but is laborious and costly. In this study, we demonstrate the promise of Fourier-transform infrared spectroscopy together with machine learning as a means for Salmonella serotyping. Using hierarchical classification, we attain optimal serovar identification accuracy, particularly for challenging-to-type serogroups. Our findings recognize the IR Biotyper as a high-throughput, scalable pathogen typing solution that offers real-time data that can enable enhanced outbreak response and prevention of foodborne disease. The approach bridges the gap between traditional microbiological practice and sophisticated analytical technology, the path to more effective, cost-saving interventions in the clinical, industrial, and regulatory settings. Application of these technologies can significantly improve Salmonella surveillance-control and Public Health outcomes.
Collapse
Affiliation(s)
- Diego Fredes-García
- Escuela de Medicina Veterinaria, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javiera Jiménez-Rodríguez
- Departamento de Laboratorios Clínicos, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro Piña-Iturbe
- Escuela de Medicina Veterinaria, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Caballero-Díaz
- Escuela de Medicina Veterinaria, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Fernando Dueñas
- Escuela de Medicina Veterinaria, Centro de Estudios e Investigación en Salud y Sociedad, Facultad de Ciencias Médicas, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Aniela Wozniak
- Departamento de Laboratorios Clínicos, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Aiko D Adell
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la vida, Universidad Andrés Bello, Santiago, Chile
| | - Andrea I Moreno-Switt
- Escuela de Medicina Veterinaria, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia García
- Departamento de Laboratorios Clínicos, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
34
|
Sun J, Zhu Y, Huang D, Li L, Pan M, Li F, Ma C. Burden of female diseases among adolescents and young adults aged 10-24 years in South Asia and Sub-Saharan Africa, 1990-2021: a systematic analysis from the Global Burden of Disease Study 2021. J Adv Res 2025:S2090-1232(25)00371-6. [PMID: 40425083 DOI: 10.1016/j.jare.2025.05.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/28/2025] [Accepted: 05/23/2025] [Indexed: 05/29/2025] Open
Abstract
INTRODUCTION Female diseases pose significant challenges in South Asia and Sub-Saharan Africa, particularly among adolescent girls and young women, who often receive insufficient attention. OBJECTIVES To report patterns and trends of female diseases among adolescent girls and young females aged 10-24 years in South Asia and Sub-Saharan Africa from 1990 to 2021. METHODS We used data from the Global Burden of Disease Study 2021 for 51 countries in South Asia and Sub-Saharan Africa between 1990 and 2021. Joinpoint Regression was used to calculate annual average percentage changes and 95 % confidence intervals to quantify temporal trends. RESULTS In 2021, South Asia and Sub-Saharan Africa had high mortality rates of maternal disorders of 6.04 (95 % uncertainty intervals 5.02, 7.39) and 17.69 (14.37, 21.78) per 100,000 population, respectively. The mortality rates for female cancers were approximately 0.98 in both regions, and the incidence rates for gynecological diseases were 16472.83 and 14480.99, per 100,000 population, respectively. From 1990 to 2021, there was an increasing trend in the number of maternal disorder deaths in Sub-Saharan Africa, as well as in all metric rates for most female cancers in both regions, and disability-adjusted life years, prevalence, and incidence rates for gynecological diseases in South Asia. Several female diseases varied across countries and were increasingly affecting younger adolescents aged 10-14 years in both regions. Although countries with lower Socio-demographic Index had a heavier burden of female diseases, no significant association was observed between the Universal Health Coverage effective coverage index and death rates for female cancers or gynecological diseases. CONCLUSIONS The burden of female diseases remains high among young females in South Asia and Sub-Saharan Africa, with younger adolescents being particularly affected. This underscores the urgent need for targeted interventions and increased investment in healthcare infrastructure to reduce the burden of female diseases in these regions.
Collapse
Affiliation(s)
- Jiahong Sun
- Department of Preventive Medicine, School of Public Health, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808 Guangdong, China
| | - Yongliang Zhu
- Department of Epidemiology and Health Statistics, School of Public Health, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Danyi Huang
- Department of Epidemiology and Health Statistics, School of Public Health, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Liuqing Li
- Department of Preventive Medicine, School of Public Health, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808 Guangdong, China
| | - Mengna Pan
- Department of Epidemiology and Health Statistics, School of Public Health, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Fei Li
- Department of Childcare and Wellness, the First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523710, China
| | - Chuanwei Ma
- Department of Epidemiology and Health Statistics, School of Public Health, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523808, China.
| |
Collapse
|
35
|
Sharma N, Baruah DJ, Duarah R, Yadav A, Bora D, Saikia R, Jarugala J, Das MR. Smart-Sandwich: A Thin Flexible Sensing Device Based on an Agarose-Chitosan-Agarose (ACA) Triple-Layer Biofilm for Onsite Monitoring of Escherichia coli. ACS Sens 2025; 10:3504-3514. [PMID: 40331915 DOI: 10.1021/acssensors.5c00038] [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: 05/08/2025]
Abstract
Portable, cost-effective, and durable thin-film sensors are essential for real-time E. coli detection, ensuring safe drinking water and public health protection. In this work, we developed a solid-state, flexible sensing device using an agarose-chitosan-agarose (ACA) sandwich biofilm for the selective colorimetric detection of E. coli in water. The chitosan in ACA biofilm functions as an artificial enzyme, exhibiting peroxidase-like activity, which catalyzes the oxidation of 3,3',5,5'-tetramethylbenzidine (TMB) in the presence of hydrogen peroxide (H2O2). Upon incubating the ACA biofilm with E. coli, the agarose layers undergo enzymatic degradation by the β-galactosidase enzyme produced by E. coli. The degradation takes place due to the cleavage of β-1,4-glycosidic bonds. This exposes the underlying chitosan layer which enhances the catalytic activity, triggering a visible color change due to TMB oxidation within 30 min. The device achieves a highly sensitive detection limit of 6.8 CFU/mL, with excellent accuracy in real samples, further supported by android-based, smartphone-assisted detection. The developed solid-state, flexible ACA biofilm offers a novel, rapid, and reliable solution for onsite E. coli detection, combining sensitivity, stability, and ease of use.
