1
|
Lin Y, Wang J, Bu F, Zhang R, Wang J, Wang Y, Huang M, Huang Y, Zheng L, Wang Q, Hu X. Bacterial extracellular vesicles in the initiation, progression and treatment of atherosclerosis. Gut Microbes 2025; 17:2452229. [PMID: 39840620 DOI: 10.1080/19490976.2025.2452229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. However, current anti-atherosclerosis drugs have shown conflicting therapeutic outcomes, thereby spurring the search for novel and effective treatments. Recent research indicates the crucial involvement of oral and gastrointestinal microbiota in atherosclerosis. While gut microbiota metabolites, such as choline derivatives, have been extensively studied and reviewed, emerging evidence suggests that bacterial extracellular vesicles (BEVs), which are membrane-derived lipid bilayers secreted by bacteria, also play a significant role in this process. However, the role of BEVs in host-microbiota interactions remains insufficiently explored. This review aims to elucidate the complex communication mediated by BEVs along the gut-heart axis. In this review, we summarize current knowledge on BEVs, with a specific focus on how pathogen-derived BEVs contribute to the promotion of atherosclerosis, as well as how BEVs from gut symbionts and probiotics may mitigate its progression. We also explore the potential and challenges associated with engineered BEVs in the prevention and treatment of atherosclerosis. Finally, we discuss the benefits and challenges of using BEVs in atherosclerosis diagnosis and treatment, and propose future research directions to address these issues.
Collapse
Affiliation(s)
- Yuling Lin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Bu
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China
| | - Ruyi Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junhui Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yubing Wang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiumei Hu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Li L, Xu X, Cheng P, Yu Z, Li M, Yu Z, Cheng W, Zhang W, Sun H, Song X. Klebsiella pneumoniae derived outer membrane vesicles mediated bacterial virulence, antibiotic resistance, host immune responses and clinical applications. Virulence 2025; 16:2449722. [PMID: 39792030 PMCID: PMC11730361 DOI: 10.1080/21505594.2025.2449722] [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: 07/01/2024] [Revised: 11/14/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025] Open
Abstract
Klebsiella pneumoniae is a gram-negative pathogen that can cause multiple diseases including sepsis, urinary tract infections, and pneumonia. The escalating detections of hypervirulent and antibiotic-resistant isolates are giving rise to growing public concerns. Outer membrane vesicles (OMVs) are spherical vesicles containing bioactive substances including lipopolysaccharides, peptidoglycans, periplasmic and cytoplasmic proteins, and nucleic acids. Emerging studies have reported various roles of OMVs in bacterial virulence, antibiotic resistance, stress adaptation, and host interactions, whereas knowledge on their roles in K. pneumoniae is currently unclear. In this review, we summarized recent progress on the biogenesis, components, and biological function of K. pneumoniae OMVs, the impact and action mechanism in virulence, antibiotic resistance, and host immune response. We also deliberated on the potential of K. pneumoniae OMVs in vaccine development, as diagnostic biomarkers, and as drug nanocarriers. In conclusion, K. pneumoniae OMVs hold great promise in the prevention and control of infectious diseases, which merits further investigation.
Collapse
Affiliation(s)
- Lifeng Li
- Henan International Joint Laboratory of Children’s Infectious Diseases, Department of Neonatology, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Xinxiu Xu
- Department of Neurology, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Ping Cheng
- Henan International Joint Laboratory of Children’s Infectious Diseases, Department of Neonatology, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Zengyuan Yu
- Henan International Joint Laboratory of Children’s Infectious Diseases, Department of Neonatology, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Mingchao Li
- Henan International Joint Laboratory of Children’s Infectious Diseases, Department of Neonatology, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Zhidan Yu
- Henan International Joint Laboratory of Children’s Infectious Diseases, Department of Neonatology, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Weyland Cheng
- Henan International Joint Laboratory of Children’s Infectious Diseases, Department of Neonatology, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Wancun Zhang
- Henan International Joint Laboratory of Children’s Infectious Diseases, Department of Neonatology, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Huiqing Sun
- Henan International Joint Laboratory of Children’s Infectious Diseases, Department of Neonatology, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Xiaorui Song
- Henan International Joint Laboratory of Children’s Infectious Diseases, Department of Neonatology, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| |
Collapse
|
3
|
Zhou D, Yang X, Gao Y, Zheng R. The mechanism of outer membrane vesicle-mediated resistance to carbapenem antibiotics. Microb Pathog 2025; 205:107654. [PMID: 40316062 DOI: 10.1016/j.micpath.2025.107654] [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: 09/17/2024] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
The escalating prevalence of carbapenem resistance in Gram-negative bacteria presents a critical therapeutic challenge, demanding urgent elucidation of novel resistance mechanisms. This review systematically examines the emerging role of outer membrane vesicles (OMVs) as multifunctional mediators of carbapenem resistance, synthesizing recent advances in understanding their biological properties and mechanistic contributions. Through comprehensive analysis of β-lactamase dissemination pathways, we demonstrate that OMVs are extracellular vectors facilitating antibiotic degradation through enzymatic cargo delivery while concurrently acting as genetic transmission vehicles for resistance determinants. Crucially, OMVs exhibit functional versatility in enhancing bacterial survival via dual mechanisms: structurally, by promoting biofilm matrix formation that establishes antibiotic-protected niches, and immunologically, through modulation of host-pathogen interactions that impair microbial clearance. The review further identifies OMV-mediated antibiotic sequestration and competitive binding as underappreciated resistance amplifiers. These insights refine our understanding of resistance evolution and reveal OMV biogenesis pathways as promising therapeutic targets. This synthesis establishes OMVs as central players in carbapenem resistance architecture, providing a strategic framework for developing countermeasures against multidrug-resistant infections.
Collapse
Affiliation(s)
- Dan Zhou
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China; Department of Clinical Laboratory, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of College of Medical, Kunming University of Science and Technology, China
| | - Xiaoyu Yang
- Department of Clinical Laboratory, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; Regenerative Medicine Research Center, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of College of Medical, Kunming University of Science and Technology, China
| | - Yuhong Gao
- Department of Clinical Laboratory, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of College of Medical, Kunming University of Science and Technology, China
| | - Rui Zheng
- Department of Clinical Laboratory, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China; The Affiliated Hospital of College of Medical, Kunming University of Science and Technology, China.
| |
Collapse
|
4
|
Huang X, Hu Z, Shang W, Chen J, Hu Q, Zhou Y, Ding R, Yin J, Li M, Liu H, Dou J, Peng H, Rao Y, Liu L, Wang Y, Tan L, Yang Y, Wu J, Xiao C, Yang Y, Rao X. Beta-Lactam Antibiotics Promote Extracellular Vesicle Production of Staphylococcus aureus Through ROS-Mediated Lipid Metabolic Reprogramming. J Extracell Vesicles 2025; 14:e70077. [PMID: 40314062 PMCID: PMC12046293 DOI: 10.1002/jev2.70077] [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: 11/21/2024] [Accepted: 03/26/2025] [Indexed: 05/03/2025] Open
Abstract
Bacterial extracellular vesicles (EVs) are natural reservoirs of biological active substances. They exhibit promising application in developing bioproducts such as vaccine, drug-delivery system and anticancer agent. However, the low yield of naturally secreted EVs during bacterial growth is a bottleneck factor that restricts EV applications. In this study, we showed that sub-minimum inhibitory concentration (MIC) of β-lactams boosted EV production in various Staphylococcus aureus strains. The expression of penicillin-binding protein (PBP) genes increased after β-lactam treatment, and the inactivation of alternative PBPs promoted EV secretion of S. aureus. We also demonstrated that sub-MIC β-lactams promoted EV production via a reactive oxygen species (ROS)-dependent pathway. Deletion of redundant pbp genes enhanced oxacillin (OXA)-stimulated ROS levels. Transcriptomic and lipidomic analyses revealed that OXA-induced ROS triggered lipid metabolic reprogramming in S. aureus. Particularly, ROS promoted lipid peroxidation (LPO) and increased the biosynthesis of phosphatidic acid (PA) and lipoteichoic acid (LTA) that contributed to EV generation. Furthermore, OXA treatment altered the diversity of EV-loaded proteins. OXA-treated ∆ agr /OXAEVs induced stronger Dengue EDIII-specific antibodies in BALB/c mice than did ∆ agrEVs. Overall, this study provided mechanic insights into β-lactam-promoted EV production in S. aureus, and highlighted the potential strategies to prepare EVs for various applications.
Collapse
Affiliation(s)
- Xiaonan Huang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Zhen Hu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Weilong Shang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Juan Chen
- Department of PharmacyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Qiwen Hu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yumin Zhou
- Department of DermatologySouthwest HospitalArmy Medical UniversityChongqingChina
| | - Ruolan Ding
- Department of MicrobiologySchool of MedicineChongqing UniversityChongqingChina
| | - Jing Yin
- Department of NeurologyFirst Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Mengyang Li
- Department of MicrobiologySchool of MedicineChongqing UniversityChongqingChina
| | - He Liu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Jianxiong Dou
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Huagang Peng
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yifan Rao
- Department of Emergency MedicineXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Lu Liu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yuting Wang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Li Tan
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yuhua Yang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Jianghong Wu
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
- Department of Emergency MedicineXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Chuan Xiao
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yi Yang
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Xiancai Rao
- Department of MicrobiologyCollege of Basic Medical SciencesKey Laboratory of Microbial Engineering under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
- Department of MicrobiologySchool of MedicineChongqing UniversityChongqingChina
| |
Collapse
|
5
|
Wang WB, Wan JY, Yu DJ, Du HX, Zhou HF, Wan HT, Yang JH. Chlorogenic acid inhibits virulence and resistance gene transfer in outer membrane vesicles of carbapenem-resistant Klebsiella pneumoniae. Front Pharmacol 2025; 16:1562096. [PMID: 40230687 PMCID: PMC11994928 DOI: 10.3389/fphar.2025.1562096] [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: 01/16/2025] [Accepted: 03/07/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction Carbapenem-resistant Klebsiella pneumoniae (CRKp) infection poses a significant global public health challenge, with the misuse of antibiotics further contributing to the development of resistance and triggering harmful inflammatory responses. Outer membrane vesicles (OMVs) released by CRKp under sub-lethal concentration of MEM pressure (KOMV-MEM) exhibit enhanced virulence and greater efficiency in transferring resistance genes. Methods We investigated the inhibitory effects of chlorogenic acid (CA) on KOMV-MEM characteristics and its protective role in KOMV-MEM infected mice. Based on LC-MS proteomic analysis of vesicles, we screened for potential targets of KOMV-MEM in promoting macrophage (MØ) pyroptosis pathways and inducing resistance gene transfer. Subsequently, computational predictions and experimental validation were performed to determine how CA regulates these mechanisms. Results This study confirmed that, under MEM pressure, the exacerbated infection levels in CRKp-inoculated mice are attributable to the high virulence of KOMV-MEM. Computational and experimental results demonstrated that CA inhibits pyroptosis by reducing MØ capture of KOMV-MEM through blocking the interaction between GroEL and LOX-1. Furthermore, CA prevents the spread of resistance genes by disrupting the conjugation and transfer processes between KOMV-MEM and recipient bacteria. Finally, in vitro and in vivo assays showed that CA inhibits KOMV-MEM resistance enzymes, thereby preventing the hydrolysis of MEM in the environment and depriving susceptible bacteria of protection. Discussion These findings provide the first confirmation that CA can inhibit both the virulence and the transmission of drug resistance in KOMV-MEM. This underscores the potential of CA treatment as a promising antimicrobial strategy against CRKp infection.
