1
|
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
|
2
|
Wagner TM, Torres-Puig S, Yimthin T, Irobalieva RN, Heller M, Kaessmeyer S, Démoulins T, Jores J. Extracellular vesicles of minimalistic Mollicutes as mediators of immune modulation and horizontal gene transfer. Commun Biol 2025; 8:674. [PMID: 40301684 PMCID: PMC12041197 DOI: 10.1038/s42003-025-08099-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/16/2025] [Indexed: 05/01/2025] Open
Abstract
Extracellular vesicles (EVs) are central components of bacterial secretomes, including the small, cell wall-less Mollicutes. Although EV release in Mollicutes has been reported, EV proteomic composition and function have not been explored yet. We developed a protocol for isolating EVs of the pathogens Mycoplasma mycoides subsp. capri (Mmc) and Mycoplasma (Mycoplasmopsis) bovis and examined their functionality. Proteomic analysis demonstrated that EVs mirror the proteome of the EV-producing bacteria. EVs exhibited nuclease activity, effectively digesting both circular and linear DNA. Notably, M. bovis EVs elicited immune responses in bovine primary blood cells, like those induced by live M. bovis. Our findings reveal that EVs can carry plasmids and enable their horizontal transfer, known as vesiduction. Specifically, the natural plasmid pKMK1, with an unknown transmission route, was detected in EVs of Mmc 152/93 and the tetM-containing pIVB08 plasmid was associated with EVs released by an Mmc GM12 strain carrying this plasmid. pIVB08 could be transferred via homo- and heterologous vesiduction to Mmc, M. capricolum subsp. capricolum and M. leachii. Vesiduction was impeded by membrane disruption but resisted DNase and Proteinase K treatment, suggesting that EVs protect their cargo. These findings enhance our understanding of Mollicutes EVs, particularly in host interactions and horizontal gene transfer.
Collapse
Affiliation(s)
- Theresa Maria Wagner
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland.
| | - Sergi Torres-Puig
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland
| | - Thatcha Yimthin
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Rossitza N Irobalieva
- Division of Veterinary Anatomy, Department of Clinical Research and Veterinary Public Health, University of Bern, Bern, Switzerland
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sabine Kaessmeyer
- Division of Veterinary Anatomy, Department of Clinical Research and Veterinary Public Health, University of Bern, Bern, Switzerland
| | - Thomas Démoulins
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland
| | - Jörg Jores
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland.
- Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, Bern, Switzerland.
| |
Collapse
|
3
|
Xing J, Niu T, Yu T, Zou B, Shi C, Wang Y, Fan S, Li M, Bao M, Sun Y, Gao K, Qiu J, Zhang D, Wang N, Jiang Y, Huang H, Cao X, Zeng Y, Wang J, Zhang S, Hu J, Zhang D, Sun W, Yang G, Yang W, Wang C. Faecalibacterium prausnitzii-derived outer membrane vesicles reprogram gut microbiota metabolism to alleviate Porcine Epidemic Diarrhea Virus infection. MICROBIOME 2025; 13:90. [PMID: 40176190 PMCID: PMC11963522 DOI: 10.1186/s40168-025-02078-x] [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: 10/28/2024] [Accepted: 03/04/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND The Porcine Epidemic Diarrhea Virus (PEDV) is one of the major challenges facing the global pig farming industry, and vaccines and treatments have proven difficult in controlling its spread. Faecalibacterium prausnitzii (F.prausnitzii), a key commensal bacterium in the gut, has been recognized as a promising candidate for next-generation probiotics due to its potential wide-ranging health benefits. A decrease in F.prausnitzii abundance has been associated with certain viral infections, suggesting its potential application in preventing intestinal viral infections. In this study, we utilized a piglet model to examine the potential role of F.prausnitzii in PEDV infections. RESULTS A piglet model of PEDV infection was established and supplemented with F.prausnitzii, revealing that F.prausnitzii mitigated PEDV infection. Further studies found that outer membrane vesicles (OMVs) are the main functional components of F.prausnitzii, and proteomics, untargeted metabolomics, and small RNA-seq were used to analyze the composition of OMVs. Exhaustion of the gut microbiota demonstrated that the function of Fp. OMVs relies on the presence of the gut microbiota. Additionally, metagenomic analysis indicated that Fp. OMVs altered the gut microbiota composition, enhancing the abundance of Faecalibacterium prausnitzii, Prevotellamassilia timonensis, and Limosilactobacillus reuteri. Untargeted metabolomics analysis showed that Fp. OMVs increased phosphatidylcholine (PC) levels, with PC identified as a key metabolite in alleviating PEDV infection. Single-cell sequencing revealed that PC altered the relative abundance of intestinal cells, increased the number of intestinal epithelial cells, and reduced necroptosis in target cells. PC treatment in infected IPEC-J2 and Vero cells alleviated necroptosis and reduced the activation of the RIPK1-RIPK3-MLKL signaling axis, thereby improving PEDV infection. CONCLUSION F.prausnitzii and its OMVs play a critical role in mitigating PEDV infections. These findings provide a promising strategy to ameliorate PEDV infection in piglets. Video Abstract.
Collapse
Affiliation(s)
- JunHong Xing
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - TianMing Niu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Tong Yu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - BoShi Zou
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - ChunWei Shi
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - YingJie Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - ShuHui Fan
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - MingHan Li
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - MeiYing Bao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Yu Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - KuiPeng Gao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - JingJing Qiu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - DongXing Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - YanLong Jiang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - HaiBin Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - JianZhong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - ShuMin Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - JingTao Hu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Di Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - WuSheng Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - GuiLian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| | - WenTao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| | - ChunFeng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| |
Collapse
|
4
|
Rima M, Dakramanji M, El Hayek E, El Khoury T, Fajloun Z, Rima M. Unveiling the wonders of bacteria-derived extracellular vesicles: From fundamental functions to beneficial applications. Heliyon 2025; 11:e42509. [PMID: 40028522 PMCID: PMC11869109 DOI: 10.1016/j.heliyon.2025.e42509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/26/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
Extracellular vesicles (EVs), are critical mediators of intercellular communication and exhibit significant potential across various biomedical domains. These nano-sized, membrane-encapsulated entities have captured substantial interest due to their diverse roles in pathogenesis and promising therapeutic applications. EVs manage numerous physiological processes by transferring bioactive molecules, including proteins, lipids, and nucleic acids, between cells. This review delves into the factors influencing the properties of EVs, such as temperature and stress conditions, which collectively influence their size, composition, and functional attributes. We also describe the emerging roles of EVs, emphasizing their involvement in microbial interactions, immune modulation, antimicrobial resistance spread and their potential as innovative diagnostic and therapeutic instruments. Despite their promising applications, the advancement of EV-based therapies faces several challenges, which will also be discussed. By elucidating these critical elements, we aim to provide a comprehensive overview of the transformative potential of EVs in revolutionizing diagnostics and therapeutics in medicine.
Collapse
Affiliation(s)
- Mariam Rima
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, 1300, Tripoli, Lebanon
| | - Mariam Dakramanji
- Department of Biological Sciences, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Elie El Hayek
- Department of Biological Sciences, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Tia El Khoury
- Department of Biological Sciences, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Ziad Fajloun
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, 1300, Tripoli, Lebanon
- Department of Biology, Faculty of Sciences 3, Campus Michel Slayman Ras Maska, Lebanese University, 1352, Tripoli, Lebanon
| | - Mohamad Rima
- Department of Biological Sciences, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| |
Collapse
|
5
|
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
|
6
|
Wang B, Farhan MHR, Yuan L, Sui Y, Chu J, Yang X, Li Y, Huang L, Cheng G. Transfer dynamics of antimicrobial resistance among gram-negative bacteria. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176347. [PMID: 39306135 DOI: 10.1016/j.scitotenv.2024.176347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 09/09/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
Antimicrobial resistance (AMR) in gram-negative bacteria (GNBs) is a significant global health concern, exacerbated by mobile genetic elements (MGEs). This review examines the transfer of antibiotic resistance genes (ARGs) within and between different species of GNB facilitated by MGEs, focusing on the roles of plasmids and phages. The impact of non-antibiotic chemicals, environmental factors affecting ARG transfer frequency, and underlying molecular mechanisms of bacterial resistance evolution are also discussed. Additionally, the study critically assesses the impact of fitness costs and compensatory evolution driven by MGEs in host organisms, shedding light on the transfer frequency of ARGs and host evolution within ecosystems. Overall, this comprehensive review highlights the factors and mechanisms influencing ARG movement among diverse GNB species and underscores the importance of implementing holistic One-Health strategies to effectively address the escalating public health challenges associated with AMR.
Collapse
Affiliation(s)
- Bangjuan Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Muhammad Haris Raza Farhan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Linlin Yuan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yuxin Sui
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Jinhua Chu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xiaohan Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yuxin Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Guyue Cheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.
| |
Collapse
|
7
|
Shen Z, Qin J, Xiang G, Chen T, Nurxat N, Gao Q, Wang C, Zhang H, Liu Y, Li M. Outer membrane vesicles mediating horizontal transfer of the epidemic blaOXA-232 carbapenemase gene among Enterobacterales. Emerg Microbes Infect 2024; 13:2290840. [PMID: 38044873 PMCID: PMC10810626 DOI: 10.1080/22221751.2023.2290840] [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: 09/05/2023] [Revised: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 12/05/2023]
Abstract
OXA-232 is one of the most common OXA-48-like carbapenemase derivatives and is widely disseminated in nosocomial settings across countries. The blaOXA-232 gene is located on a 6-kb non-conjugative ColKP3-type plasmid, while the dissemination of blaOXA-232 into different Enterobacterales species and the polyclonal dissemination of OXA-232-producing K. pneumoniae revealed the horizontal transfer of blaOXA-232. However, it's still unclear how this non-conjugative ColKP3 plasmid could facilitate the mobilization of blaOXA-232. Here, we observed the in vivo intraspecies transfer of blaOXA-232 during a nosocomial outbreak of OXA-232-producing K. pneumoniae. We demonstrated the presence of ColKP3 OXA-232 plasmid in the outer membrane vesicles (OMVs) derived from clinical isolates, and OMVs could facilitate the horizontal transfer of blaOXA-232 among Enterobacterales. In contrast, for the most prevalent carbapenemase genes, including blaKPC-2 and blaNDM-1, though the presence of carbapenemase genes and plasmid backbones in the vesicular lumen was observed, OMVs couldn't promote effective transformation, probably due to the low copy number of plasmids in clinical isolates and the low number of plasmids loaded into vesicles. Conjugation assay revealed that the epidemic IncX3 NDM-1 and IncFII(pHN7A8)/IncR KPC-2 plasmids were conjugative and could be horizontally transferred via independent conjugation or with the help of a co-existent conjugative plasmid. For the large-size and low-copy number conjugative plasmids carrying carbapenemase genes, OMVs-mediated gene exchange may only serve as an alternative pathway for horizontal transfer. In conclusion, diverse mobilization strategies were employed by plasmids harbouring carbapenemase genes, and plasmids display a proper choice of mobility pathway due to their individual properties.