Collapse
Affiliation(s)
- Nidhi Sharma
- Materials Sciences Group, Coal, Energy and Materials Sciences Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam ,India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Diksha J Baruah
- Materials Sciences Group, Coal, Energy and Materials Sciences Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam ,India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rituparna Duarah
- Materials Sciences Group, Coal, Energy and Materials Sciences Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam ,India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Archana Yadav
- Centre for Biotechnology, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam ,India
| | - Dipjyoti Bora
- Materials Sciences Group, Coal, Energy and Materials Sciences Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam ,India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ratul Saikia
- Centre for Biotechnology, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam ,India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jayaramudu Jarugala
- Materials Sciences Group, Coal, Energy and Materials Sciences Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam ,India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manash R Das
- Materials Sciences Group, Coal, Energy and Materials Sciences Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam ,India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
36
|
DeLeo FR, Freedman B, Porter AR, Opoku-Temeng C, Kobayashi SD, Chen L, Kreiswirth BN. Evaluation of capsule polysaccharide (CPS)-specific antibodies for broad recognition of prominent multidrug-resistant Klebsiella pneumoniae. Microbiol Spectr 2025:e0333824. [PMID: 40401961 DOI: 10.1128/spectrum.03338-24] [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: 12/20/2024] [Accepted: 04/24/2025] [Indexed: 05/23/2025] Open
Abstract
Klebsiella pneumoniae is primarily an opportunistic pathogen known for causing healthcare-associated infections in individuals with significant risk factors and comorbidities. These bacteria are typically multidrug-resistant (MDR), a phenotype conferred in part by the production of extended-spectrum beta-lactamases and/or carbapenemases. By comparison, so-called hypervirulent K. pneumoniae (hvKp) are defined by their ability to cause severe community-acquired infections in otherwise healthy individuals. Although hvKp lineages have historically not been MDR, there has been a recent emergence of strains with both hypervirulence and multidrug resistance phenotypes. Treatment of infections caused by MDR K. pneumoniae can be difficult, and new preventative measures are needed. As a step toward the development of a vaccine directed to prevent or moderate infections caused by these pathogens, we tested the ability of capsule polysaccharide (CPS) derived from eight selected K. pneumoniae capsule types (KLs) to elicit rabbit antibodies that recognize important KLs isolated from human infections. Seventy-one out of 84 (84.5%) contemporary K. pneumoniae clinical isolates tested were recognized by CPS-specific rabbit antisera. There was the unexpected binding of the antibodies to some isolates with KLs not included in the CPS-antigen cocktails. Notably, rabbit IgG purified from CPS-specific antisera promoted and/or enhanced human PMN bactericidal activity toward all but one of the selected clinical isolates that were not killed by PMNs outright (in the absence of specific antibody). These data provide support to the idea that a CPS-antigen cocktail could be developed to protect against the K. pneumoniae KLs that are the most frequent cause of human infections.IMPORTANCEKlebsiella pneumoniae is among the leading causes of death by infectious agents. Many of the prominent K. pneumoniae lineages are resistant to multiple classes of antibiotics, and options for treatment are limited. New countermeasures that prevent infections are needed. Here we tested the ability of capsule polysaccharide (CPS) antigen mixtures (or cocktails) to elicit rabbit antibodies that recognize K. pneumoniae from a large collection of extended-spectrum beta-lactamase-producing clinical isolates. Importantly, these antibodies had the ability to promote opsonophagocytic killing by human PMNs. Our results provide proof-of-concept for a CPS vaccine cocktail approach that could be developed to prevent infections caused by the most important K. pneumoniae lineages.
Collapse
Affiliation(s)
- Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Brett Freedman
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Adeline R Porter
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Clement Opoku-Temeng
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Scott D Kobayashi
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Liang Chen
- Division of Clinical and Translational Therapeutics, Department of Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Barry N Kreiswirth
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| |
Collapse
|
37
|
Jurado-Martín I, Tomás-Cortázar J, Rezk N, Hou Y, Saínz-Mejías M, Bruce R, Startseva M, Ma C, McClean S. The novel antigen, lipopolysaccharide export protein LptH, protects mice against Pseudomonas aeruginosa acute pneumonia in monovalent and multivalent vaccines. Vaccine 2025; 56:127145. [PMID: 40262371 DOI: 10.1016/j.vaccine.2025.127145] [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: 03/07/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen that is a leading cause of morbidity and mortality worldwide in susceptible patients, particularly in those with respiratory disorders. The rising prevalence of multidrug-resistant strains and the failure of previous P. aeruginosa vaccine candidates in clinical trials highlight the urgent need to investigate novel vaccine antigens. In this study, we evaluated the protective potential of two antigen candidates, LptH and OprM, previously identified based on their involvement in host-cell attachment in a murine acute pneumonia model. Recombinant Escherichia coli BL21 clones overexpressing these proteins showed 8.8- and 3.5-fold increased attachment to 16HBE14o- cells in vitro, confirming their role in host-cell attachment. Immunisation with rLptH significantly reduced bacterial burden in the lungs by 1.12 log10 CFU and improved animal welfare scores compared to adjuvant-only controls. Serological and immunophenotyping analyses revealed that the monovalent rLptH vaccine stimulated antigen-specific IgG1 and IgG2c isotype production, and enhanced IFN-γ and IL-17 recall responses in the spleen. Moreover, a trivalent vaccine comprising rLptH and two other P. aeruginosa antigens, rFtsZ, and rOpmH, achieved a 2.33 log10 CFU reduction in lung bacterial burden, and 1.85 log10 CFU reduction in dissemination. These encouraging findings support the potential of LptH as a promising antigen for the development of a protective multivalent vaccine against P. aeruginosa infections.