Collapse
Affiliation(s)
- Wen-Ba Wang
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jia-Yang Wan
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dao-Jun Yu
- Department of Medical Laboratory, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Xia Du
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui-Fen Zhou
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hai-Tong Wan
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie-Hong Yang
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
6
|
Chatrizeh M, Tian J, Rogers M, Feturi F, Wu G, Firek B, Nikonov R, Cass L, Sheppeck A, Ramos-Jiménez RG, Ohja L, Caroll A, Henkel M, Azar J, Aneja RK, Campfield B, Simon D, Morowitz MJ. Plant based enteral nutrition outperforms artificial nutrition in mitigating consequences of antibiotic-induced dysbiosis in mice and humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.19.25323813. [PMID: 40166543 PMCID: PMC11957089 DOI: 10.1101/2025.03.19.25323813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Malnutrition, gut inflammation, and antibiotic induced dysbiosis (AID) are omnipresent risk factors for poor clinical outcomes among critically ill patients. We previously showed that commercially available plant-based enteral nutrition (PBEN) preserves a commensal microbiome when compared to commonly used forms of commercially available artificial enteral nutrition (AEN). This study reveals that PBEN is superior to artificial enteral nutrition (AEN) in recovering from antibiotic-induced dysbiosis (AID) in mice and humans. PBEN effectively mitigates anemia, leukopenia, restores naïve lymphocyte populations, and reduces bone marrow myeloid cell expansion. Animals randomized to PBEN also fared better in response to infectious challenges after antibiotics. A pilot clinical study validated these findings, showing increased gut commensals, reduced pathogens, and improved leukocyte balance in critically ill patients receiving PBEN compared to AEN. These results suggest PBEN offers a practical dietary approach to mitigate antibiotic-associated complications and improve clinical outcomes among hospitalized patients requiring supplemental nutrition.
Collapse
Affiliation(s)
- Mona Chatrizeh
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jianmin Tian
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew Rogers
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Firuz Feturi
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guojun Wu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Brian Firek
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Roman Nikonov
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Cass
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexandra Sheppeck
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Lavnish Ohja
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ali Caroll
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mathew Henkel
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Justin Azar
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rajesh K Aneja
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian Campfield
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dennis Simon
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pediatric Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael J Morowitz
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Lead contact
| |
Collapse
|
7
|
Liu X, Xu Q, Yang X, Heng H, Yang C, Yang G, Peng M, Chan EWC, Chen S. Capsular polysaccharide enables Klebsiella pneumoniae to evade phagocytosis by blocking host-bacteria interactions. mBio 2025; 16:e0383824. [PMID: 39950808 PMCID: PMC11898582 DOI: 10.1128/mbio.03838-24] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025] Open
Abstract
Capsule polysaccharide (CPS) is among the most important virulence factors of Klebsiella pneumoniae. Previous studies demonstrated that CPS plays multiple functional roles, but the mechanism by which this virulence factor enhances the survival fitness of K. pneumoniae remains unclear. In this work, we demonstrate that CPS is the main cellular component that not only elicits the host immune response to K. pneumoniae but also enables this pathogen to survive for a prolonged period under adverse environmental conditions. Consistently, our in vitro experiments suggest that CPS prevents K. pneumoniae from phagocytosis, rendering the encapsulated strain more difficult to be eradicated by the host. We also found that phagocytosis of K. pneumoniae is partially mediated by LOX-1, a scavenger receptor of the host, and that CPS may impede interaction between LOX-1 and this pathogenic bacteria, therefore reducing the phagocytosis process. These findings provide insights into the pathogenic mechanisms of this important clinical pathogen and should facilitate the design of new strategies to combat K. pneumoniae infections. IMPORTANCE Klebsiella pneumoniae has become one of the most important clinical bacterial pathogens due to its evolution into hyperresistant and hypervirulent phenotypes. The mechanism of virulence of this pathogen is not well understood, particularly because it differs from other Enterobacteriaceae pathogens such as Escherichia coli and Salmonella. The capsule polysaccharide (CPS) of this pathogen is well recognized for contributing to the virulence of K. pneumoniae, but the exact mechanisms underlying its contribution are unclear. In this study, we demonstrated that CPS does not directly contribute to the host response; rather, it forms an external coat that blocks host recognition and prevents immune cells from binding to receptor proteins on K. pneumoniae, thus inhibiting phagocytosis, which makes it more challenging for the body to fight off infections. Understanding these mechanisms is vital for developing new treatments against K. pneumoniae infections, ultimately improving patient outcomes and public health.
Collapse
Affiliation(s)
- Xiaoxuan Liu
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- The State Key Laboratory of Chemical Biology and Drug Discovery, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Qi Xu
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- The State Key Laboratory of Chemical Biology and Drug Discovery, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Xuemei Yang
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- The State Key Laboratory of Chemical Biology and Drug Discovery, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Heng Heng
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- The State Key Laboratory of Chemical Biology and Drug Discovery, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Chen Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- The State Key Laboratory of Chemical Biology and Drug Discovery, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Guan Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Mingxiu Peng
- Shenzhen Key Laboratory for Food Biological Safety Control, The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Edward Wai-Chi Chan
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- The State Key Laboratory of Chemical Biology and Drug Discovery, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- Shenzhen Key Laboratory for Food Biological Safety Control, The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Sheng Chen
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- The State Key Laboratory of Chemical Biology and Drug Discovery, Faculty of Science, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
- Shenzhen Key Laboratory for Food Biological Safety Control, The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
8
|
Xie Y, Shi Y, Wang L, Li C, Wu M, Xu J. Outer membrane vesicle contributes to the Pseudomonas aeruginosa resistance to antimicrobial peptides in the acidic airway of bronchiectasis patients. MedComm (Beijing) 2025; 6:e70084. [PMID: 39896756 PMCID: PMC11782972 DOI: 10.1002/mco2.70084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/29/2024] [Accepted: 11/23/2024] [Indexed: 02/04/2025] Open
Abstract
Pseudomonas aeruginosa is the predominant pathogen causing chronic infection in the airway of patients with bronchiectasis (BE), a chronic respiratory disease with high prevalence worldwide. Environmental factors are vital for bacterial successful colonization. Here, with sputa and bronchoalveolar lavage fluids, we determined that the concentration of airway antimicrobial peptide LL-37 and lactate was elevated in BE patients, especially in those infected with P. aeruginosa. The in vitro antibacterial assay revealed the bactericidal activity of LL-37 against the clinical P. aeruginosa isolates, which were dampened in the acidic condition. P. aeruginosa production of outer membrane vesicles (OMVs) enhanced in the lactate-adjusted acidic condition. Transcriptomic analysis suggested that OMVs induce the hyperproduction of the chemical compound 2-heptyl-4-quinolone (HHQ) in the bacterial population, which was verified by high-performance liquid chromatography. The positively charged HHQ interfered with the binding of LL-37 to bacterial cell membrane, potentiating the P. aeruginosa resistance to LL-37. To our knowledge, this is a new resistance mechanism of P. aeruginosa against antimicrobial peptides and may provide theoretical support for the development of new antibacterial therapies.
Collapse
Affiliation(s)
- Yingzhou Xie
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of MedicineTongji UniversityShanghaiChina
| | - Yi‐Han Shi
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of MedicineTongji UniversityShanghaiChina
| | - Le‐Le Wang
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of MedicineTongji UniversityShanghaiChina
| | - Cheng‐Wei Li
- Department of Pulmonary and Critical Care MedicineHuashan Hospital, Fudan UniversityShanghaiChina
| | - Min Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, and Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| | - Jin‐Fu Xu
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of MedicineTongji UniversityShanghaiChina
- Department of Respiratory and Critical Care MedicineHuadong Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
9
|
Li P, Lu M, Peng T, Wu Y, Zhu L, Liu Y, Zhang W, Xiang T. An improvised one-step OptiPrep cushion ultracentrifugation method for outer membrane vesicles isolation of Klebsiella pneumoniae. BMC Microbiol 2024; 24:548. [PMID: 39732632 DOI: 10.1186/s12866-024-03649-y] [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: 04/05/2024] [Accepted: 11/13/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) play a crucial role in intraspecies and interspecies communication, significantly influencing physiological and pathological processes. Outer membrane vesicles (OMVs) secreted by Gram-negative bacteria are rich in components from the parent cells and are important for bacterial communication, immune evasion, and pathogenic mechanisms. However, the extraction and purification of OMVs face numerous challenges due to their small size and heterogeneity. RESULTS This study proposes an innovative strategy that combines traditional differential centrifugation (DC) with one-step ultracentrifugation (ODG) to develop a dual differential gradient centrifugation (DDGC) method for extracting outer membrane vesicles from Klebsiella pneumoniae. By comparing the DC and DDGC extraction methods, we found that OMVs extracted by DDGC exhibited more typical morphology, clearer backgrounds, and more uniform particle size distribution. The lipid polysaccharide (LPS) content in OMVs extracted by DDGC was significantly higher than that obtained by DC, and the outer membrane protein content was also greater, demonstrating enhanced biological activity. Biological activity assays indicated that OMVs extracted by DDGC showed stronger cytotoxicity to A549 lung epithelial cells, a significant decrease in cell viability, and higher levels of inflammatory factor expression(IL-6, TNF-α, IL-1β, and IL-8). CONCLUSION Our study demonstrates the advantages of the DDGC method in extracting K. pneumoniae OMVs, showing improvements in morphology, particle size distribution, protein content, and biological activity. This provides a solid foundation for further exploration of the biological functions of OMVs and their potential applications in the biomedical field.
Collapse
Affiliation(s)
- Ping Li
- Department of Respiratory and Critical Care Medicine, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiang Xi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, 330052, P.R. China
| | - Ming Lu
- Department of Respiratory and Critical Care Medicine, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiang Xi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, 330052, P.R. China
| | - Tingxiu Peng
- Department of Respiratory and Critical Care Medicine, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yifan Wu
- Department of Respiratory and Critical Care Medicine, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lanlan Zhu
- Department of Respiratory and Critical Care Medicine, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiang Xi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, 330052, P.R. China
| | - Yang Liu
- Jiang Xi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, 330052, P.R. China
- Department of Clinical Microbiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong wai zheng jie No. 17, Nanchang, 330006, PR China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
- Jiang Xi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, 330052, P.R. China.
| | - Tianxin Xiang
- Jiang Xi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, 330052, P.R. China.