Collapse
Affiliation(s)
- Zhen Shen
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Juanxiu Qin
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Guoxiu Xiang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Tianchi Chen
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Nadira Nurxat
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qianqian Gao
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Chen Wang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Haomin Zhang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yao Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
8
|
Li J, Li C, Han Y, Yang J, Hu Y, Xu H, Zhou Y, Zuo J, Tang Y, Lei C, Li C, Wang H. Bacterial membrane vesicles from swine farm microbial communities harboring and safeguarding diverse functional genes promoting horizontal gene transfer. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175639. [PMID: 39168346 DOI: 10.1016/j.scitotenv.2024.175639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/05/2024] [Accepted: 08/17/2024] [Indexed: 08/23/2024]
Abstract
Antibiotic resistance (AMR) poses a significant global health challenge, with swine farms recognized as major reservoirs of antibiotic resistance genes (ARGs). Recently, bacterial membrane vesicles (BMVs) have emerged as novel carriers mediating horizontal gene transfer. However, little is known about the ARGs carried by BMVs in swine farm environments and their transfer potential. This study investigated the distribution, sources, and microbiological origins of BMVs in three key microbial habitats of swine farms (feces, soil, and fecal wastewater), along with the ARGs and mobile genetic elements (MGEs) they harbor. Characterization of BMVs revealed particle sizes ranging from 20 to 500 nm and concentrations from 108 to 1012 particles/g, containing DNA and proteins. Metagenomic sequencing identified BMVs predominantly composed of members of the Proteobacteria phyla, including Pseudomonadaceae, Moraxellaceae, and Enterobacteriaceae, carrying diverse functional genes encompassing resistance to 14 common antibiotics and 74,340 virulence genes. Notably, multidrug resistance, tetracycline, and chloramphenicol resistance genes were particularly abundant. Furthermore, BMVs harbored various MGEs, primarily plasmids, and demonstrated the ability to protect their DNA cargo from degradation and facilitate horizontal gene transfer, including the transmission of resistance genes. In conclusion, this study reveals widespread presence of BMVs carrying ARGs and potential virulence genes in swine farm feces, soil, and fecal wastewater. These findings not only provide new insights into the role of extracellular DNA in the environment but also highlight concerns regarding the gene transfer potential mediated by BMVs and associated health risks.
Collapse
Affiliation(s)
- Jinpeng Li
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Chao Li
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650000, China.
| | - Yun Han
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Jian Yang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Yulian Hu
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Heting Xu
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Yi Zhou
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Jing Zuo
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Yizhi Tang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Changwei Lei
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Cui Li
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China
| | - Hongning Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu 610000, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China.
| |
Collapse
|
9
|
Xuan S, Xuan G. Bacterial membrane vesicles: formation, functions, and roles in bacterial-phage interactions. World J Microbiol Biotechnol 2024; 40:329. [PMID: 39304539 DOI: 10.1007/s11274-024-04148-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: 07/25/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Outer membrane vesicles (OMVs) are nano-sized vesicles actively released by Gram-negative bacteria, playing a crucial role in bacterial survival and interactions with phages. This review focuses on OMVs and succinctly delineates the stimuli instigating OMV formation, their functional repertoire, and their involvement in bacterial-phage interplays. Initially, the discussion centers on the drivers prompting OMV genesis, encompassing both extrinsic environmental pressures and intrinsic regulatory mechanisms within bacterial systems. Subsequently, a comprehensive examination of OMVs' multifaceted functions in bacterial physiology ensues, spanning signaling cascades, nutrient transport, antibiotic resilience, and evasion of immune surveillance. Particular emphasis is placed on elucidating the paramount significance of OMVs in mediating bacterial-phage dynamics. OMVs function as decoys, providing protection to bacterial hosts against phages, and concurrently promoting the spread of phage receptors, thereby rendering phage-resistant strains susceptible to phage invasion. This comprehensive review deepens our comprehension of membrane vesicles biogenesis in bacteria and their pivotal role in microbial community dynamics.
Collapse
Affiliation(s)
- Shichao Xuan
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, 266400, China
| | - Guanhua Xuan
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, 266400, China.
| |
Collapse
|
10
|
Zeng Y, Li T, Chen X, Fang X, Fang C, Liang X, Liu J, Yang Y. Oral administration of Lactobacillus plantarum expressing aCD11c modulates cellular immunity alleviating inflammatory injury due to Klebsiella pneumoniae infection. BMC Vet Res 2024; 20:399. [PMID: 39244529 PMCID: PMC11380324 DOI: 10.1186/s12917-024-04248-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Klebsiella pneumoniae (KP), responsible for acute lung injury (ALI) and inflammation of the gastrointestinal tract, is a zoonotic pathogen that poses a threat to livestock farming worldwide. Nevertheless, there is currently no validated vaccine to prevent KP infection. The development of mucosal vaccines against KP using Lactobacillus plantarum (L. plantarum) is an effective strategy. RESULTS Firstly, the L. plantarum strains NC8-pSIP409-aCD11c' and NC8-pLc23-aCD11c were constructed via homologous recombination to express the aCD11c protein either inducibly or constitutively. Both NC8-pSIP409-aCD11c' and NC8-pLc23-aCD11c strains could enhance the adhesion and invasion of L. plantarum on bone marrow-derived dendritic cells (BMDCs), and stimulate the activation of BMDCs compared to the control strain NC8-pSIP409 in vitro. Following oral immunization of mice with NC8-pSIP409-aCD11c' and NC8-pLc23-aCD11c, the cellular, humoral, and mucosal immunity were significantly improved, as evidenced by the increased expression of CD4+ IL-4+ T cells in the spleen, IgG in serum, and secretory IgA (sIgA) in the intestinal lavage fluid (ILF). Furthermore, the protective effects of L. plantarum against inflammatory damage caused by KP infection were confirmed by assessing the bacterial loads in various tissues, lung wet/dry ratio (W/D), levels of inflammatory cytokines, and histological evaluation, which influenced T helper 17 (Th17) and regulatory T (Treg) cells in peripheral blood and lung. CONCLUSIONS Both the inducible and constitutive L. plantarum strains NC8-pSIP409-aCD11c' and NC8-pLc23-aCD11c have been found to stimulate cellular and humoral immunity levels and alleviate the inflammatory response caused by KP infection. These findings have provided a basis for the development of a novel vaccine against KP.
Collapse
Affiliation(s)
- Yang Zeng
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Tiantian Li
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Xueyang Chen
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Xiaowei Fang
- College of Agriculture, Yangtze University, Jingzhou, 434025, China
| | - Chun Fang
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Xiongyan Liang
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China
| | - Jing Liu
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China.
| | - Yuying Yang
- College of Animal Science and Technology, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
11
|
Xu M, Ke H, Zang Y, Gou H, Yang D, Shi K, Zhang K, Li Y, Jiang Z, Chu P, Zhai S, Li C. Outer membrane vesicles secreted from Actinobacillus pleuropneumoniae isolate disseminating the floR resistance gene to Enterobacteriaceae. Front Microbiol 2024; 15:1467847. [PMID: 39301187 PMCID: PMC11410613 DOI: 10.3389/fmicb.2024.1467847] [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: 07/21/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
Actinobacillus pleuropneumoniae, a significant respiratory pig pathogen, is causing substantial losses in the global swine industry. The resistance spectrum of A. pleuropneumoniae is expanding, and multidrug resistance is a severe issue. Horizontal gene transfer (HGT) plays a crucial role in the development of the bacterial genome by facilitating the dissemination of resistance determinants. However, the horizontal transfer of resistance genes via A. pleuropneumoniae-derived outer membrane vesicles (OMVs) has not been previously reported. In this study, we used Illumina NovaSeq and PacBio SequeI sequencing platforms to determine the whole genome sequence of A. pleuropneumoniae GD2107, a multidrug-resistant (MDR) isolate from China. We detected a plasmid in the isolate named pGD2107-1; the plasmid was 5,027 bp in size with 7 putative open reading frames (ORF) and included the floR resistance genes. The carriage of resistance genes in A. pleuropneumoniae OMVs was identified using a polymerase chain reaction (PCR) assay, and then we thoroughly evaluated the influence of OMVs on the horizontal transfer of drug-resistant plasmids. The transfer of the plasmid to recipient bacteria via OMVs was confirmed by PCR. In growth competition experiments, all recipients carrying the pGD2107-1 plasmid exhibited a fitness cost compared to the corresponding original recipients. This study revealed that OMVs could mediate interspecific horizontal transfer of the resistance plasmid pGD2107-1 into Escherichia coli recipient strains and significantly enhance the resistance of the transformants. In summary, A. pleuropneumoniae-OMVs play the pivotal role of vectors for dissemination of the floR gene spread and may contribute to more antimicrobial resistance gene transfer in other Enterobacteriaceae.