Collapse
Affiliation(s)
- Irene Jurado-Martín
- School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Julen Tomás-Cortázar
- School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Nouran Rezk
- School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Yueran Hou
- School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Maite Saínz-Mejías
- School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Rhys Bruce
- School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Maryna Startseva
- School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Chaoying Ma
- School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Siobhán McClean
- School of Biomolecular and Biomedical Science and UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
38
|
Nie X, Tong Y, Li M, Ning Z, Fan H. Bacteriophage therapy for critical antibiotic-resistant Gram-positive bacteria: A systematic review of clinical researches. Microbiol Res 2025; 298:128231. [PMID: 40424685 DOI: 10.1016/j.micres.2025.128231] [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: 02/12/2025] [Revised: 04/25/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025]
Abstract
The emergence of antibiotic-resistant bacteria compromises medical interventions and poses a significant threat to global public health systems. Bacteriophage (phage) therapy offers a promising, natural, safe, and effective antimicrobial alternative, particularly advantageous for combating Gram-positive bacteria with increasing resistance. This systematic review synthesizes clinical cases published in recent 15 years, evaluating the safety and efficacy of phage therapy in treating Gram-positive bacterial infections. It details the mechanisms of action and applications of phages in treating Gram-positive bacterial infections, critically assessing phage cocktail, phage-assisted regimens, and phage-derived agents. The review further studies phage's interaction with human host, commensal microbiota, and immune system. Through the rigorous analysis, it identifies phage therapy's potential implementation obstacles, and provides valuable perspectives for future research and clinical treatment.
Collapse
Affiliation(s)
- Xinyu Nie
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100089, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100089, China.
| | - Mengzhe Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100089, China.
| | - Zhenbo Ning
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100089, China.
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100089, China; School of Life Sciences, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
39
|
Wan J, Wu M, Zhu Z, Liu X, Li J, Wu Z, Li Y, Zhang Y, Zhang Y, Wang Y, Yang F, Li M, Gu J, Luo X. Vaccination of a CdrA fragment conferred protection against Pseudomonas aeruginosa in wound infection via inhibition of biofilm formation. Vaccine 2025; 56:127185. [PMID: 40311213 DOI: 10.1016/j.vaccine.2025.127185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/26/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
Due to a prolonged course of infection aggravated by drug resistance, Pseudomonas aeruginosa (PA) remains a global health concern, raising an urgent need for an effective vaccine. However, no PA vaccines have been approved to date, possibly attributed to the role of biofilm formation in PA pathogenesis. CdrA (Cyclic-di-GMP-regulated adhesin A) is a large adhesion protein of PA, which plays a crucial role in bacterial biofilm formation. In this study, we produced the recombinant CdrA fragment (CdrA-F1) in E. coli based on the 3D structure predicted by AlphaFold. Immunization with CdrA-F1 induced a predominantly Th2-type immune response and conferred protection in a mouse PA wound infection model. Additionally, anti-CdrA-F1 antibodies effectively blocked the initial attachment of bacteria, thereby inhibiting biofilm formation both in vivo and in vitro, thereby exerting a protective effect. Our findings suggest that CdrA-F1 is a promising subunit vaccine candidate against PA infections, particularly those related to biofilm formation.
Collapse
Affiliation(s)
- Jiqing Wan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Meilin Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Zifan Zhu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Xinglong Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China; Department of Respiratory Diseases, Xinqiao Hospital, Army Medical University, Chongqing 400038, China
| | - Junyi Li
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Zhengzheng Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Yuhang Li
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Yueyue Zhang
- Chongqing Yuanlun Biopharmaceuticals Inc, Chongqing 400038, China
| | - Yi Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Ying Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Feng Yang
- Chongqing Yuanlun Biopharmaceuticals Inc, Chongqing 400038, China
| | - Mengxia Li
- Cancer Center of Daping Hospital, Army Medical University, Chongqing, 400038, China
| | - Jiang Gu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China.