- Jiangxi Medical Center for Critical Public Health Events, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330052, P.R. China.
| |
Collapse
|
10
|
Haracic E, Waters JK, Nguyen Thi Nguyen T, Kostoulias X, Davies BJ, Yu L, Peleg AY, Bulone V, Short FL, Eijkelkamp BA. Fatty Acid Uptake in Klebsiella pneumoniae and the Landscape of Its Infectious Niches. ACS Infect Dis 2024; 10:4048-4056. [PMID: 39259670 DOI: 10.1021/acsinfecdis.4c00307] [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: 09/13/2024]
Abstract
Klebsiella pneumoniae is consistently ranked among the most problematic multidrug-resistant bacterial pathogens in healthcare systems. Developing novel treatments requires a better understanding of its interaction with the host environment. Although bacteria can synthesize fatty acids, emerging findings suggest a potential preference for their acquisition from the host. Fatty acid profiling of mice revealed a dramatic increase in the level of hepatic lipids during K. pneumoniae infection. The K. pneumoniae fatty acid composition and uptake capabilities were found to be largely clonally conserved. Correlations between fatty acid uptake, outer membrane vesicle production, and cell permeability were observed, but this did not translate to alterations in cell morphology, capsule production, or antimicrobial susceptibility. Importantly, hyper-capsulation did not prevent the uptake of hydrophobic fatty acids. The uptake of a saturated fatty acid by hypervirulent K. pneumoniae isolate may provide insights into the clinical association of K. pneumoniae infections with hyperlipidemic and/or obese individuals.
Collapse
Affiliation(s)
- Ella Haracic
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - Jack K Waters
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - To Nguyen Thi Nguyen
- Infection Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC 3168, Australia
| | - Xenia Kostoulias
- Infection Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC 3168, Australia
- Department of Infectious Diseases, Alfred Health and School of Translational Medicine, Monash University, Prahan, VIC 3181, Australia
| | - Brynley J Davies
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - Long Yu
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Anton Y Peleg
- Infection Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC 3168, Australia
- Department of Infectious Diseases, Alfred Health and School of Translational Medicine, Monash University, Prahan, VIC 3181, Australia
| | - Vincent Bulone
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Francesca L Short
- Infection Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC 3168, Australia
| | - Bart A Eijkelkamp
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
11
|
Caselli L, Du G, Micciulla S, Traini T, Sebastiani F, Diedrichsen RG, Köhler S, Skoda MWA, van der Plas MJA, Malmsten M. Photocatalytic Degradation of Bacterial Lipopolysaccharides by Peptide-Coated TiO 2 Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2024; 16:60056-60069. [PMID: 39443826 PMCID: PMC11551910 DOI: 10.1021/acsami.4c15706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
In this study, we report the degradation of smooth and rough lipopolysaccharides (LPS) from Gram-negative bacteria and of lipoteichoic acid (LTA) from Gram-positive bacteria by peptide-coated TiO2 nanoparticles (TiO2 NPs). While bare TiO2 NPs displayed minor binding to both LPS and LTA, coating TiO2 NPs with the antimicrobial peptide LL-37 dramatically increased the level of binding to both LPS and LTA, decorating these uniformly. Importantly, peptide coating did not suppress reactive oxygen species generation of TiO2 NPs; hence, UV illumination triggered pronounced degradation of LPS and LTA by peptide-coated TiO2 NPs. Structural consequences of oxidative degradation were examined by neutron reflectometry for smooth LPS, showing that degradation occurred preferentially in its outer O-antigen tails. Furthermore, cryo-TEM and light scattering showed lipopolysaccharide fragments resulting from degradation to be captured by the NP/lipopolysaccharide coaggregates. The capacity of LL-37-TiO2 NPs to capture and degrade LPS and LTA was demonstrated to be of importance for their ability to suppress lipopolysaccharide-induced activation in human monocytes at simultaneously low toxicity. Together, these results suggest that peptide-coated photocatalytic NPs offer opportunities for the confinement of infection and inflammation.
Collapse
Affiliation(s)
- Lucrezia Caselli
- Department
of Physical Chemistry 1, Lund University, Lund SE-22100, Sweden
| | - Guanqun Du
- Department
of Physical Chemistry 1, Lund University, Lund SE-22100, Sweden
| | | | - Tanja Traini
- Department
of Pharmacy, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Federica Sebastiani
- Department
of Physical Chemistry 1, Lund University, Lund SE-22100, Sweden
- Department
of Pharmacy, University of Copenhagen, Copenhagen DK-2100, Denmark
| | | | - Sebastian Köhler
- LINXS
Institute of Advanced Neutron and X-ray Science, Scheelevagen 19, Lund 22370, Sweden
| | - Maximilian W. A. Skoda
- ISIS
Pulsed Neutron and Muon Source, Rutherford
Appleton Laboratory, Harwell OX11 0QX, U.K.
| | | | - Martin Malmsten
- Department
of Physical Chemistry 1, Lund University, Lund SE-22100, Sweden
- Department
of Pharmacy, University of Copenhagen, Copenhagen DK-2100, Denmark
| |
Collapse
|
12
|
Lei TY, Liao BB, Yang LR, Wang Y, Chen XB. Hypervirulent and carbapenem-resistant Klebsiella pneumoniae: A global public health threat. Microbiol Res 2024; 288:127839. [PMID: 39141971 DOI: 10.1016/j.micres.2024.127839] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 08/16/2024]
Abstract
The evolution of hypervirulent and carbapenem-resistant Klebsiella pneumoniae can be categorized into three main patterns: the evolution of KL1/KL2-hvKp strains into CR-hvKp, the evolution of carbapenem-resistant K. pneumoniae (CRKp) strains into hv-CRKp, and the acquisition of hybrid plasmids carrying carbapenem resistance and virulence genes by classical K. pneumoniae (cKp). These strains are characterized by multi-drug resistance, high virulence, and high infectivity. Currently, there are no effective methods for treating and surveillance this pathogen. In addition, the continuous horizontal transfer and clonal spread of these bacteria under the pressure of hospital antibiotics have led to the emergence of more drug-resistant strains. This review discusses the evolution and distribution characteristics of hypervirulent and carbapenem-resistant K. pneumoniae, the mechanisms of carbapenem resistance and hypervirulence, risk factors for susceptibility, infection syndromes, treatment regimens, real-time surveillance and preventive control measures. It also outlines the resistance mechanisms of antimicrobial drugs used to treat this pathogen, providing insights for developing new drugs, combination therapies, and a "One Health" approach. Narrowing the scope of surveillance but intensifying implementation efforts is a viable solution. Monitoring of strains can be focused primarily on hospitals and urban wastewater treatment plants.
Collapse
Affiliation(s)
- Ting-Yu Lei
- College of Pharmaceutical Science, Dali University, Dali 671000, China.
| | - Bin-Bin Liao
- College of Pharmaceutical Science, Dali University, Dali 671000, China.
| | - Liang-Rui Yang
- First Affiliated Hospital of Dali University, Yunnan 671000, China.
| | - Ying Wang
- College of Pharmaceutical Science, Dali University, Dali 671000, China.
| | - Xu-Bing Chen
- College of Pharmaceutical Science, Dali University, Dali 671000, China.
| |
Collapse
|
13
|
Zhu S, Dai D, Li H, Huang J, Kang W, Yang Y, Zhong Y, Xiang Y, Liu C, He J, Liang Z. Bovine Neutrophil β-Defensin-5 Provides Protection against Multidrug-Resistant Klebsiella pneumoniae via Regulating Pulmonary Inflammatory Response and Metabolic Response. Int J Mol Sci 2024; 25:10506. [PMID: 39408834 PMCID: PMC11477005 DOI: 10.3390/ijms251910506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Klebsiella pneumoniae (K. pneumoniae), a kind of zoonotic bacteria, is among the most common antibiotic-resistant pathogens, and it causes nosocomial infections that pose a threat to public health. In this study, the roles of synthetic bovine neutrophil β-defensin-5 (B5) in regulating inflammatory response and metabolic response against multidrug-resistant K. pneumoniae infection in a mouse model were investigated. Mice were administrated intranasally with 20 μg of B5 twice and challenged with K. pneumoniae three days after B5 pretreatment. Results showed that B5 failed to directly kill K. pneumoniae in vitro, but it provided effective protection against multidrug-resistant K. pneumoniae via decreasing the bacterial load in the lungs and spleen, and by alleviating K. pneumoniae-induced histopathological damage in the lungs. Furthermore, B5 significantly enhanced the mRNA expression of TNF-α, IL-1β, Cxcl1, Cxcl5, Ccl17, and Ccl22 and obviously enhanced the rapid recruitment of macrophages and dendritic cells in the lungs in the early infection phase, but significantly down-regulated the levels of TNF-α, IL-1β, and IL-17 in the lungs in the later infection phase. Moreover, RNA-seq results showed that K. pneumoniae infection activated signaling pathways related to natural killer cell-mediated cytotoxicity, IL-17 signaling pathway, inflammatory response, apoptosis, and necroptosis in the lungs, while B5 inhibited these signaling pathways. Additionally, K. pneumoniae challenge led to the suppression of glycerophospholipid metabolism, the phosphotransferase system, the activation of microbial metabolism in diverse environments, and metabolic pathways in the lungs. However, B5 significantly reversed these metabolic responses. Collectively, B5 can effectively regulate the inflammatory response caused by K. pneumoniae and offer protection against K. pneumoniae. B5 may be applied as an adjuvant to the existing antimicrobial therapy to control multidrug-resistant K. pneumoniae infection. Our study highlights the potential of B5 in enhancing pulmonary bacterial clearance and alleviating K. pneumoniae-caused inflammatory damage.
Collapse
Affiliation(s)
- Shuxin Zhu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Dejia Dai
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Han Li
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Jingsheng Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Weichao Kang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Yunmei Yang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Yawen Zhong
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Yifei Xiang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Chengzhi Liu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| |
Collapse
|
14
|
Kew C, Prieto-Garcia C, Bhattacharya A, Tietgen M, MacNair CR, Carfrae LA, Mello-Vieira J, Klatt S, Cheng YL, Rathore R, Gradhand E, Fleming I, Tan MW, Göttig S, Kempf VAJ, Dikic I. The aryl hydrocarbon receptor and FOS mediate cytotoxicity induced by Acinetobacter baumannii. Nat Commun 2024; 15:7939. [PMID: 39261458 PMCID: PMC11390868 DOI: 10.1038/s41467-024-52118-7] [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: 03/07/2023] [Accepted: 08/27/2024] [Indexed: 09/13/2024] Open
Abstract
Acinetobacter baumannii is a pathogenic and multidrug-resistant Gram-negative bacterium that causes severe nosocomial infections. To better understand the mechanism of pathogenesis, we compare the proteomes of uninfected and infected human cells, revealing that transcription factor FOS is the host protein most strongly induced by A. baumannii infection. Pharmacological inhibition of FOS reduces the cytotoxicity of A. baumannii in cell-based models, and similar results are also observed in a mouse infection model. A. baumannii outer membrane vesicles (OMVs) are shown to activate the aryl hydrocarbon receptor (AHR) of host cells by inducing the host enzyme tryptophan-2,3-dioxygenase (TDO), producing the ligand kynurenine, which binds AHR. Following ligand binding, AHR is a direct transcriptional activator of the FOS gene. We propose that A. baumannii infection impacts the host tryptophan metabolism and promotes AHR- and FOS-mediated cytotoxicity of infected cells.