Collapse
Affiliation(s)
- Minsheng Xu
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Haiyi Ke
- Guangdong Gaozhou Agricultural School, Maoming, China
| | - Yingan Zang
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Hongchao Gou
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Dongxia Yang
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Keda Shi
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Kunli Zhang
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Yan Li
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Zhiyong Jiang
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Pinpin Chu
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Shaolun Zhai
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Chunling Li
- Guangdong Academy of Agricultural Sciences, Institute of Animal Health, Guangzhou, China
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China
- Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| |
Collapse
|
12
|
Sun H, Xiao D, Li X, Sun T, Meng F, Shao X, Ding Y, Li Y. Study on the chemical composition and anti-fungi activities of anthraquinones and its glycosides from Rumex japonicus Houtt. J Nat Med 2024; 78:929-951. [PMID: 39103726 DOI: 10.1007/s11418-024-01834-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/18/2024] [Indexed: 08/07/2024]
Abstract
Fungi, such as Trichophyton rubrum (T. rubrum) and Microsporum canis Bodin Anamorph (M. canis Bodin Anamorph) are the main pathogens of dermatophysis. According to ancient books records, Rumex japonicus Houtt. (RJH) has a miraculous effect on the treatment of dermatophysis. To reveal the anti-fungi (T. rubrum and M. canis Bodin Anamorph) components and its mechanism of the Rumex japonicus Houtt. The vinegar extraction and alcohol precipitation, HPLC and nuclear magnetic resonance spectroscopy (NMR) were employed for analyzing the chemical compositions of RJH; in vitro anti-fungal experiment was investigated including test the minimum inhibitory concentration (MIC) and the minimum fungicidal concentration (MFC), spore germination rate, nucleic acid, protein leakage rate, biofilm structure, and the mechanism of anti-fungal and anti-fungal biofilms in RJH. Seven anthraquinones and their glycoside compounds were obtained in this study respectively, such as chrysophanol, physcion, aloe-emodin, emodin, rhein, emodin-8-O-β-D-glucoside and chrysophanol-8-O-β-D-glucoside. In vitro anti-fungal experiment results showed that RJH extracts have good anti-fungal activity for dermatophytic fungi. Among them, the MIC of the rhein, emodin and aloe-emodin against T. rubrum are 1.9 µg/ml, 3.9 µg/ml and 15.6 µg/ml, respectively; the MIC of emodin and aloe-emodin against M. canis Bodin Anamorph are 7.8 µg/ml and 62.5 µg/ml, respectively. In addition, its active components can inhibit fungal spore germination and the formation of bud tube, change cell membrane permeability, prevent hyphal growth, destroy biofilm structure, and down-regulate the expression of agglutinin-like sequence family 1 of the adhesion phase of biofilm growth. The study shows that RJH play a fungicidal role.
Collapse
Affiliation(s)
- He Sun
- Department of School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, People's Republic of China
| | - Dandan Xiao
- Department of Marine Life Sciences, Jeju National University, Jeju, 690-756, Korea
| | - Xue Li
- Department of School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, People's Republic of China
| | - Tong Sun
- Department of School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, People's Republic of China
| | - Fanying Meng
- Department of School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, People's Republic of China
| | - Xinting Shao
- Department of School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, People's Republic of China
| | - Yuling Ding
- Department of School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, People's Republic of China.
| | - Yong Li
- Department of School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, People's Republic of China.
| |
Collapse
|
13
|
Rocha Minarini LAD. Exploring bacterial extracellular vesicles: Focus on WHO critical priority pathogens. CURRENT TOPICS IN MEMBRANES 2024; 94:225-246. [PMID: 39370208 DOI: 10.1016/bs.ctm.2024.06.009] [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: 10/08/2024]
Abstract
Bacterial extracellular vesicles (EVs) are cell-derived particles with a phospholipidic bilayer structure and diameter ranging from 20 to 250 nm, comprising a varied of components, including bioactive proteins, lipids, DNA, RNA, and other metabolites. These EVs play an essential role in bacterial and host function and are recognized as essential keys in cell-to-cell communication and pathogenesis. Due to these characteristics and functions, EVs exhibit great potential for biomedical applications and are promising tools for the development of drug delivery systems and vaccines, as well as for use in disease diagnostics. An interesting focus of this review is on the clinical relevance of EVs, with a particular emphasis on two critical pathogens, Acinetobacter baumannii and Klebsiella pneumoniae. Insights into the outer membrane vesicles (OMVs) derived from these bacteria underscore their roles in antimicrobial resistance and pathogenicity. Additionally, the review explores OMV-based vaccine strategies as a promising means to mitigating these pathogens.
Collapse
Affiliation(s)
- Luciene Andrade da Rocha Minarini
- Laboratório Multidisciplinar em Saúde e Meio Ambiente, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Campus Diadema, Diadema, SP, Brazil.
| |
Collapse
|
14
|
Zhang Y, Li W, Wu Y, Tian X, Li G, Zhou Y, Sun J, Liao X, Liu Y, Wang Y, Yu Y. Chitosan oligosaccharide accelerates the dissemination of antibiotic resistance genes through promoting conjugative plasmid transfer. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:133922. [PMID: 38442604 DOI: 10.1016/j.jhazmat.2024.133922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024]
Abstract
The dissemination of antibiotic resistance genes (ARGs), especially via plasmid-mediated horizontal gene transfer, poses a pervasive threat to global health. Chitosan-oligosaccharide (COS) is extensively utilized in medicine, plant and animal husbandry. However, their impact on microflora implies the potential to exert selective pressure on plasmid transfer. To explore the role of COS in facilitating the dissemination of ARGs via plasmid conjugation, we established in vitro mating models. The addition of COS to conjugation mixtures significantly enhanced the transfer of RP4 plasmid and mcr-1 positive IncX4 plasmid in both intra- and inter-specific. Phenotypic and transcriptome analysis revealed that COS enhanced intercellular contact by neutralizing cell surface charge and increasing cell surface hydrophobicity. Additionally, COS increased membrane permeability by inhibiting the Tol-Pal system, thereby facilitating plasmid conjugative transfer. Furthermore, COS served as the carbon source and was metabolized by E. coli, providing energy for plasmid conjugation through regulating the expression of ATPase and global repressor factor-related genes in RP4 plasmid. Overall, these findings improve our awareness of the potential risks associated with the presence of COS and the spread of bacterial antibiotic resistance, emphasizing the need to establish guidelines for the prudent use of COS and its discharge into the environment.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Wenjie Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Yashuang Wu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Xiaomin Tian
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Gong Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Yufeng Zhou
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Xiaoping Liao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Yahong Liu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yang Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yang Yu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
15
|
Burt M, Angelidou G, Mais CN, Preußer C, Glatter T, Heimerl T, Groß R, Serrania J, Boosarpu G, Pogge von Strandmann E, Müller JA, Bange G, Becker A, Lehmann M, Jonigk D, Neubert L, Freitag H, Paczia N, Schmeck B, Jung AL. Lipid A in outer membrane vesicles shields bacteria from polymyxins. J Extracell Vesicles 2024; 13:e12447. [PMID: 38766978 PMCID: PMC11103557 DOI: 10.1002/jev2.12447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
The continuous emergence of multidrug-resistant bacterial pathogens poses a major global healthcare challenge, with Klebsiella pneumoniae being a prominent threat. We conducted a comprehensive study on K. pneumoniae's antibiotic resistance mechanisms, focusing on outer membrane vesicles (OMVs) and polymyxin, a last-resort antibiotic. Our research demonstrates that OMVs protect bacteria from polymyxins. OMVs derived from Polymyxin B (PB)-stressed K. pneumoniae exhibited heightened protective efficacy due to increased vesiculation, compared to OMVs from unstressed Klebsiella. OMVs also shield bacteria from different bacterial families. This was validated ex vivo and in vivo using precision cut lung slices (PCLS) and Galleria mellonella. In all models, OMVs protected K. pneumoniae from PB and reduced the associated stress response on protein level. We observed significant changes in the lipid composition of OMVs upon PB treatment, affecting their binding capacity to PB. The altered binding capacity of single OMVs from PB stressed K. pneumoniae could be linked to a reduction in the lipid A amount of their released vesicles. Although the amount of lipid A per vesicle is reduced, the overall increase in the number of vesicles results in an increased protection because the sum of lipid A and therefore PB binding sites have increased. This unravels the mechanism of the altered PB protective efficacy of OMVs from PB stressed K. pneumoniae compared to control OMVs. The lipid A-dependent protective effect against PB was confirmed in vitro using artificial vesicles. Moreover, artificial vesicles successfully protected Klebsiella from PB ex vivo and in vivo. The findings indicate that OMVs act as protective shields for bacteria by binding to polymyxins, effectively serving as decoys and preventing antibiotic interaction with the cell surface. Our findings provide valuable insights into the mechanisms underlying antibiotic cross-protection and offer potential avenues for the development of novel therapeutic interventions to address the escalating threat of multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Marie Burt
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
| | - Georgia Angelidou
- Core Facility for Metabolomics and Small Molecules Mass SpectrometryMax Planck Institute for Terrestrial MicrobiologyMarburgGermany
- Core Facility for Mass Spectrometry and ProteomicsMax Planck Institute for terrestrial MicrobiologyMarburgGermany
| | - Christopher Nils Mais
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Christian Preußer
- Institute for Tumor ImmunologyPhilipps‐University MarburgMarburgGermany
- Core Facility ‐ Extracellular VesiclesPhilipps‐University MarburgMarburgGermany
| | - Timo Glatter
- Core Facility for Mass Spectrometry and ProteomicsMax Planck Institute for terrestrial MicrobiologyMarburgGermany
| | - Thomas Heimerl
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Rüdiger Groß
- Institute of Molecular VirologyUlm University Medical CenterUlmGermany
| | - Javier Serrania
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Gowtham Boosarpu
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
| | - Elke Pogge von Strandmann
- Institute for Tumor ImmunologyPhilipps‐University MarburgMarburgGermany
- Core Facility ‐ Extracellular VesiclesPhilipps‐University MarburgMarburgGermany
| | - Janis A. Müller
- Institute of VirologyPhilipps‐University MarburgMarburgGermany
| | - Gert Bange
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Anke Becker