| | - Xiaoli Luo
- Department of Respiratory Diseases, Xinqiao Hospital, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
40
|
Wachholz Junior D, Pontes RG, Hryniewicz BM, Kubota LT. Exploring a CRISPR/Cas12a-powered impedimetric biosensor for amplification-free detection of a pathogenic bacterial DNA. Biosens Bioelectron 2025:117607. [PMID: 40419416 DOI: 10.1016/j.bios.2025.117607] [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: 12/17/2024] [Revised: 04/30/2025] [Accepted: 05/18/2025] [Indexed: 05/28/2025]
Abstract
Timely and precise detection of bacterial infections is essential for improving patient outcomes and reducing healthcare costs, especially for sepsis, where delayed diagnosis increases mortality. Traditional culture- and PCR-based methods are time consuming and require complex sample processing, making them unsuitable for rapid diagnostics in resource-limited settings. CRISPR/Cas-based methods, particularly when combined with electrochemical sensing, offer a promising alternative for rapid point-of-care (POC) diagnostics of bacterial infections due to their simplicity and specificity. This study proposes a label-free impedimetric biosensor using the CRISPR/Cas12a system for rapid and amplification-free detection of Staphylococcus aureus DNA, a primary pathogen responsible for sepsis. By leveraging CRISPR/Cas12a's target-activated collateral cleavage on non-specific DNA reporters we investigated the impact of using a protospacer adjacent motif (PAM) sequence on detection sensitivity and specificity. Our biosensor demonstrated ultra-sensitive detection, with limit of detection as low as 20 aM for dsDNA targets in buffer and without any pre-amplification steps. The study also confirmed CRISPR specificity's dependence on the PAM sequence, showing that mismatches on targeting sequences reduces cleavage efficiency, with a drastic reduction in trans-cleavage activity for single mismatch in PAM-containing sequences. Additionally, we examined how the DNA reporter affects performance, noting reduced cleavage efficiency when a ssDNA target was paired with a dsDNA reporter. Furthermore, validation experiments using human serum samples confirmed the biosensor's accuracy for bacterial DNA detection in clinical settings. This work advances CRISPR-powered electrochemical biosensors, providing a detailed discussion on developing a highly sensitive, fast and amplification-free tool for early detection of sepsis-causing bacteria.
Collapse
Affiliation(s)
- Dagwin Wachholz Junior
- Institute of Chemistry, University of Campinas - UNICAMP, 13083-970, Campinas, Brazil; National Institute of Science and Technology in Bioanalytic (INCTBio), Campinas, Brazil.
| | - Rafael Gonçalves Pontes
- Institute of Chemistry, University of Campinas - UNICAMP, 13083-970, Campinas, Brazil; National Institute of Science and Technology in Bioanalytic (INCTBio), Campinas, Brazil
| | - Bruna M Hryniewicz
- Institute of Chemistry, University of Campinas - UNICAMP, 13083-970, Campinas, Brazil; National Institute of Science and Technology in Bioanalytic (INCTBio), Campinas, Brazil
| | - Lauro Tatsuo Kubota
- Institute of Chemistry, University of Campinas - UNICAMP, 13083-970, Campinas, Brazil; National Institute of Science and Technology in Bioanalytic (INCTBio), Campinas, Brazil
| |
Collapse
|
41
|
Chan YT, Chair SY, Gao RT, Ng MH, Lee VWY. Effect of vaccine education intervention on vaccine uptake and vaccine knowledge among older adults: A systematic review with meta-analysis. Vaccine 2025; 56:127182. [PMID: 40318347 DOI: 10.1016/j.vaccine.2025.127182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/14/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
Educational intervention is one approach for providing vaccine knowledge and encouraging vaccine uptake. However, up till now, no systematic review reporting the effectiveness of vaccine educational intervention on vaccine uptake and/or knowledge enhancement for disease preventeable by the vaccine of interest, compared to conventional/standard or no intervention, for older adults aged 60 or above was found. Six databases, including PubMed, OVID, CINAHL, Web of Science, the Cochrane Library, and the British Nursing Index, were searched. Vaccine educational interventions in this study were interventions that encourage vaccine uptake and/or provide knowledge enhancement on diseases prevented by the vaccine. Conventional/standard or no intervention refers to vaccine educational intervention that existed prior to the commencement of the intervention or no vaccine educational intervention was performed at all. All included studies were randomized control trials. The appraisal was performed with the Cochrane Risk of Bias Tool Version 1 (RoB 1). Meta-analysis was performed with RevMan Web. Ten studies were included in the meta-analysis, with a sample size of 419,523. This review demonstrated that vaccine educational intervention improves vaccine uptake (Relative Risk: 1.19, p = 0.008, 95 % confidence interval: 1.05-1.35). Furthermore, interactive vaccine educational intervention had a statistically significant effect on improving vaccine uptake among older adults (Relative Risk = 1.33, 95 % confidence interval: 1.08-1.62, p = 0.006). Neither continuous nor conducted once only intervention were found to have statistically significant improvement in vaccine uptake in the subgroup analysis. Vaccine educational interventions were found to have statistically significant effects on vaccine knowledge enhancement (Mean Difference: 0.42, 95 % confidence interval: 0.11-0.74, p = 0.008). The findings demonstrate that educational interventions significantly encourage vaccine uptake and knowledge enhancement in older adults. However, the limited amount of included literature and the high heterogeneity observed may limit the generalizability of the findings of this study. Further RCTs are recommended to bring more assertive results.