Collapse
Affiliation(s)
- Chun Kew
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
| | - Cristian Prieto-Garcia
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Anshu Bhattacharya
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
| | - Manuela Tietgen
- Institute for Medical Microbiology and Infection Control, Hospital of the Goethe University, Frankfurt, Germany
- University Center of Competence for Infection Control of the State of Hesse, Frankfurt, Germany
| | - Craig R MacNair
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Lindsey A Carfrae
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - João Mello-Vieira
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
| | - Stephan Klatt
- Institute for Vascular Signalling, Department of Molecular Medicine, CPI, Goethe University, Frankfurt, Germany
| | - Yi-Lin Cheng
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Rajeshwari Rathore
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Elise Gradhand
- Department of Pathology, Dr. Senckenberg Institute of Pathology, Goethe University, Frankfurt, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Department of Molecular Medicine, CPI, Goethe University, Frankfurt, Germany
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Stephan Göttig
- Institute for Medical Microbiology and Infection Control, Hospital of the Goethe University, Frankfurt, Germany
| | - Volkhard A J Kempf
- Institute for Medical Microbiology and Infection Control, Hospital of the Goethe University, Frankfurt, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany.
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany.
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Branch Translational Medicine and Pharmacology, Frankfurt, Germany.
- Max Planck Institute of Biophysics, Frankfurt, Germany.
| |
Collapse
|
15
|
Zhang CH, Lu DC, Liu Y, Wang L, Sethi G, Ma Z. The role of extracellular vesicles in pyroptosis-mediated infectious and non-infectious diseases. Int Immunopharmacol 2024; 138:112633. [PMID: 38986299 DOI: 10.1016/j.intimp.2024.112633] [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: 04/06/2024] [Revised: 06/22/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
Pyroptosis, a lytic and pro-inflammatory cell death, is important in various pathophysiological processes. Host- and bacteria-derived extracellular vesicles (EVs), as natural nanocarriers messengers, are versatile mediators of intercellular communication between different types of cells. Recently, emerging research has suggested that EVs exhibit multifaceted roles in disease progression by manipulating pyroptosis. This review focuses on new findings concerning how EVs shape disease progression in infectious and non-infectious diseases by regulating pyroptosis. Understanding the characteristics and activity of EVs-mediated pyroptotic death may conducive to the discovery of novel mechanisms and more efficient therapeutic targets in infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Cai-Hua Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China; Department of Oncology, People's Hospital Affiliated to Chongqing Three Gorges Medical College, Chongqing 404100, China
| | - Ding-Ci Lu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Ying Liu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore; Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore.
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China.
| |
Collapse
|
16
|
Bai D, Zhou C, Du J, Zhao J, Gu C, Wang Y, Zhang L, Lu N, Zhao Y. TrxR1 is involved in the activation of Caspase-11 by regulating the oxidative-reductive status of Trx-1. Redox Biol 2024; 75:103277. [PMID: 39059206 PMCID: PMC11327437 DOI: 10.1016/j.redox.2024.103277] [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: 05/29/2024] [Revised: 07/15/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024] Open
Abstract
Sepsis is a common complication of infections that significantly impacts the survival of critically patients. Currently, effective pharmacological treatment strategies are lacking. Auranofin, known as an inhibitor of Thioredoxin reductase (TrxR), exhibits anti-inflammatory activity, but its role in sepsis is not well understood. Here, we demonstrate the significant inhibitory effect of Auranofin on sepsis in a cecal ligation and puncture (CLP) mouse model. In vitro, Auranofin inhibits pyroptosis triggered by Caspase-11 activation. Further investigations reveal that inhibiting TrxR1 suppresses macrophage pyroptosis induced by E. coli, while TrxR2 does not exhibit this effect. TrxR1, functioning as a reductase, regulates the oxidative-reductive status of Thioredoxin-1 (Trx-1). Mechanistically, the modulation of Trx-1's reductive activity by TrxR1 may be involved in Caspase-11 activation-induced pyroptosis. Additionally, inhibiting TrxR1 maintains Trx-1 in its oxidized state. The oxidized form of Trx-1 interacts with Caveolin-1 (CAV1), regulating outer membrane vesicle (OMV) internalization. In summary, our study suggests that inhibiting TrxR1 suppresses OMV internalization by maintaining the oxidized form of Trx-1, thereby restricting Caspase-11 activation and alleviating sepsis.
Collapse
Affiliation(s)
- Dongsheng Bai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Chen Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Jiaying Du
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Jiawei Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Chunyang Gu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - YuXiang Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Lulu Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China.
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China.
| | - Yue Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China.
| |
Collapse
|
17
|
Qiu Z, Yuan K, Cao H, Chen S, Chen F, Mo F, Guo G, Peng J. Cross-talk of MLST and transcriptome unveiling antibiotic resistance mechanism of carbapenem resistance Acinetobacter baumannii clinical strains isolated in Guiyang, China. Front Microbiol 2024; 15:1394775. [PMID: 38946905 PMCID: PMC11211267 DOI: 10.3389/fmicb.2024.1394775] [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: 03/02/2024] [Accepted: 05/23/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction Acinetobacter baumannii (A. baumannii) is an important opportunistic pathogen causing nosocomial infection in the clinic. The occurrence rate of antibiotic resistance is increasing year by year, resulting in a highly serious situation of bacterial resistance. Methods To better understand the local epidemiology of multidrug-resistant A. baumannii, an investigation was conducted on the antibiotic resistance of different types of A. baumannii and its relationship with the genes of A. baumannii. Furthermore, the molecular mechanism underlying antibiotic resistance in A. baumannii was investigated through transcriptome analysis. Results These results showed that a total of 9 STs were detected. It was found that 99% of the strains isolated in the hospital belonged to the same STs, and the clone complex CC208 was widely distributed in various departments and all kinds of samples. Furthermore, these A. baumannii strains showed high resistance to ertapenem, biapenem, meropenem, and imipenem, among which the resistance to ertapenem was the strongest. The detection rate of bla OXA-51 gene in these carbapenem resistance A. baumannii (CRAB) reached 100%; Additionally, the transcriptome results showed that the resistance genes were up-regulated in resistance strains, and these genes involved in biofilm formation, efflux pumps, peptidoglycan biosynthesis, and chaperonin synthesis. Discussion These results suggest that the CC208 STs were the main clonal complex, and showed high carbapenem antibiotic resistance. All these resistant strains were distributed in various departments, but most of them were distributed in intensive care units (ICU). The bla OXA-23 was the main antibiotic resistance genotype; In summary, the epidemic trend of clinical A. baumannii in Guiyang, China was analyzed from the molecular level, and the resistance mechanism of A. baumannii to carbapenem antibiotics was analyzed with transcriptome, which provided a theoretical basis for better control of A. baumannii.
Collapse
Affiliation(s)
- Zhilang Qiu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Kexin Yuan
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Huijun Cao
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Sufang Chen
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Feifei Chen
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Fei Mo
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Guo Guo
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Basic Medical College, Guizhou Medical University, Guiyang, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, China
| | - Jian Peng
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Basic Medical College, Guizhou Medical University, Guiyang, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, China
| |
Collapse
|
18
|
Jiang B, Huang J. Influences of bacterial extracellular vesicles on macrophage immune functions. Front Cell Infect Microbiol 2024; 14:1411196. [PMID: 38873097 PMCID: PMC11169721 DOI: 10.3389/fcimb.2024.1411196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Bacterial extracellular vesicles (EVs) are crucial mediators of information transfer between bacteria and host cells. Macrophages, as key effector cells in the innate immune system, have garnered widespread attention for their interactions with bacterial EVs. Increasing evidence indicates that bacterial EVs can be internalized by macrophages through multiple pathways, thereby influencing their immune functions. These functions include inflammatory responses, antimicrobial activity, antigen presentation, and programmed cell death. Therefore, this review summarizes current research on the interactions between bacterial EVs and macrophages. This will aid in the deeper understanding of immune modulation mediated by pathogenic microorganisms and provide a basis for developing novel antibacterial therapeutic strategies.
Collapse
Affiliation(s)
- Bowei Jiang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Junyun Huang
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
19
|
Jia Y, Zhang L, Xu J, Xiang L. Recent advances in cell membrane camouflaged nanotherapeutics for the treatment of bacterial infection. Biomed Mater 2024; 19:042006. [PMID: 38697197 DOI: 10.1088/1748-605x/ad46d4] [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: 09/14/2023] [Accepted: 05/01/2024] [Indexed: 05/04/2024]
Abstract
Infectious diseases caused by bacterial infections are common in clinical practice. Cell membrane coating nanotechnology represents a pioneering approach for the delivery of therapeutic agents without being cleared by the immune system in the meantime. And the mechanism of infection treatment should be divided into two parts: suppression of pathogenic bacteria and suppression of excessive immune response. The membrane-coated nanoparticles exert anti-bacterial function by neutralizing exotoxins and endotoxins, and some other bacterial proteins. Inflammation, the second procedure of bacterial infection, can also be suppressed through targeting the inflamed site, neutralization of toxins, and the suppression of pro-inflammatory cytokines. And platelet membrane can affect the complement process to suppress inflammation. Membrane-coated nanoparticles treat bacterial infections through the combined action of membranes and nanoparticles, and diagnose by imaging, forming a theranostic system. Several strategies have been discovered to enhance the anti-bacterial/anti-inflammatory capability, such as synthesizing the material through electroporation, pretreating with the corresponding pathogen, membrane hybridization, or incorporating with genetic modification, lipid insertion, and click chemistry. Here we aim to provide a comprehensive overview of the current knowledge regarding the application of membrane-coated nanoparticles in preventing bacterial infections as well as addressing existing uncertainties and misconceptions.
Collapse
Affiliation(s)
- Yinan Jia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Zhang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junhua Xu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
20
|
Wang Y, Li S, Wang T, Zou M, Peng X. Extracellular Vesicles From Mycoplasma gallisepticum: Modulators of Macrophage Activation and Virulence. J Infect Dis 2024; 229:1523-1534. [PMID: 37929888 DOI: 10.1093/infdis/jiad486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/21/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023] Open
Abstract
Extracellular vesicles (EVs) mediate intercellular communication by transporting proteins. To investigate the pathogenesis of Mycoplasma gallisepticum, a major threat to the poultry industry, we isolated and characterized M. gallisepticum-produced EVs. Our study highlights the significant impact of M. gallisepticum-derived EVs on immune function and macrophage apoptosis, setting them apart from other M. gallisepticum metabolites. These EVs dose-dependently enhance M. gallisepticum adhesion and proliferation, simultaneously modulating Toll-like receptor 2 and interferon γ pathways and thereby inhibiting macrophage activation. A comprehensive protein analysis revealed 117 proteins in M. gallisepticum-derived EVs, including established virulence factors, such as GapA, CrmA, VlhA, and CrmB. Crucially, these EV-associated proteins significantly contribute to M. gallisepticum infection. Our findings advance our comprehension of M. gallisepticum pathogenesis, offering insights for preventive strategies and emphasizing the pivotal role of M. gallisepticum-derived EVs and their associated proteins. This research sheds light on the composition and crucial role of M. gallisepticum-derived EVs in M. gallisepticum pathogenesis, aiding our fight against M. gallisepticum infections.