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Mareike Lehmann
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
- Comprehensive Pneumology Center (CPC), Institute of Lung Health and ImmunityHelmholtz Zentrum MünchenGerman Center for Lung Research (DZL)MunichGermany
- Institute for Lung Health (ILH)GiessenGermany
| | - Danny Jonigk
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH)German Center of Lung Research (DZL)HannoverGermany
- Institute of PathologyUniversity Medical Center RWTH University of AachenAachenGermany
| | - Lavinia Neubert
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH)German Center of Lung Research (DZL)HannoverGermany
- Institute of PathologyHannover Medical SchoolHannoverGermany
| | - Hinrich Freitag
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH)German Center of Lung Research (DZL)HannoverGermany
- Institute of PathologyHannover Medical SchoolHannoverGermany
| | - Nicole Paczia
- Core Facility for Metabolomics and Small Molecules Mass SpectrometryMax Planck Institute for Terrestrial MicrobiologyMarburgGermany
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
- Institute for Lung Health (ILH)GiessenGermany
- Department of Medicine, Pulmonary and Critical Care MedicineUniversity Medical Center MarburgUniversities of Giessen and Marburg Lung CenterPhilipps‐University MarburgMarburgGermany
- Member of the German Center for Infectious Disease Research (DZIF)MarburgGermany
- Core Facility Flow Cytometry – Bacterial VesiclesPhilipps‐University MarburgMarburgGermany
| | - Anna Lena Jung
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
- Core Facility Flow Cytometry – Bacterial VesiclesPhilipps‐University MarburgMarburgGermany
| |
Collapse
|
16
|
Dell'Annunziata F, Ciaglia E, Folliero V, Lopardo V, Maciag A, Galdiero M, Puca AA, Franci G. Klebsiella pneumoniae-OMVs activate death-signaling pathways in Human Bronchial Epithelial Host Cells (BEAS-2B). Heliyon 2024; 10:e29017. [PMID: 38644830 PMCID: PMC11031753 DOI: 10.1016/j.heliyon.2024.e29017] [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: 08/02/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/23/2024] Open
Abstract
The programmed cell death pathways of apoptosis are important in mammalian cellular protection from infections. The activation of these pathways depends on the presence of membrane receptors that bind bacterial components to activate the transduction mechanism. In addition to bacteria, these mechanisms can be activated by outer membrane vesicles (OMVs). OMVs are spherical vesicles of 20-250 nm diameter, constitutively released by Gram-negative bacteria. They contain several bacterial determinants including proteins, DNA/RNA and proteins, that activate different cellular processes in host cells. This study focused on Klebsiella pneumoniae-OMVs in activating death mechanisms in human bronchial epithelial cells (BEAS-2B). Characterization of purified OMVs was achieved by scanning electron microscopy, nanoparticle tracking analysis and protein profiling. Cell viability was assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay while apoptotic induction was measured by flow cytometry and confirmed by western blotting. The OMVs produced showed a spherical morphology, with a diameter of 137.2 ± 41 nm and a vesicular density of 7.8 × 109 particles/mL Exposure of cell monolayers to 50 μg of K. pneumoniae-OMV for 14 h resulted in approximately 25 % cytotoxicity and 41.15-41.14 % of cells undergoing early and late apoptosis. Fluorescence microscopy revealed reduced cellular density, the presence of apoptotic bodies, chromatin condensation, and nuclear membrane blebbing in residual cells. Activation of caspases -3 and -9 and dysregulation of BAX, BIM and Bcl-xL indicated the activation of mitochondria-dependent apoptosis. Furthermore, a decrease in the antioxidant enzymes superoxide dismutase, catalase and glutathione peroxidase involved endoplasmic reticulum stress with the potential formation of reactive oxygen species. These findings provide evidence for the role of OMVs in apoptosis and involvement in the pathogenesis of K. pneumoniae infections.
Collapse
Affiliation(s)
- Federica Dell'Annunziata
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, Salerno, Italy
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, Salerno, Italy
| | - Veronica Folliero
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, Salerno, Italy
| | - Valentina Lopardo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, Salerno, Italy
| | - Anna Maciag
- Cardiovascular Research Unit, IRCCS MultiMedica, 20138, Milan, Italy
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
- Complex Operative Unity of Virology and Microbiology, University Hospital of Campania “Luigi Vanvitelli", 80138, Naples, Italy
| | - Annibale Alessandro Puca
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, Salerno, Italy
- Cardiovascular Research Unit, IRCCS MultiMedica, 20138, Milan, Italy
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, Salerno, Italy
- Clinical Pathology and Microbiology Unit, San Giovanni di Dio e Ruggi D'Aragona University Hospital, 84126, Salerno, Italy
| |
Collapse
|
17
|
Muñoz-Echeverri LM, Benavides-López S, Geiger O, Trujillo-Roldán MA, Valdez-Cruz NA. Bacterial extracellular vesicles: biotechnological perspective for enhanced productivity. World J Microbiol Biotechnol 2024; 40:174. [PMID: 38642254 PMCID: PMC11032300 DOI: 10.1007/s11274-024-03963-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/19/2024] [Indexed: 04/22/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are non-replicative nanostructures released by Gram-negative and Gram-positive bacteria as a survival mechanism and inter- and intraspecific communication mechanism. Due to BEVs physical, biochemical, and biofunctional characteristics, there is interest in producing and using them in developing new therapeutics, vaccines, or delivery systems. However, BEV release is typically low, limiting their application. Here, we provide a biotechnological perspective to enhance BEV production, highlighting current strategies. The strategies include the production of hypervesiculating strains through gene modification, bacteria culture under stress conditions, and artificial vesicles production. We discussed the effect of these production strategies on BEVs types, morphology, composition, and activity. Furthermore, we summarized general aspects of BEV biogenesis, functional capabilities, and applications, framing their current importance and the need to produce them in abundance. This review will expand the knowledge about the range of strategies associated with BEV bioprocesses to increase their productivity and extend their application possibilities.
Collapse
Affiliation(s)
- Laura M Muñoz-Echeverri
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán CDMX, C.P. 04510, México
| | - Santiago Benavides-López
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México
- Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio B, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán CDMX, C.P. 04510, México
| | - Otto Geiger
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Av. Universidad s/n, Cuernavaca, Morelos, CP 62210, México
| | - Mauricio A Trujillo-Roldán
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 Carretera, Tijuana-Ensenada, Baja California, 22860, México
| | - Norma A Valdez-Cruz
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México.
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 Carretera, Tijuana-Ensenada, Baja California, 22860, México.
| |
Collapse
|
18
|
Zhao M, He S, Wen R, Li C, Chen X, Lin X, Wang H, Tang Y. Membrane vesicles derived from Enterococcus faecalis promote the co-transfer of important antibiotic resistance genes located on both plasmids and chromosomes. J Antimicrob Chemother 2024; 79:320-326. [PMID: 38109479 DOI: 10.1093/jac/dkad381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/28/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Bacterial membrane vesicles (BMVs) are novel vehicles of antibiotic resistance gene (ARG) transfer in Gram-negative bacteria, but their role in the spread of ARGs in Gram-positive bacteria has not been defined. The purpose of this study was to evaluate the role of MVs in the transmission of antimicrobial resistance in Gram-positive bacteria. METHODS A linezolid-resistant Enterococcus faecalis CQ20 of swine origin was selected as the donor strain. Linezolid-susceptible E. faecalis SC032 of human origin, Enterococcus faecium BM4105 and Escherichia coli were selected as recipient strains. The presence of plasmids (pCQ20-1 and pCQ20-2) and an optrA-carrying transposon Tn6674 in CQ20, MVs and vesiculants was verified by WGS or PCR. MVs were isolated with density gradient centrifugation, and MV-mediated transformation was performed to assess the horizontal transferability of MVs. The MICs for CQ20 and its vesiculants were determined by the broth microdilution method. RESULTS CQ20-derived MVs (CQ20-MV) were isolated, and PCR identified the presence of two plasmids and the optrA gene in the CQ20-MVs. MV-mediated transformation to E. faecalis SC032 and E. faecium BM4105 was successfully performed, and the WGS data also showed that both plasmids pCQ20-1 and pCQ20-2 and optrA-carrying transposon Tn6674 were transferred to E. faecalis SC032 and E. faecium BM4105, but failed for E. coli. Additionally, vesiculants that had acquired ARGs still had the ability to spread these genes via MVs. CONCLUSIONS To our knowledge, this is the first report of MV-mediated co-transfer of ARG-carrying plasmids and transposons in the Gram-positive bacterium E. faecium.
Collapse
Affiliation(s)
- Mengyu Zhao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, China
| | - Shuang He
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, China
| | - Renqiao Wen
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, China
| | - Chao Li
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, China
| | - Xinggui Chen
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, China
| | - Xiaolong Lin
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, China
| | - Hongning Wang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, China
| | - Yizhi Tang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Lehmkuhl J, Schneider JS, Werth KLV, Scherff N, Mellmann A, Kampmeier S. Role of membrane vesicles in the transmission of vancomycin resistance in Enterococcus faecium. Sci Rep 2024; 14:1895. [PMID: 38253607 PMCID: PMC10803344 DOI: 10.1038/s41598-024-52310-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/17/2024] [Indexed: 01/24/2024] Open
Abstract
Clonal transmission and horizontal gene transfer (HGT) contribute to the spread of vancomycin-resistant enterococci (VRE) in global healthcare. Our study investigated vesiduction, a HGT mechanism via membrane vesicles (MVs), for vanA and vanB genes that determine vancomycin resistance. We isolated MVs for VRE of different sequence types (STs) and analysed them by nanoparticle tracking analysis. Selected MV samples were subjected to DNA sequence analysis. In resistance transfer experiments, vancomycin-susceptible enterococci were exposed to MVs and bacterial supernatants of VRE. Compared to bacteria grown in lysogeny broth (MVs/LB), cultivation under vancomycin stress (MVs/VAN) resulted in increased particle concentrations of up to 139-fold (ST80). As a key finding, we could show that VRE isolates of ST80 and ST117 produced remarkably more vesicles at subinhibitory antibiotic concentrations (approx. 9.2 × 1011 particles/ml for ST80 and 2.4 × 1011 particles/ml for ST117) than enterococci of other STs (range between 1.8 × 1010 and 5.3 × 1010 particles/ml). In those MV samples, the respective resistance genes vanA and vanB were completely verifiable using sequence analysis. Nevertheless, no vancomycin resistance transfer via MVs to vancomycin-susceptible Enterococcus faecium was phenotypically detectable. However, our results outline the potential of future research on ST-specific MV properties, promising new insights into VRE mechanisms.