Collapse
Affiliation(s)
- Yee Tang Chan
- JC School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Sek Ying Chair
- The Nethersole School of Nursing, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Rui Tong Gao
- The Nethersole School of Nursing, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Ming Him Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Vivian Wing Yan Lee
- Centre for Learning Enhancement And Research, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| |
Collapse
|
42
|
López-Miranda JL, González-Reyna MA, Elizalde-Mata A, Mares-Briones F, Esparza R, Molina GA, Estévez M. A green approach for biomedical and photocatalytic applications using sargassum-derived nanocellulose foams decorated with MO nanoparticles (M = Zn, Cu). Int J Biol Macromol 2025; 315:144439. [PMID: 40403815 DOI: 10.1016/j.ijbiomac.2025.144439] [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: 01/24/2025] [Revised: 04/30/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
Since 2011, Sargassum algae have been invading the shores of the Caribbean, where it has been expanding at an unprecedented rate, generating large quantities of decomposing organic matter and harming the region's economy and ecosystems. As an alternative to mitigate this complex issue, the present study proposes the use and application of Sargassum natans VIII as a source for extracting nanocellulose fibers and as a stabilizing agent during the co-precipitation synthesis of ZnO and CuO quasi-spherical nanoparticles with average sizes of 29 nm and 42 nm, respectively. The environmental and biological properties were evaluated, showing a synergistic effect between nanoparticles and nanocellulose. By forming a decorated foam, the synthesis process described enables us to optimize nanoparticle dispersion, stability, and functional performance within the nanocellulose matrix and a functional composite. With this approach, we achieve improved surface activity of the NPs, resulting in enhanced performance in their anti-inflammatory, antibacterial, and photocatalytic properties. The nanocellulose/CuO inhibited the growth of both Gram-positive and Gram-negative bacterial strains, whereas the nanocellulose/ZnO exhibited antibacterial activity only against Gram-positive strains. On the other hand, nanocellulose/ZnO presented higher anti-inflammatory properties than those of CuO, mainly at high concentrations. Finally, both materials were effective in degrading methylene blue and Congo Red. Therefore, this type of nanomaterial, with its high potential in biomedical and environmental applications, represents an economical, sustainable, highly efficient, reusable, and environmentally friendly material, offering promise in these areas.
Collapse
Affiliation(s)
- J Luis López-Miranda
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Qro., 76230, Mexico
| | - M A González-Reyna
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Qro., 76230, Mexico
| | - Alberto Elizalde-Mata
- Posgrado en Ciencia e Ingeniería de Materiales, Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Blvd. Juriquilla 3000, Querétaro 76230, Mexico
| | - Fabian Mares-Briones
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Qro., 76230, Mexico
| | - Rodrigo Esparza
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Qro., 76230, Mexico
| | - Gustavo A Molina
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Qro., 76230, Mexico; Instituto de Ingeniería, Universidad Nacional Autónoma de México, Edificio 17, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, Mexico
| | - Miriam Estévez
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Qro., 76230, Mexico.
| |
Collapse
|
43
|
Zhao DW, Lohans CT. Combatting Pseudomonas aeruginosa with β-Lactam Antibiotics: A Revived Weapon? Antibiotics (Basel) 2025; 14:526. [PMID: 40426592 PMCID: PMC12108352 DOI: 10.3390/antibiotics14050526] [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: 04/25/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Pseudomonas aeruginosa is a significant threat to public health as an aggressive, opportunistic pathogen. The use of β-lactam antibiotics such as penicillins, cephalosporins, monobactams, and carbapenems remains a front-line treatment against P. aeruginosa. However, the widespread use of β-lactams has led to the emergence of β-lactam-resistant isolates that significantly increase the economic burden and risk of mortality in patients. With the declining productivity of the antibiotic discovery pipeline, research has investigated synergistic agents to revive the use of β-lactam antibiotics against β-lactam-resistant P. aeruginosa. In this review, we summarize the mechanism of β-lactam antibiotics and provide an overview of major mechanisms associated with β-lactam resistance in P. aeruginosa. We then describe the background and use of three promising classes of agents that have shown extensive beneficial effects with β-lactam antibiotics against P. aeruginosa, namely β-lactamase inhibitors, bacteriophages, and antimicrobial peptides. The current understanding of the mechanisms of these synergistic agents is discussed. Lastly, we provide an overview of the current barriers impeding antibiotic development, and offer a glimpse into recent advances of artificial intelligence-based discovery that may serve as a new foundation for antimicrobial discovery and treatment.
Collapse
Affiliation(s)
| | - Christopher T. Lohans
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada;
| |
Collapse
|
44
|
Kowalski CH, Nguyen UT, Lawhorn S, Smith TJ, Corrigan RM, Suh WS, Kalan L, Barber MF. Skin mycobiota-mediated antagonism against Staphylococcus aureus through a modified fatty acid. Curr Biol 2025; 35:2266-2281.e8. [PMID: 40233753 DOI: 10.1016/j.cub.2025.03.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 02/17/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025]
Abstract
Microbiota promote host health by inhibiting pathogen colonization, yet how host-resident fungi or mycobiota contribute to this process remains unclear. The human skin mycobiota is uniquely stable compared with other body sites and dominated by skin-adapted yeasts of the genus Malassezia. We observe that colonization of human skin by Malassezia sympodialis significantly reduces subsequent colonization by the prominent bacterial pathogen Staphylococcus aureus. In vitro, M. sympodialis generates a hydroxyl palmitic acid isomer from environmental sources that has potent bactericidal activity against S. aureus in the context of skin-relevant stressors and is sufficient to impair S. aureus skin colonization. Leveraging experimental evolution to pinpoint mechanisms of S. aureus adaptation in response to antagonism by Malassezia, we identified multiple mutations in the stringent response regulator Rel that promote survival against M. sympodialis and provide a competitive advantage on human skin when M. sympodialis is present. Similar Rel alleles have been reported in S. aureus clinical isolates, and natural Rel variants are sufficient for tolerance to M. sympodialis antagonism. Partial stringent response activation underlies tolerance to clinical antibiotics, with both laboratory-evolved and natural Rel variants conferring multidrug tolerance in a manner that is dependent on the alternative sigma factor SigB. These findings demonstrate the ability of the mycobiota to mediate pathogen colonization resistance through generation of a hydroxy palmitic acid isomer, identify new mechanisms of bacterial adaptation in response to microbiota antagonism, and reveal the potential for microbiota-driven evolution to shape pathogen antibiotic susceptibility.