Collapse
Affiliation(s)
- Yingjie Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shiying Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tengfei Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mengyun Zou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
21
|
Thaden JT, Ahn R, Ruffin F, Gjertson DW, Hoffmann A, Fowler VG, Yeaman MR. Use of Transcriptional Signatures to Differentiate Pathogen-Specific and Treatment-Specific Host Responses in Patients With Bacterial Bloodstream Infections. J Infect Dis 2024; 229:1535-1545. [PMID: 38001039 PMCID: PMC11095544 DOI: 10.1093/infdis/jiad498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Clinical outcomes in bacterial bloodstream infections (BSIs) are influenced by bacterial species, host immunity, and antibiotic therapy. The mechanisms by which such factors influence outcomes are poorly understood. We aimed to identify bacterial- and antibiotic-specific host transcriptional signatures in patients with bacterial BSI. METHODS RNA sequencing was performed on blood samples from patients with BSI due to gram-negative (GN) versus gram-positive (GP) pathogens: Escherichia coli (n = 30) or Klebsiella pneumoniae (n = 28) versus methicillin-susceptible Staphylococcus aureus (MSSA) (n = 24) or methicillin-resistant S. aureus (MRSA) (n = 58). Patients were matched by age, sex, and race. RESULTS No significant host transcriptome differences were detected in patients with E. coli versus K. pneumoniae BSI, so these were considered together as GN BSI. Relative to S. aureus BSI, patients with GN BSI had increased activation of the classic complement system. However, the most significant signal was a reduction in host transcriptional signatures involving mitochondrial energy transduction and oxidative burst in MRSA versus MSSA. This attenuated host transcriptional signature remained after controlling for antibiotic therapy. CONCLUSIONS Given the importance of immune cellular energetics and reactive oxygen species in eliminating hematogenous or intracellular MRSA, these findings may offer insights into its persistence relative to other bacterial BSIs.
Collapse
Affiliation(s)
- Joshua T Thaden
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
| | - Richard Ahn
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
| | - David W Gjertson
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Alexander Hoffmann
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Vance G Fowler
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
| | - Michael R Yeaman
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- Department of Medicine, Divisions of Molecular Medicine and Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, USA
- Institute for Infection & Immunity, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| |
Collapse
|
22
|
Marchant P, Vivanco E, Silva A, Nevermann J, Fuentes I, Barrera B, Otero C, Calderón IL, Gil F, Fuentes JA. β-lactam-induced OMV release promotes polymyxin tolerance in Salmonella enterica sv. Typhi. Front Microbiol 2024; 15:1389663. [PMID: 38591031 PMCID: PMC10999688 DOI: 10.3389/fmicb.2024.1389663] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
The rise of multidrug-resistant bacteria is a global concern, leading to a renewed reliance on older antibiotics like polymyxins as a last resort. Polymyxins, cationic cyclic peptides synthesized nonribosomally, feature a hydrophobic acyl tail and positively charged residues. Their antimicrobial mechanism involves initial interaction with Gram-negative bacterial outer-membrane components through polar and hydrophobic interactions. Outer membrane vesicles (OMVs), nano-sized proteoliposomes secreted from the outer membrane of Gram-negative bacteria, play a crucial role in tolerating harmful molecules, including cationic peptides such as polymyxins. Existing literature has documented environmental changes' impact on modulating OMV properties in Salmonella Typhimurium. However, less information exists regarding OMV production and characteristics in Salmonella Typhi. A previous study in our laboratory showed that S. Typhi ΔmrcB, a mutant associated with penicillin-binding protein (PBP, a β-lactam antibiotic target), exhibited hypervesiculation. Consequently, this study investigated the potential impact of β-lactam antibiotics on promoting polymyxin tolerance via OMVs in S. Typhi. Our results demonstrated that sub-lethal doses of β-lactams increased bacterial survival against polymyxin B in S. Typhi. This phenomenon stems from β-lactam antibiotics inducing hypervesiculation of OMVs with higher affinity for polymyxin B, capturing and diminishing its biologically effective concentration. These findings suggest that β-lactam antibiotic use may inadvertently contribute to decreased polymyxin effectivity against S. Typhi or other Gram-negative bacteria, complicating the effective treatment of infections caused by these pathogens. This study emphasizes the importance of evaluating the influence of β-lactam antibiotics on the interaction between OMVs and other antimicrobial agents.
Collapse
Affiliation(s)
- Pedro Marchant
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Erika Vivanco
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Andrés Silva
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Jan Nevermann
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Ignacio Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Boris Barrera
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Santiago, Chile
| | - Carolina Otero
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Iván L. Calderón
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Fernando Gil
- Microbiota-Host Interactions and Clostridia Research Group, Universidad Andres Bello, Santiago, Chile
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
23
|
Yu MSC, Chiang DM, Reithmair M, Meidert A, Brandes F, Schelling G, Ludwig C, Meng C, Kirchner B, Zenner C, Muller L, Pfaffl MW. The proteome of bacterial membrane vesicles in Escherichia coli-a time course comparison study in two different media. Front Microbiol 2024; 15:1361270. [PMID: 38510998 PMCID: PMC10954253 DOI: 10.3389/fmicb.2024.1361270] [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: 01/10/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction Bacteria inhabit the in- and outside of the human body, such as skin, gut or the oral cavity where they play an innoxious, beneficial or even pathogenic role. It is well known that bacteria can secrete membrane vesicles (MVs) like eukaryotic cells with extracellular vesicles (EVs). Several studies indicate that bacterial membrane vesicles (bMVs) play a crucial role in microbiome-host interactions. However, the composition of such bMVs and their functionality under different culture conditions are still largely unknown. Methods To gain a better insight into bMVs, we investigated the composition and functionality of E. coli (DSM 105380) bMVs from the culture media Lysogeny broth (LB) and RPMI 1640 throughout the different phases of growth (lag-, log- and stationary-phase). bMVs from three time points (8 h, 54 h, and 168 h) and two media (LB and RPMI 1640) were isolated by ultracentrifugation and analyzed using nanoparticle tracking analysis (NTA), cryogenic electron microscopy (Cryo-EM), conventional transmission electron microscopy (TEM) and mass spectrometry-based proteomics (LC-MS/MS). Furthermore, we examined pro-inflammatory cytokines IL-1β and IL-8 in the human monocyte cell line THP-1 upon bMV treatment. Results Particle numbers increased with inoculation periods. The bMV morphologies in Cryo-EM/TEM were similar at each time point and condition. Using proteomics, we identified 140 proteins, such as the common bMV markers OmpA and GroEL, present in bMVs isolated from both media and at all time points. Additionally, we were able to detect growth-condition-specific proteins. Treatment of THP-1 cells with bMVs of all six groups lead to significantly high IL-1β and IL-8 expressions. Conclusion Our study showed that the choice of medium and the duration of culturing significantly influence both E. coli bMV numbers and protein composition. Our TEM/Cryo-EM results demonstrated the presence of intact E. coli bMVs. Common E. coli proteins, including OmpA, GroEL, and ribosome proteins, can consistently be identified across all six tested growth conditions. Furthermore, our functional assays imply that bMVs isolated from the six groups retain their function and result in comparable cytokine induction.
Collapse
Affiliation(s)
- Mia S. C. Yu
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich (TUM), Freising, Germany
| | - Dapi Menglin Chiang
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich (TUM), Freising, Germany
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marlene Reithmair
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany
| | - Agnes Meidert
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Florian Brandes
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Gustav Schelling
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Christina Ludwig
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich (TUM), Freising, Germany
| | - Chen Meng
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich (TUM), Freising, Germany
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich (TUM), Freising, Germany
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany
| | - Christian Zenner
- Intestinal Microbiome, ZIEL – Institute for Food & Health, School of Life Sciences, Technical University of Munich (TUM), Freising, Germany
| | - Laurent Muller
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital of Basel, Basel, Switzerland
| | - Michael W. Pfaffl
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich (TUM), Freising, Germany
| |
Collapse
|
24
|
Zheng P, He J, Fu Y, Yang Y, Li S, Duan B, Yang Y, Hu Y, Yang Z, Wang M, Liu Q, Zheng X, Hua L, Li W, Li D, Ding Y, Yang X, Bai H, Long Q, Huang W, Ma Y. Engineered Bacterial Biomimetic Vesicles Reprogram Tumor-Associated Macrophages and Remodel Tumor Microenvironment to Promote Innate and Adaptive Antitumor Immune Responses. ACS NANO 2024; 18:6863-6886. [PMID: 38386537 DOI: 10.1021/acsnano.3c06987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Tumor-associated macrophages (TAMs) are among the most abundant infiltrating leukocytes in the tumor microenvironment (TME). Reprogramming TAMs from protumor M2 to antitumor M1 phenotype is a promising strategy for remodeling the TME and promoting antitumor immunity; however, the development of an efficient strategy remains challenging. Here, a genetically modified bacterial biomimetic vesicle (BBV) with IFN-γ exposed on the surface in a nanoassembling membrane pore structure was constructed. The engineered IFN-γ BBV featured a nanoscale structure of protein and lipid vesicle, the existence of rich pattern-associated molecular patterns (PAMPs), and the costimulation of introduced IFN-γ molecules. In vitro, IFN-γ BBV reprogrammed M2 macrophages to M1, possibly through NF-κB and JAK-STAT signaling pathways, releasing nitric oxide (NO) and inflammatory cytokines IL-1β, IL-6, and TNF-α and increasing the expression of IL-12 and iNOS. In tumor-bearing mice, IFN-γ BBV demonstrated a targeted enrichment in tumors and successfully reprogrammed TAMs into the M1 phenotype; notably, the response of antigen-specific cytotoxic T lymphocyte (CTL) in TME was promoted while the immunosuppressive myeloid-derived suppressor cell (MDSC) was suppressed. The tumor growth was found to be significantly inhibited in both a TC-1 tumor and a CT26 tumor. It was indicated that the antitumor effects of IFN-γ BBV were macrophage-dependent. Further, the modulation of TME by IFN-γ BBV produced synergistic effects against tumor growth and metastasis with an immune checkpoint inhibitor in an orthotopic 4T1 breast cancer model which was insensitive to anti-PD-1 mAb alone. In conclusion, IFN-γ-modified BBV demonstrated a strong capability of efficiently targeting tumor and tuning a cold tumor hot through reprogramming TAMs, providing a potent approach for tumor immunotherapy.