Collapse
Affiliation(s)
- Johanna Lehmkuhl
- Institute of Hygiene, University Hospital Münster, 48149, Münster, Germany
| | | | | | - Natalie Scherff
- Institute of Hygiene, University Hospital Münster, 48149, Münster, Germany
| | - Alexander Mellmann
- Institute of Hygiene, University Hospital Münster, 48149, Münster, Germany
| | - Stefanie Kampmeier
- Institute of Hygiene, University Hospital Münster, 48149, Münster, Germany.
- Institute for Hygiene and Microbiology, University of Würzburg, 97080, Würzburg, Germany.
| |
Collapse
|
20
|
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: 38] [Impact Index Per Article: 19.0] [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
|
21
|
Fan F, Wang J, Chen H, Wei L, Zhang Z. Isolation and protein MdtQ analysis of outer membrane vesicles released by carbapenem-resistant Klebsiella pneumoniae. Microb Pathog 2023; 183:106325. [PMID: 37640276 DOI: 10.1016/j.micpath.2023.106325] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CRKP) has emerged as a leading public health problem, and is increasingly being reported worldwide with resistance to a wide spectrum of antibiotics. Recent reports have demonstrated that the outer membrane vesicles (OMVs) of gram-negative bacteria are potent resistance factors, but their role in the drug resistance of CRKP has not been elucidated. In order to investigate the effects of OMV components on drug resistance and to explore the mechanism of antimicrobial resistance in CRKP, we isolated the OMVs through ultracentrifugation, separated the OMV proteins through mass spectrometry (MS), and performed bioinformatics analysis. A total of 3,192 proteins were detected by nano LC-MS/MS analysis, with 108 (61.4%) cytoplasmic proteins, 50 (28.4%) cytoplasmic membrane proteins, nine (5.1%) periplasmic proteins, six (3.4%) outer membrane proteins, two (1.1%) extracellular proteins, and one (0.6%) other protein detected in the vesicles. MdtQ was detected as the only multidrug resistance outer membrane protein. Further experiments confirmed that MdtQ included the 1440 BP sequence and had a unique three-dimensional structure. To superimpose MdtQ with KPC-2 resistant proteins, I7ACB1, I7AKP2, and Q93LQ9, the root mean square deviation (RMSD) values were calculated (0.379, 0.671, and 1.35, respectively). I7ACB1 had the lowest RMSD value, indicating that it had the best superimposition effect. Furthermore, MdtQ had 20 biological pocket structures, and the four most important pockets were evenly distributed around the inner perimeter of its three-dimensional structure. These findings may provide a theoretical basis for controlling the spread of bacterial resistance in the future.
Collapse
Affiliation(s)
- Fangfang Fan
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, 233030, China
| | - Jiaqi Wang
- Department of Clinical Laboratory, Shanghai Clinical Medical College of Qingdao University, Shanghai Deji Hospital, Shanghai, 200331, China
| | - Hong Chen
- Department of Clinical Laboratory, Shanghai Clinical Medical College of Qingdao University, Shanghai Deji Hospital, Shanghai, 200331, China
| | - Li Wei
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, 233030, China.
| | - Zhen Zhang
- Department of Clinical Laboratory, Shanghai Clinical Medical College of Qingdao University, Shanghai Deji Hospital, Shanghai, 200331, China.
| |
Collapse
|
22
|
Zhang X, Wang Y, Fan R, Zhang L, Li Z, Zhang Y, Zheng W, Wang L, Liu B, Quan C. Quantitative Proteomic Analysis of Outer Membrane Vesicles from Fusobacterium nucleatum Cultivated in the Mimic Cancer Environment. Microbiol Spectr 2023; 11:e0039423. [PMID: 37341631 PMCID: PMC10434195 DOI: 10.1128/spectrum.00394-23] [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: 01/29/2023] [Accepted: 05/25/2023] [Indexed: 06/22/2023] Open
Abstract
Fusobacterium nucleatum is a Gram-negative bacterium that has been identified as an important pathogenic gut bacterium associated with colorectal cancer. Compared with the normal intestine, the pH value of the tumor microenvironment is weakly acidic. The metabolic changes of F. nucleatum in the tumor microenvironment, especially the protein composition of its outer membrane vesicles, remain unclear. Here, we systematically analyzed the effect of environmental pH on the proteome of outer membrane vesicles (OMVs) from F. nucleatum by tandem mass tag (TMT) labeling-high-resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. A total of 991 proteins were identified in acidic OMVs (aOMVs) and neutral OMVs (nOMVs), including known virulence proteins and putative virulence proteins. Finally, 306 upregulated proteins and 360 downregulated proteins were detected in aOMVs, and approximately 70% of the expression of OMV proteins was altered under acidic conditions. A total of 29 autotransporters were identified in F. nucleatum OMVs, and 13 autotransporters were upregulated in aOMVs. Interestingly, three upregulated autotransporters (D5REI9, D5RD69, and D5RBW2) show homology to the known virulence factor Fap2, suggesting that they may be involved in various pathogenic pathways such as the pathway for binding with colorectal cancer cells. Moreover, we found that more than 70% of MORN2 domain-containing proteins may have toxic effects on host cells. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses demonstrated that a number of proteins were significantly enriched in multiple pathways involving fatty acid synthesis and butyrate synthesis. Seven metabolic enzymes involved in fatty acid metabolism pathways were identified in the proteomic data, of which 5 were upregulated and 2 were downregulated in aOMVs, while 14 metabolic enzymes involved in the butyric acid metabolic pathway were downregulated in aOMVs. In conclusion, we found a key difference in virulence proteins and pathways in the outer membrane vesicles of F. nucleatum between the tumor microenvironment pH and normal intestinal pH, which provides new clues for the prevention and treatment of colorectal cancer. IMPORTANCE F. nucleatum is an opportunistic pathogenic bacterium that can be enriched in colorectal cancer tissues, affecting multiple stages of colorectal cancer development. OMVs have been demonstrated to play key roles in pathogenesis by delivering toxins and other virulence factors to host cells. By employing quantitative proteomic analysis, we found that the pH conditions could affect the protein expression of the outer membrane vesicles of F. nucleatum. Under acidic conditions, approximately 70% of the expression of proteins in OMVs was altered. Several virulence factors, such as type 5a secreted autotransporter (T5aSSs) and membrane occupation and recognition nexus (MORN) domain-containing proteins, were upregulated under acidic conditions. A large number of proteins showed significant enrichments in multiple pathways involving fatty acid synthesis and butyrate synthesis. Proteomics analysis of the outer membrane vesicles secreted by pathogenic bacteria in the acidic tumor microenvironment is of great significance for elucidating the pathogenicity mechanism and its application in vaccine and drug delivery vehicles.
Collapse
Affiliation(s)
- Xuqiang Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Yuxin Wang
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Ruochen Fan
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, Liaoning, China
| | - Liying Zhang
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, Liaoning, China
| | - Zhuting Li
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Yanmei Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Wei Zheng
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Lulu Wang
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, Liaoning, China
| | - Baoquan Liu
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Chunshan Quan
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| |
Collapse
|
23
|
Silva E Carvalho I, Pratavieira S, Salvador Bagnato V, Alves F. Sonophotodynamic inactivation of Pseudomonas aeruginosa biofilm mediated by curcumin. BIOFOULING 2023; 39:606-616. [PMID: 37537876 DOI: 10.1080/08927014.2023.2241385] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/20/2023] [Accepted: 07/23/2023] [Indexed: 08/05/2023]
Abstract
The inactivation of Pseudomonas aeruginosa biofilm is a major challenge, as biofilms are less responsive to conventional treatments and responsible for persistent infections. This has led to the investigation of alternative approaches for biofilm control such as photodynamic (PDI) and sonodynamic (SDI) inactivation. The combination of them, known as Sonophotodynamic Inactivation (SPDI), has improved the effectiveness of the process. Curcumin, a well-established photosensitizer, has been identified as a potential sonosensitizer. This study evaluated the most effective combination for SPDI against P. aeruginosa biofilms in vitro, varying curcumin concentrations and ultrasound intensities. The results indicated that the inactivation was directly proportional to the curcumin concentration. Using curcumin 120 µM and 3.0 W.cm-2 of ultrasound intensity, SPDI demonstrated the highest and the best synergistic results, equivalent to 6.9 ± 2.1 logs of reduction. PDI reduced 0.7 ± 0.9 log and SDI had no effect. In conclusion, SPDI with curcumin is a promising approach for biofilm inactivation.
Collapse
Affiliation(s)
| | | | - Vanderlei Salvador Bagnato
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
- Department of Biomedical Engineering, College of Engineering, TX A&M University, College Station, TX, USA
| | - Fernanda Alves
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| |
Collapse
|
24
|
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
|
25
|
Moura de Sousa J, Lourenço M, Gordo I. Horizontal gene transfer among host-associated microbes. Cell Host Microbe 2023; 31:513-527. [PMID: 37054673 DOI: 10.1016/j.chom.2023.03.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Horizontal gene transfer is an important evolutionary force, facilitating bacterial diversity. It is thought to be pervasive in host-associated microbiomes, where bacterial densities are high and mobile elements are frequent. These genetic exchanges are also key for the rapid dissemination of antibiotic resistance. Here, we review recent studies that have greatly extended our knowledge of the mechanisms underlying horizontal gene transfer, the ecological complexities of a network of interactions involving bacteria and their mobile elements, and the effect of host physiology on the rates of genetic exchanges. Furthermore, we discuss other, fundamental challenges in detecting and quantifying genetic exchanges in vivo, and how studies have contributed to start overcoming these challenges. We highlight the importance of integrating novel computational approaches and theoretical models with experimental methods where multiple strains and transfer elements are studied, both in vivo and in controlled conditions that mimic the intricacies of host-associated environments.