Collapse
Affiliation(s)
- Caitlin H Kowalski
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA.
| | - Uyen Thy Nguyen
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Susannah Lawhorn
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - T Jarrod Smith
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Rebecca M Corrigan
- Florey Institute, School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK; The School of Medicine, University College Dublin, Belfield, Dublin 4, Dublin D04 V1W8, Ireland
| | - Won Se Suh
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Lindsay Kalan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Matthew F Barber
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA; Department of Biology, University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
45
|
Alhosani F, Islayem D, Almansoori S, Zaka A, Nayfeh L, Rezk A, Yousef AF, Pappa AM, Nayfeh A. Antibiofilm activity of ZnO-Ag nanoparticles against Pseudomonas aeruginosa. Sci Rep 2025; 15:17321. [PMID: 40389571 PMCID: PMC12089420 DOI: 10.1038/s41598-025-02372-6] [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: 02/17/2025] [Accepted: 05/13/2025] [Indexed: 05/21/2025] Open
Abstract
Biofilm-related infections remain a major concern in clinical settings due to the increasing challenge of antimicrobial resistance to conventional antimicrobial treatments. Surface coatings of nanomaterials that can effectively prevent biofilm formation and disrupt established biofilms are essential to addressing this challenge. In this study, a ZnO-Ag nanocomposite was synthesized via a dry chemical method and characterized using XRD, XPS, TEM, SEM-EDX, and AFM, confirming the presence of highly crystalline and pure ZnO and Ag nanoparticles with sharp nanoscale features. The nanocomposite demonstrated potent antibiofilm activity against Pseudomonas aeruginosa, a common Gram-negative biofilm-forming pathogen. Surface-coated glass slides prevented initial biofilm formation, while treatment with higher nanocomposite concentrations (≥ 0.25 g/L) significantly disrupted pre-formed biofilms and altered biofilm architecture, as shown by SEM and crystal violet assays. Mechanistic investigations suggested that nanoparticle surface sharpness may contribute to membrane disruption, and EPR analysis confirmed the generation of reactive oxygen species (ROS), particularly superoxide and methyl radicals, under light exposure. These results highlight the composite's strong potential for integration into surfaces prone to bacterial colonization, offering a practical approach for reducing biofilm-related complications.
Collapse
Affiliation(s)
- Fatima Alhosani
- Department of Electrical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Deema Islayem
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
- Biotechnology Center (BTC), Khalifa University, Abu Dhabi, United Arab Emirates
| | - Shamma Almansoori
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Awais Zaka
- Department of Chemical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
- Research and Innovation Center for Graphene and 2D Materials (RIC-2D), Khalifa University, Abu Dhabi, United Arab Emirates
| | - Laith Nayfeh
- Department of Electrical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ayman Rezk
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
- Research and Innovation Center for Graphene and 2D Materials (RIC-2D), Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ahmed F Yousef
- Department of Biological Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Biotechnology Center (BTC), Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Membranes and Advanced Water Technology (CMAT), Khalifa University, Abu Dhabi, United Arab Emirates
| | - Anna Maria Pappa
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ammar Nayfeh
- Department of Electrical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates.
- Research and Innovation Center for Graphene and 2D Materials (RIC-2D), Khalifa University, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
46
|
Wang S, Yao S. Distribution and drug resistance analysis of pathogens in early-stage digestive tract perforation complicated with peritonitis. Sci Rep 2025; 15:17308. [PMID: 40389620 PMCID: PMC12089619 DOI: 10.1038/s41598-025-02543-5] [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: 10/24/2024] [Accepted: 05/14/2025] [Indexed: 05/21/2025] Open
Abstract
To investigate the distribution and drug resistance of pathogens associated with early-stage digestive tract perforation with peritonitis. A retrospective analysis was conducted on patients with digestive tract perforation and peritonitis at Huadu District People's Hospital of Guangzhou from Jan. 2020 to Aug. 2024. The selected patients were divided into two groups: the upper digestive tract (UDT) group and the lower digestive tract (LDT) group. General clinical characteristics and intraoperative secretions culture results were compared and analyzed. The study included 831 patients; 41.28% were in UDT group followed 58.72% in LDT group. 694 strains that isolated comprised 503 Gram-negative bacteria (GNB), 93 g-positive bacteria (GPB) and 98 fungi. Compared to LDT group, the UDT group had a higher positive rate of GPB and fungi but a lower positive rate of GNB. The most common pathogens among GNB were E.coli, Pseudomonas aeruginosa, and Klebsiella pneumoniae; among GPB were Streptococcus anginosus, Enterococcus aviae, and Streptococcus constellations; among fungi were Candida albicans, Candida glabrata, and Candida Cruxalis. We further analyzed drug susceptibility results to related antibacterial drugs. The findings from this study have significant implications for guiding initial empirical antimicrobial therapy for patients with digestive tract perforation and peritonitis.