Collapse
Affiliation(s)
- Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Ying Yang
- Cell Biology & Molecular Biology Laboratory of Experimental Teaching Center, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, People's Republic of China
| | - Shuqin Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Kunming Medical University, Kunming 650500, People's Republic of China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Kunming Medical University, Kunming 650500, People's Republic of China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Kunming Medical University, Kunming 650500, People's Republic of China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Liangqun Hua
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Centers for Disease Control and Prevention, Kunming 530112, People's Republic of China
| | - Yiting Ding
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Hongmei Bai
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| |
Collapse
|
25
|
Fan F, Chen G, Deng S, Wei L. Proteomic analysis of meropenem-induced outer membrane vesicles released by carbapenem-resistant Klebsiella pneumoniae. Microbiol Spectr 2024; 12:e0291723. [PMID: 38236023 PMCID: PMC10846168 DOI: 10.1128/spectrum.02917-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024] Open
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CRKP) is an important multidrug resistance (MDR) pathogen that threatens human health and is the main source of hospital-acquired infection. Outer membrane vesicles (OMVs) are extracellular vesicles derived from Gram-negative bacteria and contain materials involved in bacterial survival and pathogenesis. They also contribute to cellular communication to nearby or distant recipient cells and influence their functions and phenotypes. In this study, we sought to understand the mechanism of bacterial response to meropenem pressure and explore the relationship between pathogenic proteins and the high pathogenicity of bacteria. We performed whole-genome PacBio sequencing on a clinical CRKP strain, and its OMVs were characterized using nanoparticle tracking analysis, transmission electron microscopy, and proteomic analysis. Thousands of vesicle proteins have been identified in mass spectrometry-based high-throughput proteomics analyses of K. pneumoniae OMVs. Protein functionality analysis showed that the OMVs were predominantly involved in metabolic, intracellular compartments, nucleic acid binding, survival, defense, and antibiotic resistance, such as Chromosome partition protein MukB, 3-methyl-2-oxobutanoate hydroxymethyltransferase, methionine-tRNA ligase, Heat shock protein 60 family chaperone GroEL, and Gamma-glutamyl phosphate reductase. Additionally, a protein-protein interaction network demonstrated that OMVs from meropenem-treated K. pneumoniae showed the highest connectivity in DNA polymerase I, phenylalanine-tRNA ligase beta subunit, DNA-directed RNA polymerase subunit beta, methionine-tRNA ligase, DNA-directed RNA polymerase subunit beta, and DNA-directed RNA polymerase subunit alpha. The OMVs proteome expression profile indicates increased secretion of stress proteins released from meropenem-treated K. pneumoniae, which provides clues for revealing the biogenesis and pathophysiological functions of Gram-negative bacteria OMVs. The significant differentially expressed proteins identified in this study are of great significance for exploring effective control strategies for CRKP infection.IMPORTANCEMeropenem is one of the main antibiotics used in the clinical treatment of carbapenem-resistant Klebsiella pneumoniae (CRKP). This study demonstrated that some important metabolic changes occurred in meropenem-induced CRKP-outer membrane vesicles (OMVs), The OMVs proteome expression profile indicates increased secretion of stress proteins released from meropenem-induced Klebsiella pneumoniae. Furthermore, this is the first study to discuss the protein-protein interaction network of the OMVs released by CRKP, especially under antibiotic stress.
Collapse
Affiliation(s)
- Fangfang Fan
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China
| | - Guangzhang Chen
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China
| | - Siqian Deng
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China
| | - Li Wei
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China
| |
Collapse
|
26
|
Liu BD, Akbar R, Oliverio A, Thapa K, Wang X, Fan GC. BACTERIAL EXTRACELLULAR VESICLES IN THE REGULATION OF INFLAMMATORY RESPONSE AND HOST-MICROBE INTERACTIONS. Shock 2024; 61:175-188. [PMID: 37878470 PMCID: PMC10921997 DOI: 10.1097/shk.0000000000002252] [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] [Indexed: 10/27/2023]
Abstract
ABSTRACT Extracellular vesicles (EVs) are a new revelation in cross-kingdom communication, with increasing evidence showing the diverse roles of bacterial EVs (BEVs) in mammalian cells and host-microbe interactions. Bacterial EVs include outer membrane vesicles released by gram-negative bacteria and membrane vesicles generated from gram-positive bacteria. Recently, BEVs have drawn attention for their potential as biomarkers and therapeutic tools because they are nano-sized and can deliver bacterial cargo into host cells. Importantly, exposure to BEVs significantly affects various physiological and pathological responses in mammalian cells. Herein, we provide a comprehensive overview of the various effects of BEVs on host cells (i.e., immune cells, endothelial cells, and epithelial cells) and inflammatory/infectious diseases. First, the biogenesis and purification methods of BEVs are summarized. Next, the mechanisms and pathways identified by BEVs that stimulate either proinflammatory or anti-inflammatory responses are highlighted. In addition, we discuss the mechanisms by which BEVs regulate host-microbe interactions and their effects on the immune system. Finally, this review focuses on the contribution of BEVs to the pathogenesis of sepsis/septic shock and their therapeutic potential for the treatment of sepsis.
Collapse
Affiliation(s)
- Benjamin D. Liu
- Department of Chemistry and Biochemistry, The Ohio State University College of Arts and Sciences, Columbus, OH, 43210, USA
| | - Rubab Akbar
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Anna Oliverio
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kajol Thapa
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xiaohong Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
27
|
Xie J, Haesebrouck F, Van Hoecke L, Vandenbroucke RE. Bacterial extracellular vesicles: an emerging avenue to tackle diseases. Trends Microbiol 2023; 31:1206-1224. [PMID: 37330381 DOI: 10.1016/j.tim.2023.05.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 06/19/2023]
Abstract
A growing body of research, especially in recent years, has shown that bacterial extracellular vesicles (bEVs) are one of the key underlying mechanisms behind the pathogenesis of various diseases like pulmonary fibrosis, sepsis, systemic bone loss, and Alzheimer's disease. Given these new insights, bEVs are proposed as an emerging vehicle that can be used as a diagnostic tool or to tackle diseases when used as a therapeutic target. To further boost the understanding of bEVs in health and disease we thoroughly discuss the contribution of bEVs in disease pathogenesis and the underlying mechanisms. In addition, we speculate on their potential as novel diagnostic biomarkers and how bEV-related mechanisms can be exploited as therapeutic targets.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium; Department of Pathobiology, Pharmacology, and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Freddy Haesebrouck
- Department of Pathobiology, Pharmacology, and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
28
|
Pu D, Zhao J, Chang K, Zhuo X, Cao B. "Superbugs" with hypervirulence and carbapenem resistance in Klebsiella pneumoniae: the rise of such emerging nosocomial pathogens in China. Sci Bull (Beijing) 2023; 68:2658-2670. [PMID: 37821268 DOI: 10.1016/j.scib.2023.09.040] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/19/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
Although hypervirulent Klebsiella pneumoniae (hvKP) can produce community-acquired infections that are fatal in young and adult hosts, such as pyogenic liver abscess, endophthalmitis, and meningitis, it has historically been susceptible to antibiotics. Carbapenem-resistant K. pneumoniae (CRKP) is usually associated with urinary tract infections acquired in hospitals, pneumonia, septicemias, and soft tissue infections. Outbreaks and quick spread of CRKP in hospitals have become a major challenge in public health due to the lack of effective antibacterial treatments. In the early stages of K. pneumoniae development, HvKP and CRKP first appear as distinct routes. However, the lines dividing the two pathotypes are vanishing currently, and the advent of carbapenem-resistant hypervirulent K. pneumoniae (CR-hvKP) is devastating as it is simultaneously multidrug-resistant, hypervirulent, and highly transmissible. Most CR-hvKP cases have been reported in Asian clinical settings, particularly in China. Typically, CR-hvKP develops when hvKP or CRKP acquires plasmids that carry either the carbapenem-resistance gene or the virulence gene. Alternatively, classic K. pneumoniae (cKP) may acquire a hybrid plasmid carrying both genes. In this review, we provide an overview of the key antimicrobial resistance mechanisms, virulence factors, clinical presentations, and outcomes associated with CR-hvKP infection. Additionally, we discuss the possible evolutionary processes and prevalence of CR-hvKP in China. Given the wide occurrence of CR-hvKP, continued surveillance and control measures of such organisms should be assigned a higher priority.
Collapse
Affiliation(s)
- Danni Pu
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, China
| | - Jiankang Zhao
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, China
| | - Kang Chang
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, China
| | - Xianxia Zhuo
- Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, China; Department of Pulmonary and Critical Care Medicine, Capital Medical University, Beijing 100069, China
| | - Bin Cao
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, China; Department of Pulmonary and Critical Care Medicine, Capital Medical University, Beijing 100069, China; Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
29
|
Yadav S, Shah D, Dalai P, Agrawal-Rajput R. The tale of antibiotics beyond antimicrobials: Expanding horizons. Cytokine 2023; 169:156285. [PMID: 37393846 DOI: 10.1016/j.cyto.2023.156285] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/02/2023] [Accepted: 06/24/2023] [Indexed: 07/04/2023]
Abstract
Antibiotics had proved to be a godsend for mankind since their discovery. They were once the magical solution to the vexing problem of infection-related deaths. German scientist Paul Ehrlich had termed salvarsan as the silver bullet to treatsyphilis.As time passed, the magic of newly discovered silver bullets got tarnished with raging antibiotic resistance among bacteria and associated side-effects. Still, antibiotics remain the primary line of treatment for bacterial infections. Our understanding of their chemical and biological activities has increased immensely with advancement in the research field. Non-antibacterial effects of antibiotics are studied extensively to optimise their safer, broad-range use. These non-antibacterial effects could be both useful and harmful to us. Various researchers across the globe including our lab are studying the direct/indirect effects and molecular mechanisms behind these non-antibacterial effects of antibiotics. So, it is interesting for us to sum up the available literature. In this review, we have briefed the possible reason behind the non-antibacterial effects of antibiotics, owing to the endosymbiotic origin of host mitochondria. We further discuss the physiological and immunomodulatory effects of antibiotics. We then extend the review to discuss molecular mechanisms behind the plausible use of antibiotics as anticancer agents.
Collapse
Affiliation(s)
- Shivani Yadav
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Dhruvi Shah
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Parmeswar Dalai
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India.
| |
Collapse
|
30
|
Yang J, Jia F, Qiao Y, Hai Z, Zhou X. Correlation between bacterial extracellular vesicles and antibiotics: A potentially antibacterial strategy. Microb Pathog 2023:106167. [PMID: 37224984 DOI: 10.1016/j.micpath.2023.106167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 05/26/2023]
Abstract
Bacterial extracellular vesicles (BEVs) are proteoliposome nanoparticles that are secreted by both Gram-negative (G-) and Gram-positive (G+) bacteria. BEVs have significant roles in various physiological processes of bacteria, including driving inflammatory responses, regulating bacterial pathogenesis, and promoting bacterial survival in diverse environments. Recently, there has been increasing interest in the use of BEVs as a potential solution to antibiotic resistance. BEVs have shown great promise as a new approach to antibiotics, as well as a drug-delivery tool in antimicrobial strategies. In this review, we provide a summary of recent scientific advances in BEVs and antibiotics, including BEV biogenesis, ability to kill bacteria, potential for delivering antibiotics, and their role in the development of vaccines or as immune adjuvants. We propose that BEVs provide a novel antimicrobial strategy that would be beneficial against the increasing threat of antibiotic resistance.