Collapse
Affiliation(s)
- Jorge Moura de Sousa
- Institut Pasteur, Université Paris Cité, CNRS, UMR3525, Microbial Evolutionary Genomics, Paris, 75015 Paris, France
| | - Marta Lourenço
- Institut Pasteur, Université Paris Cité, Biodiversity and Epidemiology of Bacterial Pathogens, F-75015 Paris, France
| | - Isabel Gordo
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande,6, Oeiras, Portugal.
| |
Collapse
|
26
|
Hussein M, Jasim R, Gocol H, Baker M, Thombare VJ, Ziogas J, Purohit A, Rao GG, Li J, Velkov T. Comparative Proteomics of Outer Membrane Vesicles from Polymyxin-Susceptible and Extremely Drug-Resistant Klebsiella pneumoniae. mSphere 2023; 8:e0053722. [PMID: 36622250 PMCID: PMC9942579 DOI: 10.1128/msphere.00537-22] [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: 10/29/2022] [Accepted: 12/06/2022] [Indexed: 01/10/2023] Open
Abstract
Outer membrane vesicles (OMVs) secreted by Gram-negative bacteria serve as transporters for the delivery of cargo such as virulence and antibiotic resistance factors. OMVs play a key role in the defense against membrane-targeting antibiotics such as the polymyxin B. Herein, we conducted comparative proteomics of OMVs from paired Klebsiella pneumoniae ATCC 700721 polymyxin-susceptible (polymyxin B MIC = 0.5 mg/L) and an extremely resistant (polymyxin B MIC ≥128 mg/L), following exposure to 2 mg/L of polymyxin B. Comparative profiling of the OMV subproteome of each strain revealed proteins from multiple perturbed pathways, particularly in the polymyxin-susceptible strain, including outer membrane assembly (lipopolysaccharide, O-antigen, and peptidoglycan biosynthesis), cationic antimicrobial peptide resistance, β-lactam resistance, and quorum sensing. In the polymyxin-susceptible strain, polymyxin B treatment reduced the expression of OMV proteins in the pathways related to adhesion, virulence, and the cell envelope stress responses, whereas, in the polymyxin-resistant strain, the proteins involved in LPS biosynthesis, RNA degradation, and nucleotide excision repair were significantly overexpressed in response to polymyxin B treatment. Intriguingly, the key polymyxin resistance enzymes 4-amino-4-deoxy-l-arabinose transferase and the PhoPQ two-component protein kinase were significantly downregulated in the OMVs of the polymyxin-susceptible strain. Additionally, a significant reduction in class A β-lactamase proteins was observed following polymyxin B treatment in the OMVs of both strains, particularly the OMVs of the polymyxin-susceptible strain. These findings shed new light on the OMV subproteome of extremely polymyxin resistant K. pneumoniae, which putatively may serve as active decoys to make the outer membrane more impervious to polymyxin attack. IMPORTANCE OMVs can help bacteria to fight antibiotics not only by spreading antibiotic resistance genes but also by acting as protective armor against antibiotics. By employing proteomics, we found that OMVs have a potential role in shielding K. pneumoniae and acting as decoys to polymyxin attack, through declining the export of proteins (e.g., 4-amino-4-deoxy-l-arabinose transferase) involved in polymyxin resistance. Furthermore, polymyxin B treatment of both strains leads to shedding of the OMVs with perturbed proteins involved in outer membrane remodeling (e.g., LPS biosynthesis) as well as pathogenic potential of K. pneumoniae (e.g., quorum sensing). The problematic extended spectrum beta-lactamases SHV and TEM were significantly reduced in both strains, suggesting that polymyxin B may act as a potentiator to sensitize the bacterium to β-lactam antibiotics. This study highlights the importance of OMVs as "molecular mules" for the intercellular transmission and delivery of resistance and cellular repair factors in the bacterial response to polymyxins.
Collapse
Affiliation(s)
- Maytham Hussein
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Raad Jasim
- Department of Pharmacology, College of Pharmacy, University of Babylon, Iraq
| | - Hakan Gocol
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Mark Baker
- Discipline of Biological Sciences, Priority Research Centre in Reproductive Biology, Faculty of Science and IT, University of Newcastle, Callaghan, New South Wales, Australia
| | - Varsha J. Thombare
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - James Ziogas
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Aayush Purohit
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Gauri G. Rao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Tony Velkov
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
27
|
Li S, Wu J, Ma N, Liu W, Shao M, Ying N, Zhu L. Prediction of genome-wide imipenem resistance features in Klebsiella pneumoniae using machine learning. J Med Microbiol 2023; 72. [PMID: 36753438 DOI: 10.1099/jmm.0.001657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Introduction. The resistance rate of Klebsiella pneumoniae (K. pneumoniae) to imipenem is increasing year by year, and the imipenem resistance mechanism of K. pneumoniae is complex. Therefore, it is urgent to develop new strategies to explore the resistance mechanism of imipenem for its effective and accurate use in clinical practice.Hypothesis/Gap sStatement. Machine learning could identify resistance features and biological process that influence microbial resistance from whole-genome sequencing (WGS) data.Aims. This work aimed to predict imipenem resistance genetic features in K. pneumoniae from whole-genome k-mer features, and analyse their function for understanding its resistance mechanism.Methods. This study analysed WGS data of K. pneumoniae combined with resistance phenotype for imipenem, and established K. pneumoniae to imipenem genotype-phenotype model to predict resistance features using chi-squared test and random forest. An external clinical dataset was used to verify prediction power of resistance features. The potential genes were identified through alignment the resistance features with the K. pneumoniae reference genome using blastn, the functions of potential genes were further analysed to explore its resistance-related signalling pathways with GO and KEGG analysis, the resistance sequence patterns were screened using streme software. Finally, the resistance features were combined and modelled through four machine-learning algorithms (logistic regression, SVM, GBDT and XGBoost) to evaluate their phenotype prediction ability.Results. A total of 16 670 imipenem resistance features were predicted from genotype-phenotype model. The 30 potential genes were identified by annotating the resistance features and corresponded to known antibiotic-related genes (mdtM, dedA, rne, etc.). GO and KEGG pathway analyses indicated the possible association of imipenem resistance with metabolism process and cell membrane. CRYCAGCDN and CGRDAAAN were found from the imipenem resistance features, which were widely presented in the reported β-lactam resistance genes (bla SHV, bla CTX-M, bla TEM, etc.), and YCYAGCMCAST with metabolic functions (organic substance metabolic process, nitrogen compound metabolic process and cellular metabolic process) was identified from the top 50 resistance features. The 25 resistance genes in the training dataset included 19 genes in the external dataset, which verified the accuracy of prediction. The area under curve values of logistics regression, SVM, GBDT and XGBoost were 0.965, 0.966, 0.969 and 0.969, respectively, indicating that the imipenem resistance features have a strong prediction power.Conclusion. Machine-learning methods could effectively predict the imipenem resistance feature in K. pneumoniae, and provide resistance sequence profiles for predicting resistance phenotype and exploring potential resistance mechanisms. It provides an important insight into the potential therapeutic strategies of K. pneumoniae resistance to imipenem, and speed up the application of machine learning in routine diagnosis.
Collapse
Affiliation(s)
- Shanshan Li
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| | - Jun Wu
- Lin'an Center for Disease Control and Prevention, Lin'an, 311300, PR China
| | - Nan Ma
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| | - Wenjia Liu
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China.,College of Electronics and Information Engineering, Hangzhou Dianzi University, Hangzhou 310018, PR China
| | - Mengjie Shao
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| | - Nanjiao Ying
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China.,Institute of Biomedical Engineering and Instrument, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| | - Lei Zhu
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China.,Institute of Biomedical Engineering and Instrument, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| |
Collapse
|
28
|
MacNair CR, Tan MW. The role of bacterial membrane vesicles in antibiotic resistance. Ann N Y Acad Sci 2023; 1519:63-73. [PMID: 36415037 DOI: 10.1111/nyas.14932] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Bacterial survival during antibiotic exposure is a complex and multifaceted phenomenon. On top of antibiotic resistance genes, biofilm formation, and persister tolerance, bacterial membrane vesicles (MVs) provide a layer of protection that has been largely overlooked. MVs are spherical nanoparticles composed of lipid membranes and are common to Gram-positive and Gram-negative bacteria. Although the importance of MVs in bacterial pathogenesis and virulence factor transport has been firmly established, a growing body of work now identifies MVs as key contributors to bacterial survival during antibiotic exposure. Herein, we highlight the ability of MVs to reduce antibiotic efficacy and transmit resistance elements. We also discuss the potential of targeting MV production as an unconventional therapeutic approach.
Collapse
Affiliation(s)
- Craig R MacNair
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, California, USA
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, California, USA
| |
Collapse
|
29
|
Li P, Luo W, Xiang TX, Jiang Y, Liu P, Wei DD, Fan L, Huang S, Liao W, Liu Y, Zhang W. Horizontal gene transfer via OMVs co-carrying virulence and antimicrobial-resistant genes is a novel way for the dissemination of carbapenem-resistant hypervirulent Klebsiella pneumoniae. Front Microbiol 2022; 13:945972. [PMID: 36532464 PMCID: PMC9751880 DOI: 10.3389/fmicb.2022.945972] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/19/2022] [Indexed: 08/15/2023] Open
Abstract
INTRODUCTION The rapidly increased isolation rate of CR-HvKP worldwide has brought great difficulties in controlling clinical infection. Moreover, it has been demonstrated that the transmission of drug-resistant genes among bacteria can be mediated by outer membrane vesicles (OMVs), which is a new way of horizontal gene transfer (HGT). The transmission of virulence genes among bacteria has also been well studied; however, it remains unclear whether virulence and drug-resistant genes can be co-transmitted simultaneously. Co-transmission of virulence and drug-resistant genes is essential for the formation and prevalence of CR-HvKP. METHODS First, we isolated OMVs from CR-HvKP by cushioned-density gradient ultracentrifugation (C-DGUC). TEM and DLS were used to examine the morphology and size of bacterial OMVs. OMV-mediated gene transfer in liquid cultures and the acquisition of the carbapenem gene and virulence gene was confirmed using colony-PCR. Antimicrobial susceptibility testing, mCIM and eCIM were conducted for the resistance of transformant. Serum killing assay, assessment of the anti-biofilm effect and galleria mellonella infection model, mucoviscosity assay, extraction and quantification of capsules were verified the virulence of transformant. Pulsed-field gel electrophoresis (PFGE), S1 nuclease-pulsed-field gel electrophoresis (S1-PFGE), Southern blotting hybridization confirmed the plasmid of transformant. RESULTS Firstly, OMVs were isolated from CR-HvKP NUHL30457 (K2, ST86). TEM and DLS analyses revealed the spherical morphology of the vesicles. Secondly, our study demonstrated that CR-HvKP delivered genetic material, incorporated DNA within the OMVs, and protected it from degradation by extracellular exonucleases. Thirdly, the vesicular lumen DNA was delivered to the recipient cells after determining the presence of virulence and carbapenem-resistant genes in the CR-HvKP OMVs. Importantly, S1-PFGE and Southern hybridization analysis of the 700603 transformant strain showed that the transformant contained both drug-resistant and virulence plasmids. DISCUSSION In the present study, we aimed to clarify the role of CRHvKP-OMVs in transmitting CR-HvKP among K. pneumoniae. Collectively, our findings provided valuable insights into the evolution of CR-HvKP.