Collapse
Affiliation(s)
- Shuxiang Wang
- Department of Critical Care Medicine, Huadu District People's Hospital, Guangzhou, Guangdong, China.
| | - Shuwen Yao
- Department of Critical Care Medicine, Huadu District People's Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
47
|
Wang Y, Xu X, Jia S, Qu M, Pei Y, Qiu S, Zhang J, Liu Y, Ma S, Lyu N, Hu Y, Li J, Zhang E, Wan B, Zhu B, Gao GF. A global atlas and drivers of antimicrobial resistance in Salmonella during 1900-2023. Nat Commun 2025; 16:4611. [PMID: 40382325 PMCID: PMC12085583 DOI: 10.1038/s41467-025-59758-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 05/01/2025] [Indexed: 05/20/2025] Open
Abstract
Although previous studies using phenotypic or/and genomic approaches monitoring have revealed the spatiotemporal distribution of antimicrobial resistance (AMR) in Salmonella in local areas, their geographical patterns and driving factors remain largely unknown at a global scale. Here, we performed an analysis of publicly available data of 208,233 Salmonella genomes in 148 countries/regions between 1900 and 2023 and explored driving indicators of AMR. Overall, we found that the geographic distribution of AMR varied depending on the location, source, and serovar. The proportion of AMR levels increased across six continents, especially in serovars Agona, Dublin, I 1,4,[5],12:i:-, Muenchen, Senftenberg, Mbandaka mainly from chickens, food, wild animals, and the environment, while decreased in Schwarzengrund and Saintpaul mainly from cattle, pigs, and turkeys. We also found that S. Typhimurium exhibiting macro, red, dry, and rough was detected as early as 1992 in the USA, earlier than in China. Moreover, we identified that antibiotic consumption, agriculture, climate, urban, health, and socioeconomic factors contribute to the development of AMR in Salmonella. We present a globally high-resolution genetic atlas of Salmonella and also identify some factors driving the rise of AMR, which can provide valuable information for understanding the transmission dynamics and evolutionary trajectories of Salmonella.
Collapse
Affiliation(s)
- Yanan Wang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.
- Longhu Laboratory, Zhengzhou, Henan, China.
- Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
| | - Xuebin Xu
- Department of Microbiology, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Shulei Jia
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Mengqi Qu
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yuhang Pei
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Shaofu Qiu
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Jianmin Zhang
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yue Liu
- Department of Microbiology, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Sufang Ma
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Na Lyu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yongfei Hu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jianli Li
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Erqin Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Bo Wan
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Baoli Zhu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Microbial Drug Resistance and Resistome, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - George F Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Key Laboratory of Microbial Drug Resistance and Resistome, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
48
|
Liu D, Ton PM, Zong D, Zarrinpar A, Ding Y. Expression of Fluorescence Reporters and Natural Products in Native Gut Escherichia coli. ACS Synth Biol 2025; 14:1557-1566. [PMID: 40138712 DOI: 10.1021/acssynbio.4c00835] [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: 03/29/2025]
Abstract
Escherichia coli is a widely studied model organism and an integral component of the human gut microbiome, offering significant potential for bacteria-based therapeutic applications. Despite this promise, engineering native E. coli strains remains challenging. In this study, we employed the chassis-independent recombinase-assisted genome engineering (CRAGE) technique to genetically engineer the native gut strain E. coli EcAZ-1 and the probiotic strain E. coli Nissle 1917 (EcN). We successfully expressed a suite of heterologous genes, including the bioluminescent lux operon, green fluorescent protein (GFP), and oxygen-independent fluorescent protein IFP2.0, in both strains. Optimization of IFP2.0 fluorescence was achieved under both aerobic and anaerobic conditions by coexpressing a heme oxygenase gene and/or supplementing the chromophore biliverdin or hemin. Additionally, we engineered these strains to biosynthesize the bioactive compounds naringenin and mycosporine-like amino acids. This work highlights the potential of native E. coli strains as versatile platforms for synthetic biology, paving the way for innovative applications in biomedical research and therapeutic development.