Collapse
Affiliation(s)
- Jiangliu Yang
- College of Life Science, Ningxia University, Yinchuan, 750021, China; Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, 750021, China
| | - Fang Jia
- Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot, 010058, China
| | - Yarui Qiao
- College of Life Science, Ningxia University, Yinchuan, 750021, China; Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, 750021, China
| | - Zhenzhen Hai
- College of Life Science, Ningxia University, Yinchuan, 750021, China; Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, 750021, China
| | - Xuezhang Zhou
- College of Life Science, Ningxia University, Yinchuan, 750021, China; Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, 750021, China.
| |
Collapse
|
31
|
Doré E, Boilard E. Bacterial extracellular vesicles and their interplay with the immune system. Pharmacol Ther 2023; 247:108443. [PMID: 37210006 DOI: 10.1016/j.pharmthera.2023.108443] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
The mammalian intestinal tract harbors trillions of microorganisms confined within this space by mucosal barriers. Despite these barriers, bacterial components may still be found elsewhere in the body, even in healthy subjects. Bacteria can release small lipid-bound particles, also named bacterial extracellular vesicles (bEV). While bacteria themselves cannot normally penetrate the mucosal defense, bEVs may infiltrate the barrier and disseminate throughout the body. The extremely diverse cargo that bEVs can carry, depending on their parent species, strain, and growth conditions, grant them an equally broad potential to interact with host cells and influence immune functions. Herein, we review the current knowledge of processes underlying the uptake of bEVs by mammalian cells, and their effect on the immune system. Furthermore, we discuss how bEVs could be targeted and manipulated for diverse therapeutic purposes.
Collapse
Affiliation(s)
- Etienne Doré
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada; Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, QC, Canada
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada; Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, QC, Canada.
| |
Collapse
|
32
|
Tang B, Yang A, Liu P, Wang Z, Jian Z, Chen X, Yan Q, Liang X, Liu W. Outer Membrane Vesicles Transmitting blaNDM-1 Mediate the Emergence of Carbapenem-Resistant Hypervirulent Klebsiella pneumoniae. Antimicrob Agents Chemother 2023; 67:e0144422. [PMID: 37052502 DOI: 10.1128/aac.01444-22] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
Dissemination of hypervirulent and carbapenem-resistant Klebsiella pneumoniae (CRKP) has been reported worldwide, posing a serious threat to antimicrobial therapy and public health. Outer membrane vesicles (OMVs) act as vectors for the horizontal transfer of virulence and resistance genes. However, K. pneumoniae OMVs that transfer carbapenem resistance genes into hypervirulent K. pneumoniae (hvKP) have been insufficiently investigated. Therefore, this study investigates the transmission of the blaNDM-1 gene encoding resistance via OMVs released from CRKP and the potential mechanism responsible for the carbapenem-resistant hypervirulent K. pneumoniae (CR-hvKP) emergence. OMVs were isolated via ultracentrifugation from CRKP with or without meropenem selective pressure. OMVs were then used to transform classical K. pneumoniae (ckp) ATCC 10031, extended-spectrum β-lactamase (ESBL)-producing K. pneumoniae ATCC 700603, and hvKP NTUH-K2044. Our results showed that meropenem treatment resulted in changes in the number and diameter of OMVs secreted by CRKP. OMVs derived from CRKP mediated the transfer of blaNDM-1 to ckp and hvKP, thereby increasing the carbapenem MIC of transformants. Further experiments confirmed that NTUH-K2044 transformants exhibited hypervirulence. Our study demonstrates, for the first time, that OMVs derived from CRKP can carry blaNDM-1 and deliver resistance genes to other K. pneumoniae strains, even hvKP. The transfer of carbapenem genes into hypervirulent strains may promote the emergence and dissemination of CR-hvKP. This study elucidates a new mechanism underlying the formation of CR-hvKP.
Collapse
Affiliation(s)
- Bin Tang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Awen Yang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Peilin Liu
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Zhiqian Wang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Zijuan Jian
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Xia Chen
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Qun Yan
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Xianghui Liang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Wenen Liu
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| |
Collapse
|
33
|
Zhang X, Qian C, Tang M, Zeng W, Kong J, Fu C, Xu C, Ye J, Zhou T. Carbapenemase-loaded outer membrane vesicles protect Pseudomonas aeruginosa by degrading imipenem and promoting mutation of antimicrobial resistance gene. Drug Resist Updat 2023; 68:100952. [PMID: 36812748 DOI: 10.1016/j.drup.2023.100952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
AIMS To investigate the effect of Klebsiella pneumoniae carbapenemase (KPC)-loaded outer membrane vesicles (OMVs) in protecting Pseudomonas aeruginosa against imipenem treatment and its mechanism. METHODS The OMVs of carbapenem-resistant Klebsiella pneumonia (CRKP) were isolated and purified from the supernatant of bacterial culture by using ultracentrifugation and Optiprep density gradient ultracentrifugation. The transmission electron microscope, bicinchoninic acid, PCR and carbapenemase colloidal gold assays were applied to characterize the OMVs. Bacterial growth and larvae infection experiments were performed to explore the protective function of KPC-loaded OMVs for P. aeruginosa under imipenem treatment. Ultra-performance liquid chromatography, antimicrobial susceptibility testing, whole-genome sequencing and bioinformatics analysis were used to investigate the mechanism of P. aeruginosa resistance phenotype mediated by OMVs. RESULTS CRKP secreted OMVs loaded with KPC, which protect P. aeruginosa from imipenem through hydrolysis of antibiotics in a dose- and time-dependent manner. Furthermore, carbapenem-resistant subpopulations were developed in P. aeruginosa by low concentrations of OMVs that were confirmed to inadequately hydrolyze imipenem. Interestingly, none of the carbapenem-resistant subpopulations obtained the exogenous antibiotic resistance genes, but all of them possessed OprD mutations, which was consistent with the mechanism of P. aeruginosa induced by sub-minimal inhibitory concentrations of imipenem. CONCLUSIONS OMVs containing KPC provide a novel route for P. aeruginosa to acquire an antibiotic-resistant phenotype in vivo.
Collapse
Affiliation(s)
- Xiaotuan Zhang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Changrui Qian
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Miran Tang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Weiliang Zeng
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jingchun Kong
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Cheng Fu
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chunquan Xu
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jianzhong Ye
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Tieli Zhou
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
34
|
Keeping alert to the hypervirulent K1, K2, K3, K5, K54 and K57 strains of Klebsiella pneumoniae within dairy production process. Microbes Infect 2023; 25:105106. [PMID: 36720402 DOI: 10.1016/j.micinf.2023.105106] [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: 04/20/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/30/2023]
Abstract
Klebsiella pneumoniae (Kp) is now recognized as an urgent threat to public health since the emergence of multidrug-resistant and hypervirulent isolates. We identified a hypervirulent K2 isolate from the milk samples possibly associated with an infection incident in children, which raised the alarm to the zoonotic potential of bovine mastitis Kp as a foodborne pathogen. Subsequently, numerous K1, K2, K3, K5, K54 and K57 strains were identified from mastitis milk samples, and showed high pathogenicity in infected mouse. Further analysis based on complete genomes found that these isolates showed closely evolutionary relationships with the human hypervirulent strains in diverse phylogenetic lineages, suggesting their potential risk to public health.
Collapse
|
35
|
Li Y, Kumar S, Zhang L, Wu H, Wu H. Characteristics of antibiotic resistance mechanisms and genes of Klebsiella pneumoniae. Open Med (Wars) 2023; 18:20230707. [PMID: 37197355 PMCID: PMC10183727 DOI: 10.1515/med-2023-0707] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 04/14/2023] [Accepted: 04/14/2023] [Indexed: 05/19/2023] Open
Abstract
Klebsiella pneumoniae is an important multidrug-resistant (MDR) pathogen that can cause a range of infections in hospitalized patients. With the growing use of antibiotics, MDR K. pneumoniae is more prevalent, posing additional difficulties and obstacles in clinical therapy. To provide a valuable reference to deeply understand K. pneumoniae, and also to provide the theoretical basis for clinical prevention of such bacteria infections, the antibiotic resistance and mechanism of K. pneumoniae are discussed in this article. We conducted a literature review on antibiotic resistance of K. pneumoniae. We ran a thorough literature search of PubMed, Web of Science, and Scopus, among other databases. We also thoroughly searched the literature listed in the papers. We searched all antibiotic resistance mechanisms and genes of seven important antibiotics used to treat K. pneumoniae infections. Antibiotics such as β-lactams, aminoglycosides, and quinolones are used in the treatment of K. pneumoniae infection. With both chromosomal and plasmid-encoded ARGs, this pathogen has diverse resistance genes. Carbapenem resistance genes, enlarged-spectrum β-lactamase genes, and AmpC genes are the most often β-lactamase resistance genes. K. pneumoniae is a major contributor to antibiotic resistance worldwide. Understanding K. pneumoniae antibiotic resistance mechanisms and molecular characteristics will be important for the design of targeted prevention and novel control strategies against this pathogen.
Collapse
Affiliation(s)
- Yanping Li
- Pharmacy Department, Jiangsu Vocational College of Medicine, 224005Yancheng, Jiangsu Province, China
- Post Graduate Centre, Management and Science University, University Drive, Off Persiaran Olahraga, Section 13, 40100, Selangor, Malaysia
| | - Suresh Kumar
- Department of Diagnostic and Allied Health Science, Faculty of Health and Life Sciences, Management and Science University, Shah Alam, Malaysia
| | - Lihu Zhang
- Pharmacy Department, Jiangsu Vocational College of Medicine, 224005Yancheng, Jiangsu Province, China
| | - Hongjie Wu
- School of Electronic and Information Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Hongyan Wu
- Pharmacy Department, Jiangsu Vocational College of Medicine, 224005Yancheng, Jiangsu Province, China
| |
Collapse
|
36
|
Schubert BD, Ku H, Kabwe M, Nguyen TH, Irving H, Tucci J. Effects of Klebsiella pneumoniae Bacteriophages on IRAK3 Knockdown/Knockout THP-1 Monocyte Cell Lines. Viruses 2022; 14:2582. [PMID: 36423191 PMCID: PMC9699088 DOI: 10.3390/v14112582] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
Bacterial sepsis characterised by an immunosuppressive and cytokine storm state is a challenge to treat clinically. While conventional antibiotics have been associated with exacerbating the cytokine storm, the role that bacteriophages may play in immune modulation of sepsis remains unclear. Bacteriophages are bacterial viruses that have the capacity to lyse specific bacteria and hence provide a natural alternative to antibiotics. K. pneumoniae is known to cause sepsis in humans, and in this study we isolated two lytic bacteriophages against this pathogen, one of which was a novel jumbo bacteriophage. We employed THP-1 monocyte cell lines, with different functional phenotypes for the interleukin-1 receptor associated kinase 3 (IRAK3- a cytoplasmic homeostatic mediator and prognostic marker of inflammation), to evaluate the role of the K. pneumoniae bacteriophages in modulating the immune response in-vitro. We showed for the first time that bacteriophages did not stimulate excessive production of tumour necrosis factor alpha, or interleukin-6, in THP-1 monocyte cell lines which displayed varying levels of IRAK3 expression.