Collapse
Affiliation(s)
- Ping Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Yichun People's Hospital, Yichun, China
| | - Wanying Luo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tian-Xin Xiang
- Department of Infectious Diseases, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Yuhuan Jiang
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Peng Liu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Dan-Dan Wei
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Linping Fan
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Shanshan Huang
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Wenjian Liao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yang Liu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
- National Regional Center for Respiratory Medicine, China-Japan Friendship Jiangxi Hospital, Nanchang, China
| | - Wei Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
30
|
Hua Y, Wang J, Huang M, Huang Y, Zhang R, Bu F, Yang B, Chen J, Lin X, Hu X, Zheng L, Wang Q. Outer membrane vesicles-transmitted virulence genes mediate the emergence of new antimicrobial-resistant hypervirulent Klebsiella pneumoniae. Emerg Microbes Infect 2022; 11:1281-1292. [PMID: 35437096 PMCID: PMC9132476 DOI: 10.1080/22221751.2022.2065935] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Hypervirulent Klebsiella pneumoniae (hvKp) is a notorious clinical pathogen that is more likely to cause severe primary and metastatic abscesses. The dissemination of antimicrobial-resistant hvKp isolates has been reported worldwide, posing a great challenge and severe clinical threat. However, the mechanisms of antimicrobial-resistant hvKp isolates prevalent worldwide are not well precise. Outer membrane vesicles (OMVs) secreted from gram-negative bacteria are an important vehicle for delivering effector molecules inter- and intra-species. To explore whether OMVs as the vector of virulence genes horizontal transfer among Klebsiella pneumoniae and to explain the potential mechanism for the development of antimicrobial-resistant hvKp isolates, we isolated OMVs from hvKp and classical Klebsiella pneumoniae (cKp) by sequential differential centrifugation, respectively. Then, the characteristics and contents of hvKp-OMVs and cKp-OMVs were analyzed. These hvKp-OMVs contain virulence genes, which could be transferred from hvKp horizontally to extended-spectrum beta lactamase (ESBL)-producing cKp, leading to the production of antimicrobial-resistant hypervirulent transformants. Further experiments confirmed the transformants exhibited antimicrobial resistance and hypervirulent phenotypes in vitro and in vivo. In short, this work demonstrated that hvKp-OMVs facilitated virulence genes transfer, allowing an increase in the virulence level of ESBL-producing cKp and providing a new mechanism for the emergence of antimicrobial-resistant hvKp isolates.
Collapse
Affiliation(s)
- Yuneng Hua
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Mei Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yiyi Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ruyi Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Fan Bu
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Biao Yang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Juanjiang Chen
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaomin Lin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiumei Hu
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Qian Wang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
31
|
The Discovery of the Role of Outer Membrane Vesicles against Bacteria. Biomedicines 2022; 10:biomedicines10102399. [PMID: 36289660 PMCID: PMC9598313 DOI: 10.3390/biomedicines10102399] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/02/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
Gram-negative bacteria are intrinsically resistant to many commercialized antibiotics. The outer membrane (OM) of Gram-negative bacteria prevents the entry of such antibiotics. Outer membrane vesicles (OMV) are naturally released from the OM of Gram-negative bacteria for a range of purposes, including competition with other bacteria. OMV may carry, as part of the membrane or lumen, molecules with antibacterial activity. Such OMV can be exposed to and can fuse with the cell surface of different bacterial species. In this review we consider how OMV can be used as tools to deliver antimicrobial agents. This includes the characteristics of OMV production and how this process can be used to create the desired antibacterial activity of OMV.
Collapse
|
32
|
In Vitro Antibacterial and Anti-Inflammatory Activity of Arctostaphylos uva-ursi Leaf Extract against Cutibacterium acnes. Pharmaceutics 2022; 14:pharmaceutics14091952. [PMID: 36145700 PMCID: PMC9501556 DOI: 10.3390/pharmaceutics14091952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/31/2022] [Accepted: 09/09/2022] [Indexed: 12/03/2022] Open
Abstract
Cutibacterium acnes (C. acnes) is the main causative agent of acne vulgaris. The study aims to evaluate the antimicrobial activity of a natural product, Arctostaphylos uva-ursi leaf extract, against C. acnes. Preliminary chemical–physical characterization of the extract was carried out by means of FT-IR, TGA and XPS analyses. Skin permeation kinetics of the extract conveyed by a toning lotion was studied in vitro by Franz diffusion cell, monitoring the permeated arbutin (as the target component of the extract) and the total phenols by HPLC and UV-visible spectrophotometry, respectively. Antimicrobial activity and time-killing assays were performed to evaluate the effects of Arctostaphylos uva-ursi leaf extract against planktonic C. acnes. The influence of different Arctostaphylos uva-ursi leaf extract concentrations on the biofilm biomass inhibition and degradation was evaluated by the crystal violet (CV) method. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) test was used to determine the viability of immortalized human keratinocytes (HaCaT) after exposure to Arctostaphylos uva-ursi leaf extract for 24 and 48 h. Levels of interleukin (IL)-1β, IL-6, IL-8 and tumour necrosis factor (TNF)-α were quantified after HaCaT cells cotreatment with Arctostaphylos uva-ursi leaf extract and heat-killed C. acnes. The minimum inhibitory concentration (MIC) which exerted a bacteriostatic action on 90% of planktonic C. acnes (MIC90) was 0.6 mg/mL. Furthermore, MIC and sub-MIC concentrations influenced the biofilm formation phases, recording a percentage of inhibition that exceeded 50 and 40% at 0.6 and 0.3 mg/mL. Arctostaphylos uva-ursi leaf extract disrupted biofilm biomass of 57 and 45% at the same concentrations mentioned above. Active Arctostaphylos uva-ursi leaf extract doses did not affect the viability of HaCaT cells. On the other hand, at 1.25 and 0.6 mg/mL, complete inhibition of the secretion of pro-inflammatory cytokines was recorded. Taken together, these results indicate that Arctostaphylos uva-ursi leaf extract could represent a natural product to counter the virulence of C. acnes, representing a new alternative therapeutic option for the treatment of acne vulgaris.
Collapse
|
33
|
Characteristics of Antibiotic Resistance and Tolerance of Environmentally Endemic Pseudomonas aeruginosa. Antibiotics (Basel) 2022; 11:antibiotics11081120. [PMID: 36009989 PMCID: PMC9404893 DOI: 10.3390/antibiotics11081120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Antibiotic-resistant bacteria remain a serious public health threat. In order to determine the percentage of antibiotic-resistant and -tolerant Pseudomonas aeruginosa cells present and to provide a more detailed infection risk of bacteria present in the environment, an isolation method using a combination of 41 °C culture and specific primers was established to evaluate P. aeruginosa in the environment. The 50 strains were randomly selected among 110 isolated from the river. The results of antibiotic susceptibility evaluation showed that only 4% of environmental strains were classified as antibiotic-resistant, while 35.7% of clinical strains isolated in the same area were antibiotic-resistant, indicating a clear difference between environmental and clinical strains. However, the percentage of antibiotic-tolerance, an indicator of potential resistance risk for strains that have not become resistant, was 78.8% for clinical strains and 90% for environmental strains, suggesting that P. aeruginosa, a known cause of nosocomial infections, has a high rate of antibiotic-tolerance even in environmentally derived strains. It suggested that the rate of antibiotic-tolerance is not elicited by the presence or absence of antimicrobial exposure. The combination of established isolation and risk analysis methods presented in this study should provide accurate and efficient information on the risk level of P. aeruginosa in various regions and samples.
Collapse
|
34
|
Impact of Escherichia coli Outer Membrane Vesicles on Sperm Function. Pathogens 2022; 11:pathogens11070782. [PMID: 35890027 PMCID: PMC9319964 DOI: 10.3390/pathogens11070782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 01/27/2023] Open
Abstract
Reproductive tract infections account for approximately 15% of male infertility cases. Escherichia coli (E. coli) represents the most frequently isolated bacterial strain in the semen of infertile men. All Gram-negative bacteria constitutively produce outer membrane vesicles (OMVs). The present study proved, for the first time, the involvement of OMVs in human sperm function. E. coli OMVs were isolated by ultracentrifugation and characterized via sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE), transmission electron microscopy (TEM) and dynamic light scattering (DLS) analysis. Human sperm was exposed to OMVs (8 µg/mL) for different times (30, 45, 60 and 90 min). The vitality, motility, morphology, ROS level and DNA fragmentation of spermatozoa were evaluated. OMVs reduced the progressive motility and increased the immobile spermatozoa amount after 30 min of treatment. In addition, a significant increase in the percentage of intracellular ROS and sperm DNA fragmentation was recorded for each vesicular exposure time. These preliminary findings prove that OMVs contribute to altering human sperm function via two mechanisms: (i) impaired motility and (ii) DNA fragmentation.
Collapse
|
35
|
Petrillo F, Petrillo A, Marrapodi M, Capristo C, Gicchino MF, Montaldo P, Caredda E, Reibaldi M, Boatti LMV, Dell’Annunziata F, Folliero V, Galdiero M. Characterization and Comparison of Ocular Surface Microbiome in Newborns. Microorganisms 2022; 10:microorganisms10071390. [PMID: 35889110 PMCID: PMC9320102 DOI: 10.3390/microorganisms10071390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 12/10/2022] Open
Abstract
The ocular microbiome is of fundamental importance for immune eye homeostasis, and its alteration would lead to an impairment of ocular functionality. Little evidence is reported on the composition of the ocular microbiota of term infants and on the impact of antibiotic prophylaxis. Methods: A total of 20 conjunctival swabs were collected from newborns at birth and after antibiotic treatment. Samples were subjected to 16S rRNA sequencing via system MiSeq Illumina. The data were processed with the MicrobAT software and statistical analysis were performed using two-way ANOVA. Results: Antibiotic prophylaxis with gentamicin altered the composition of the microbiota. In detail, a 1.5- and 2.01-fold reduction was recorded for Cutibacterium acnes (C. acnes) and Massilia timonae (M. timonae), respectively, whereas an increase in Staphylococcus spp. of 6.5 times occurred after antibiotic exposure. Conclusions: Antibiotic prophylaxis altered the ocular microbiota whose understanding could avoid adverse effects on eye health.