Collapse
Affiliation(s)
- Dake Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, Florida 32610, United States
| | - Phuong M Ton
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, Florida 32610, United States
| | - David Zong
- Division of Gastroenterology, University of California, San Diego, La Jolla, California 92093, United States
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, La Jolla, California 92093, United States
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California 92093, United States
- Division of Gastroenterology, Jennifer Moreno Department of Veterans Affairs Medical Center, La Jolla, California 92093, United States
| | - Yousong Ding
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
49
|
Lin S, Xiong Z, Zhang C, Liu S, Ding T, Yang K, He Y, Zhao Z, Zhou Z. A molecular comparative study of intestinal colonization with Staphylococcus aureus between pediatric inpatients and outpatients of different age groups. Microbiol Spectr 2025:e0239424. [PMID: 40377306 DOI: 10.1128/spectrum.02394-24] [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: 09/24/2024] [Accepted: 04/02/2025] [Indexed: 05/18/2025] Open
Abstract
Currently, Staphylococcus aureus (S. aureus) is one of the leading causes of death from infectious diseases worldwide. Our aim in this study was to investigate and compare the molecular epidemiology and antibiotic resistance of S. aureus colonizing the intestinal tract of pediatric inpatients and outpatients of different age groups. We analyzed stool samples from 1,300 patients at a children's hospital in Shenzhen, China. After culturing S. aureus, we used matrix-assisted laser desorption/ionization time-of-flight mass spectrometry to identify the strains. In addition, we performed antimicrobial susceptibility testing on isolated S. aureus strains as well as pheno- and genotypic characterization by PCR. S. aureus was detected in 104 out of 1,300 (8.0%) patients, including 20 out of 1,300 (1.5%) patients with methicillin-resistant S. aureus (MRSA). MRSA accounted for 19.2% of the S. aureus isolates. The resistance rates of S. aureus strains to penicillin, erythromycin, clindamycin, levofloxacin, and moxifloxacin were 83.7%, 34.6%, 31.7%, 3.9%, and 3.9%, respectively. None of the strains showed resistance to linezolid, daptomycin, tigecycline, vancomycin, or tetracycline. One hundred fou strains of S. aureus revealed that 49.0% (51/104) harbored enterotoxin genes, and most enterotoxin-positive strains carried only one gene type (90.2%, 46/51), while a minority carried two gene types (9.8%, 5/51). Besides, a total of 29 sequence types (STs) were identified with the three most prevalent STs: ST45, ST188, and ST6, accounting for 12.5%, 12.5%, and 9.6%. We found that the molecular characteristics of S. aureus in intestinal colonization of children have regional differences. To provide a theoretical basis for the prevention and control of S. aureus infections, increased surveillance of local S. aureus resistance and molecular epidemiological characteristics is needed.IMPORTANCEThis study assessed the clinical and molecular epidemiology of Staphylococcus aureus in pediatric patients at a children's hospital in Shenzhen, South China by means of screening stool samples for pheno- and genotypic characterization for carriage of S. aureus. Of 1,300 fecal samples screened, 104 (8.0%) were positive for S. aureus with 19.2% methicillin-resistant S. aureus. The resistance rates of S. aureus to penicillin, erythromycin, clindamycin, levofloxacin, and moxifloxacin were 83.7%, 34.6%, 31.7%, 3.9%, and 3.9%, respectively. None of the strains showed resistance to linezolid, daptomycin, tigecycline, vancomycin, or tetracycline. One hundred four strains of S. aureus revealed that 49.0% (51/104) harbored enterotoxin genes, and most enterotoxin-positive strains carried only one gene type (90.2%, 46/51), while a minority carried two gene types (9.8%, 5/51). Besides, a total of 29 sequence types (STs) were identified with the three most prevalent STs: ST45, ST188, and ST6, accounting for 12.5%, 12.5%, and 9.6%.
Collapse
Affiliation(s)
- Shaoxiang Lin
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Medical Research Institute of Maternal and Child, Shenzhen, China
| | - Zhile Xiong
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Medical Research Institute of Maternal and Child, Shenzhen, China
- Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Chao Zhang
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Medical Research Institute of Maternal and Child, Shenzhen, China
| | - Shuyan Liu
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Medical Research Institute of Maternal and Child, Shenzhen, China
| | - Tongyan Ding
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Medical Research Institute of Maternal and Child, Shenzhen, China
| | - Kaiyue Yang
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Medical Research Institute of Maternal and Child, Shenzhen, China
| | - Yunxing He
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Medical Research Institute of Maternal and Child, Shenzhen, China
| | - Zhimin Zhao
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Medical Research Institute of Maternal and Child, Shenzhen, China
| | - Zhenwen Zhou
- Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Medical Research Institute of Maternal and Child, Shenzhen, China
| |
Collapse
|
50
|
Brinkmann A, Chiriac U, Eckmann C, Frey OR, Grabein B, Hagel S, Richter D, Liebchen U. [Empirical antibiotic therapy in life-threatening infections-current concepts and controversies]. DIE ANAESTHESIOLOGIE 2025:10.1007/s00101-025-01531-8. [PMID: 40372450 DOI: 10.1007/s00101-025-01531-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/01/2025] [Indexed: 05/16/2025]
Abstract
Along with early administration and the selection of the right drug, it is important to consider pharmacokinetic and pharmacodynamic principles, especially in life-threatening infections. Therefore, successful antibiotic therapy requires a clear understanding of the relationship between microbiology, pharmacology, and intensive care medicine. Open and transparent communication among professionals is essential for improving the quality of care. This article discusses current concepts and controversies by presenting a relevant case study of community-acquired pneumonia. It aims to help clinicians select the right drug for each patient, including the correct dose, the appropriate administration, and the right duration of treatment.
Collapse
Affiliation(s)
- A Brinkmann
- Klinik für Anästhesie, operative Intensivmedizin und spezielle Schmerztherapie, Klinikum Heidenheim, Schlosshaustraße 100, 89522, Heidenheim, Deutschland.
| | - U Chiriac
- Apotheke, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - C Eckmann
- Klinik für Allgemein‑, Viszeral- und Thoraxchirurgie und ABS-Team, Klinikum Hannoversch-Münden, Universitätsmedizin Göttingen, Hannoversch-Münden, Deutschland
| | - O R Frey
- Apotheke, Kliniken Landkreis Heidenheim, Heidenheim, Deutschland
| | - B Grabein
- Stabsstelle Klinische Mikrobiologie und Krankenhaushygiene, LMU Klinikum, LMU München, München, Deutschland
| | - S Hagel
- Institut für Infektionsmedizin und Krankenhaushygiene, Universitätsklinikum Jena, Jena, Deutschland
| | - D Richter
- Klinik für Anästhesiologie, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - U Liebchen
- Klinik für Anaesthesiologie, LMU Klinikum, LMU München, München, Deutschland
| |
Collapse
|