Collapse
Affiliation(s)
- Bryce Dylan Schubert
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, P.O. Box 199, Bendigo, VIC 3550, Australia
| | - Heng Ku
- Commonwealth Scientific and Industrial Research Organisation, Oceans & Atmosphere, Ecosciences Precinct, Dutton Park, QLD 4102, Australia
| | - Mwila Kabwe
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, P.O. Box 199, Bendigo, VIC 3550, Australia
- La Trobe Institute for Molecular Science, La Trobe University, P.O. Box 199, Bendigo, VIC 3550, Australia
| | - Trang Hong Nguyen
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, P.O. Box 199, Bendigo, VIC 3550, Australia
- La Trobe Institute for Molecular Science, La Trobe University, P.O. Box 199, Bendigo, VIC 3550, Australia
| | - Helen Irving
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, P.O. Box 199, Bendigo, VIC 3550, Australia
- La Trobe Institute for Molecular Science, La Trobe University, P.O. Box 199, Bendigo, VIC 3550, Australia
| | - Joseph Tucci
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, P.O. Box 199, Bendigo, VIC 3550, Australia
- La Trobe Institute for Molecular Science, La Trobe University, P.O. Box 199, Bendigo, VIC 3550, Australia
| |
Collapse
|
37
|
Mansoor M, Hamer O, Walker E, Hill J. Antibiotics for the Secondary Prevention of Coronary Heart Disease. BRITISH JOURNAL OF CARDIAC NURSING 2022; 17:1-7. [PMID: 38812658 PMCID: PMC7616032 DOI: 10.12968/bjca.2022.0082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
It is estimated that 200 million people are living with coronary heart disease, which remains one of the leading causes of mortality and morbidity worldwide. Those living with coronary heart disease are at an increased risk of cardiovascular events such as stroke, myocardial infarction, and cardiovascular death. Pathophysiology of coronary heart disease revolves around inflammation which leads to plaque build-up. Antibiotics are known to hold anti-inflammatory and anti-oxidative properties. It is theorized that reductions in inflammation could prevent cardiovascular events which may reduce suffering, risk of death and hospital admission rates in patients with coronary heart disease. This article critically appraises a systematic review that assessed the risk of antibiotics used as secondary prevention for coronary heart disease.
Collapse
|
38
|
Chiang MH, Chang FJ, Kesavan DK, Vasudevan A, Xu H, Lan KL, Huang SW, Shang HS, Chuang YP, Yang YS, Chen TL. Proteomic Network of Antibiotic-Induced Outer Membrane Vesicles Released by Extensively Drug-Resistant Elizabethkingia anophelis. Microbiol Spectr 2022; 10:e0026222. [PMID: 35852325 PMCID: PMC9431301 DOI: 10.1128/spectrum.00262-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2022] Open
Abstract
Elizabethkingia anophelis, a nonfermenting Gram-negative bacterium, causes life-threatening health care-associated infections. E. anophelis harbors multidrug resistance (MDR) genes and is intrinsically resistant to various classes of antibiotics. Outer membrane vesicles (OMVs) are secreted by Gram-negative bacteria and contain materials involved in bacterial survival and pathogenesis. OMVs specialize and tailor their functions by carrying different components to challenging environments and allowing communication with other microorganisms or hosts. In this study, we sought to understand the characteristics of E. anophelis OMVs under different antibiotic stress conditions. An extensively drug-resistant clinical isolate, E. anophelis C08, was exposed to multiple antibiotics in vitro, and its OMVs were characterized using nanoparticle tracking analysis, transmission electron microscopy, and proteomic analysis. Protein functionality analysis showed that the OMVs were predominantly involved in metabolism, survival, defense, and antibiotic resistance processes, such as the Rag/Sus family, the chaperonin GroEL, prenyltransferase, and an HmuY family protein. Additionally, a protein-protein interaction network demonstrated that OMVs from imipenem-treated E. anophelis showed significant enrichments in the outer membrane, adenyl nucleotide binding, serine-type peptidase activity, the glycosyl compound metabolic process, and cation binding proteins. Collectively, the OMV proteome expression profile indicates that the role of OMVs is immunologically relevant and related to bacterial survival in antibiotic stress environments rather than representing a resistance point. IMPORTANCE Elizabethkingia anophelis is a bacterium often associated with nosocomial infection. This study demonstrated that imipenem-induced E. anophelis outer membrane vesicles (OMVs) are immunologically relevant and crucial for bacterial survival under antibiotic stress conditions rather than being a source of antibiotic resistance. Furthermore, this is the first study to discuss the protein-protein interaction network of the OMVs released by E. anophelis, especially under antibiotic stress. Our findings provide important insights into clinical antibiotic stewardship.
Collapse
Affiliation(s)
- Ming-Hsien Chiang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Fang-Ju Chang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Dinesh Kumar Kesavan
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Aparna Vasudevan
- International Genomics Research Centre (IGRC), Jiangsu University, Zhenjiang, China
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Huaxi Xu
- International Genomics Research Centre (IGRC), Jiangsu University, Zhenjiang, China
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Kuo-Lun Lan
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Wei Huang
- Department of Orthopedic Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hung-Sheng Shang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Ping Chuang
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Ya-Sung Yang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Te-Li Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
39
|
van Os N, Javed A, Broere F, van Dijk A, Balhuizen MD, van Eijk M, Rooijakkers SHM, Bardoel BW, Heesterbeek DAC, Haagsman HP, Veldhuizen E. Novel insights in antimicrobial and immunomodulatory mechanisms of action of PepBiotics CR-163 and CR-172. J Glob Antimicrob Resist 2022; 30:406-413. [PMID: 35840108 DOI: 10.1016/j.jgar.2022.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Recently our group developed a novel group of antimicrobial peptides termed PepBiotics, of which peptides CR-163 and CR-172 showed optimized antibacterial activity against Pseudomonas aeruginosa and Staphylococcus aureus without inducing antimicrobial resistance. In this study, the antibacterial mechanism of action and the immunomodulatory activity of these two PepBiotics was explored. METHODS RAW264.7 cells were used to determine the ability of PepBiotics to neutralize LPS-and LTA-induced activation of macrophages. Isothermal titration calorimetry and competition assays with dansyl-labeled polymyxin B determined binding characteristics to LPS and LTA. Combined bacterial killing with subsequent macrophage activation assays was performed to determine so-called silent killing'. Finally, flow cytometry of peptide-treated genetically engineered E. coli,expressing GFP and mCherry in the cytoplasm and periplasm, respectively further established the antimicrobial mechanism of PepBiotics. RESULTS Both CR-163 and CR-172 were shown to have broad-spectrum activity against ESKAPE pathogens and E. coli, using a membranolytic mechanism of action. PepBiotics could exothermically bind LPS/LTA and were able to replace polymyxin B. Finally, it was demonstrated that bacteria killed by PepBiotics were less prone to stimulate immune cells, contrary to gentamicin and heat-killed bacteria that still elicited a strong immune response CONCLUSIONS: These studies highlight the multifunctional nature of the two peptide antibiotics as both broad spectrum antimicrobial and immunomodulator. Their ability to kill bacteria and reduce unwanted subsequent immune activation is a major advantage and highlights their potential for future therapeutic use.
Collapse
Affiliation(s)
- Nico van Os
- Department of Biomolecular Health Sciences, Division Infectious Diseases & Immunology, Section Molecular Host Defence, Utrecht University, Utrecht, The Netherlands
| | - Ali Javed
- Department of Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Section Immunology, Utrecht University, Utrecht, The Netherlands
| | - Femke Broere
- Department of Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Section Immunology, Utrecht University, Utrecht, The Netherlands
| | - Albert van Dijk
- Department of Biomolecular Health Sciences, Division Infectious Diseases & Immunology, Section Molecular Host Defence, Utrecht University, Utrecht, The Netherlands
| | - Melanie D Balhuizen
- Department of Biomolecular Health Sciences, Division Infectious Diseases & Immunology, Section Molecular Host Defence, Utrecht University, Utrecht, The Netherlands
| | - Martin van Eijk
- Department of Biomolecular Health Sciences, Division Infectious Diseases & Immunology, Section Molecular Host Defence, Utrecht University, Utrecht, The Netherlands
| | - Suzan H M Rooijakkers
- Department of Medical Microbiology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Bart W Bardoel
- Department of Medical Microbiology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Dani A C Heesterbeek
- Department of Medical Microbiology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Henk P Haagsman
- Department of Biomolecular Health Sciences, Division Infectious Diseases & Immunology, Section Molecular Host Defence, Utrecht University, Utrecht, The Netherlands
| | - Edwin Veldhuizen
- Department of Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Section Immunology, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
40
|
Long Q, Zheng P, Zheng X, Li W, Hua L, Yang Z, Huang W, Ma Y. Engineered bacterial membrane vesicles are promising carriers for vaccine design and tumor immunotherapy. Adv Drug Deliv Rev 2022; 186:114321. [PMID: 35533789 DOI: 10.1016/j.addr.2022.114321] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/18/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023]
Abstract
Bacterial membrane vesicles (BMVs) have emerged as novel and promising platforms for the development of vaccines and immunotherapeutic strategies against infectious and noninfectious diseases. The rich microbe-associated molecular patterns (MAMPs) and nanoscale membrane vesicle structure of BMVs make them highly immunogenic. In addition, BMVs can be endowed with more functions via genetic and chemical modifications. This article reviews the immunological characteristics and effects of BMVs, techniques for BMV production and modification, and the applications of BMVs as vaccines or vaccine carriers. In summary, given their versatile characteristics and immunomodulatory properties, BMVs can be used for clinical vaccine or immunotherapy applications.
Collapse
|
41
|
Villageliu DN, Samuelson DR. The Role of Bacterial Membrane Vesicles in Human Health and Disease. Front Microbiol 2022; 13:828704. [PMID: 35300484 PMCID: PMC8923303 DOI: 10.3389/fmicb.2022.828704] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Bacterial membrane vesicles (MVs) are nanoparticles derived from the membrane components of bacteria that transport microbial derived substances. MVs are ubiquitous across a variety of terrestrial and marine environments and vary widely in their composition and function. Membrane vesicle functional diversity is staggering: MVs facilitate intercellular communication by delivering quorum signals, genetic information, and small molecules active against a variety of receptors. MVs can deliver destructive virulence factors, alter the composition of the microbiota, take part in the formation of biofilms, assist in the uptake of nutrients, and serve as a chemical waste removal system for bacteria. MVs also facilitate host-microbe interactions including communication. Released in mass, MVs overwhelm the host immune system and injure host tissues; however, there is also evidence that vesicles may take part in processes which promote host health. This review will examine the ascribed functions of MVs within the context of human health and disease.
Collapse
Affiliation(s)
| | - Derrick R. Samuelson
- Division of Pulmonary, Critical Care, and Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
42
|
Is the Success of Cefazolin plus Ertapenem in Methicillin-Susceptible
Staphylococcus aureus
Bacteremia Based on Release of Interleukin 1-beta? Antimicrob Agents Chemother 2022; 66:e0216621. [DOI: 10.1128/aac.02166-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cefazolin and ertapenem has been shown to be an effective salvage regimen for refractory methicillin-susceptible
Staphylococcus aureus
bacteremia. Our findings suggest cefazolin plus ertapenem
in vitro
stimulates interleukin-1β release from peripheral blood monocytes both with and without
S. aureus
presence. This IL-1β augmentation was primarily driven by ertapenem. These findings support further exploration of cefazolin plus ertapenem in MSSA bacteremia and may partially explain its marked potency
in vivo
despite modest synergy
in vitro
.
Collapse
|