Collapse
Affiliation(s)
- Francesco Petrillo
- Department of Surgical Sciences, Eye Clinic Section, University of Turin, 10124 Turin, Italy; (F.P.); (M.R.)
| | - Arianna Petrillo
- Pediatric Unit, Fondazione IRCCS “Ca’ Granda-Ospedale Maggiore-Policlinico”, 20122 Milan, Italy;
| | - Maddalena Marrapodi
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (M.M.); (M.F.G.)
| | - Carlo Capristo
- Department of Neonatology, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.C.); (P.M.); (E.C.)
| | - Maria Francesca Gicchino
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (M.M.); (M.F.G.)
| | - Paolo Montaldo
- Department of Neonatology, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.C.); (P.M.); (E.C.)
| | - Elisabetta Caredda
- Department of Neonatology, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.C.); (P.M.); (E.C.)
| | - Michele Reibaldi
- Department of Surgical Sciences, Eye Clinic Section, University of Turin, 10124 Turin, Italy; (F.P.); (M.R.)
| | | | - Federica Dell’Annunziata
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.D.); (V.F.)
| | - Veronica Folliero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.D.); (V.F.)
| | - Marilena Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.D.); (V.F.)
- Correspondence:
| |
Collapse
|
36
|
Wang Z, Wen Z, Jiang M, Xia F, Wang M, Zhuge X, Dai J. Dissemination of virulence and resistance genes among Klebsiella pneumoniae via outer membrane vesicle: An important plasmid transfer mechanism to promote the emergence of carbapenem-resistant hypervirulent Klebsiella pneumoniae. Transbound Emerg Dis 2022; 69:e2661-e2676. [PMID: 35679514 DOI: 10.1111/tbed.14615] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/15/2022] [Accepted: 06/07/2022] [Indexed: 12/01/2022]
Abstract
Klebsiella pneumoniae is well-known opportunistic enterobacteria involved in complex clinical infections in humans and animals. The domestic animals might be a source of the multidrug-resistant virulent K. pneumoniae to humans. K. pneumoniae infections in domestic animals are considered as an emergent global concern. The horizontal gene transfer plays essential roles in bacterial genome evolution by spread of virulence and resistance determinants. However, the virulence genes can be transferred horizontally via K. pneumoniae-derived outer membrane vesicles (OMVs) remains to be unreported. In this study, we performed complete genome sequencing of two K. pneumoniae HvK2115 and CRK3022 with hypervirulent or carbapenem-resistant traits. OMVs from K. pneumoniae HvK2115 and CRK3022 were purified and observed. The carriage of virulence or resistance genes in K. pneumoniae OMVs was identified. The influence of OMVs on the horizontal transfer of virulence-related or drug-resistant plasmids among K. pneumoniae strains was evaluated thoroughly. The plasmid transfer to recipient bacteria through OMVs was identified by polymerase chain reaction, pulsed field gel electrophoresis and Southern blot. This study revealed that OMVs could mediate the intraspecific and interspecific horizontal transfer of the virulence plasmid phvK2115. OMVs could simultaneously transfer two resistance plasmids into K. pneumoniae and Escherichia coli recipient strains. OMVs-mediated horizontal transfer of virulence plasmid phvK2115 could significantly enhance the pathogenicity of human carbapenem-resistant K. pneumoniae CRK3022. The CRK3022 acquired the virulence plasmid phvK2115 could become a CR-hvKp strain. It was critically important that OMVs-mediated horizontal transfer of phvK2115 lead to the coexistence of virulence and carbapenem-resistance genes in K. pneumoniae, resulting in the emerging of carbapenem-resistant hypervirulent K. pneumoniae.
Collapse
Affiliation(s)
- Zhongxing Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhe Wen
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Min Jiang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Fufang Xia
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Min Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Xiangkai Zhuge
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jianjun Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,College of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
37
|
Folliero V, Dell’Annunziata F, Roscetto E, Amato A, Gasparro R, Zannella C, Casolaro V, De Filippis A, Catania MR, Franci G, Galdiero M. Rhein: A Novel Antibacterial Compound Against Streptococcus mutans Infection. Microbiol Res 2022; 261:127062. [DOI: 10.1016/j.micres.2022.127062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 12/15/2022]
|
38
|
Li C, Wen R, Mu R, Chen X, Ma P, Gu K, Huang Z, Ju Z, Lei C, Tang Y, Wang H. Outer Membrane Vesicles of Avian PathogenicEscherichia coli Mediate the Horizontal Transmission of blaCTX-M-55. Pathogens 2022; 11:pathogens11040481. [PMID: 35456156 PMCID: PMC9025603 DOI: 10.3390/pathogens11040481] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/11/2022] [Accepted: 04/16/2022] [Indexed: 12/21/2022] Open
Abstract
The CTX-M-55 type extended-spectrum β-lactamase (ESBL) producing Enterobacteriaceae is increasing in prevalence worldwide without the transmission mechanism being fully clarified, which threatens public and livestock health. Outer membrane vesicles (OMVs) have been shown to mediate the gene horizontal transmission in some species. However, whether blaCTX-M-55 can be transmitted horizontally through OMVs in avian pathogenic Escherichia coli (APEC) has not been reported yet. To test this hypothesis, an ESBL-producing APEC was isolated and whole-genome sequencing (WGS) was performed to analyze the location of blaCTX-M-55. Ultracentrifugation and size exclusion chromatography was used to isolate and purify OMVs, and the transfer experiment of blaCTX-M-55 via OMVs was performed finally. Our results showed that the blaCTX-M-55 was located on an IncI2 plasmid. The number and diameter of OMVs secreted by ESBL-producing APEC treated with different antibiotics were significantly varied. The transfer experiment showed that the OMVs could mediate the horizontal transfer of blaCTX-M-55, and the frequency of gene transfer ranged from 10−5 to 10−6 CFU/mL with the highest frequency observed in the Enrofloxacin treatment group. These findings contribute to a better understanding of the antibiotics in promoting and disseminating resistance in the poultry industry and support the restrictions on the use of antibiotics in the poultry industry.
Collapse
Affiliation(s)
- Chao Li
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Renqiao Wen
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Rongrong Mu
- Provincial Key Laboratory for Transfusion-Transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, China;
| | - Xuan Chen
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Peng Ma
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Kui Gu
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Zheren Huang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Zijing Ju
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Changwei Lei
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Yizhi Tang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
| | - Hongning Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China; (C.L.); (R.W.); (X.C.); (P.M.); (K.G.); (Z.H.); (Z.J.); (C.L.); (Y.T.)
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, China
- Correspondence: ; Tel./Fax: +86-028-8547-1599
| |
Collapse
|
39
|
Investigation of biocidal efficacy of commercial disinfectants used in public, private and workplaces during the pandemic event of SARS-CoV-2. Sci Rep 2022; 12:5468. [PMID: 35361869 PMCID: PMC8969816 DOI: 10.1038/s41598-022-09575-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/25/2022] [Indexed: 12/24/2022] Open
Abstract
This study investigated the performance of 24 commercial disinfectants present on the market during last year according to the manufacturer’s instructions. Recently, national and international organizations of public health performed studies on disinfection products due to the increasing awareness of the potential and growing risks on human health, such as skin damage and reactions in the mucosal lining, especially for the healthcare workers in their frequent daily use. However, there are many limitations in the common cleaning/disinfection products on market as in the selection of effective disinfectants to decontaminate inanimate surfaces. We analyzed the disinfection power of hydrogen peroxide, quaternary ammonium compounds, alcohols, phenols and aldehydes used as active principles according to international guidelines. The antimicrobial properties were assessed by broth microdilution, and antibiofilm properties against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus); their virucidal efficacy was tested against Herpes simplex virus type 1 (HSV-1) and Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The quaternary ammonium compounds demonstrated better efficacy than others and in some cases ready to use products had also virucidal and antimicrobial activities after dilution at 0.125%. The scientific evidence indicates that many commercial products are used at high concentrations and high doses and this could have deleterious effects both on human health and the environment. A lower concentration of active ingredients would avoid the excessive release of chemicals into the environment and improve skin tolerance, ensuring the health and safety protection of workers, including the healthcare operators at their workplace.
Collapse
|
40
|
Zhang Y, Xu S, Yang Y, Chou SH, He J. A 'time bomb' in the human intestine-the multiple emergence and spread of antibiotic-resistant bacteria. Environ Microbiol 2021; 24:1231-1246. [PMID: 34632679 DOI: 10.1111/1462-2920.15795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 11/30/2022]
Abstract
Antibiotics have a strong killing effect on bacteria and are the first choice for the prevention and treatment of bacterial infectious diseases. Therefore, they have been widely used in the medical field, animal husbandry and planting industry. However, with the massive use of antibiotics, more and more antibiotic-resistant bacteria (ARB) have emerged. Because human intestines are rich in nutrients, have suitable temperature, and are high in bacterial abundance, they can easily become a hotbed for the spread of ARB and antibiotic-resistant genes (ARGs). When opportunistic pathogenic bacteria in the intestine acquire ARGs, the infectious diseases caused by such opportunistic pathogens will become more difficult to treat, or even impossible to cure. Therefore, ARB in the human intestine are like a 'time bomb'. In this review, we discuss the sources of intestinal ARB and the transmission routes of ARGs in the human intestine from the perspective of One Health. Further, we describe various methods to prevent the emergence of ARB and inhibit the spread of ARGs in the human intestine. Finally, we may be able to overcome ARB in the human intestine using an interdisciplinary 'One Health' approach.
Collapse
Affiliation(s)
- Yuling Zhang
- State Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Siyang Xu
- State Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yijun Yang
- State Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Shan-Ho Chou
- State Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Jin He
- State Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| |
Collapse
|