1
|
Zhao S, Xu W, Zeng W, He S, Luo X, Ge D, Duan Y, Han R, Chen CY, Yang Y, Hu Y, Zhang C. Engineered bacterial outer membrane vesicles co-delivering Angio-3 and doxorubicin to enhance tumor therapy. Colloids Surf B Biointerfaces 2025; 253:114707. [PMID: 40262304 DOI: 10.1016/j.colsurfb.2025.114707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
Bacterial outer membrane vesicles (OMVs) have emerged as versatile nanomaterial-based drug delivery systems that can stimulate systemic immune responses and facilitate precise co-delivery of multiple therapeutic agents. This study introduces a bioengineering approach that enables the co-delivery of the angiogenesis inhibitor Angio-3 and the chemotherapeutic agent doxorubicin (DOX) within OMVs, creating a potent antitumor therapeutic platform. Angio-3 displayed on the surface of OMVs inhibited angiogenesis and decreased vascular permeability, which in turn impeded the supply of nutrients necessary for tumor growth. Moreover, intrinsic properties of OMVs triggered a systemic immune response. Both in vitro and in vivo studies, including a CT26 tumor-bearing mouse model, have demonstrated that the OMV@A&D-based therapeutic regimen, which integrates antiangiogenesis, chemotherapy, and immune activation, significantly suppresses tumor proliferation. This study highlights the potential of bioengineered OMVs in revolutionizing cancer therapy by offering a multifaceted and synergistic platform that enhances therapeutic outcomes.
Collapse
Affiliation(s)
- Shuai Zhao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Wenxuan Xu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Wanting Zeng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Shuailin He
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Xuan Luo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Dejie Ge
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Yamin Duan
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Rui Han
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Chin-Yu Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Yong Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Yunhong Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China.
| | - Cheng Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China.
| |
Collapse
|
2
|
Inoue H, Kawano K, Kawamoto J, Ogawa T, Kurihara T. Rapid screening and identification of genes involved in bacterial extracellular membrane vesicle production using a curvature-sensing peptide. J Bacteriol 2025:e0049724. [PMID: 40183544 DOI: 10.1128/jb.00497-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/09/2025] [Indexed: 04/05/2025] Open
Abstract
Bacteria secrete extracellular membrane vesicles (EMVs). Physiological functions and biotechnological applications of these lipid nanoparticles have been attracting significant attention. However, the details of the molecular basis of EMV biogenesis have not yet been fully elucidated. In our previous work, an N-terminus-substituted FAAV peptide labeled with nitrobenzoxadiazole (NBD; nFAAV5-NBD) was developed. This peptide can sense the curvature of a lipid bilayer and selectively bind to EMVs even in the presence of cells. Here, we applied nFAAV5-NBD to a genome-wide screening of hyper- and hypo-vesiculation transposon mutants of a Gram-negative bacterium, Shewanella vesiculosa HM13, to identify the genes involved in EMV production. We analyzed the transposon insertion sites in hyper- and hypo-vesiculation mutants and identified 16 and six genes, respectively, with a transposon inserted within or near them. Targeted gene-disrupted mutants of the identified genes showed that the lack of putative dipeptidyl carboxypeptidase, glutamate synthase β-subunit, LapG protease, metallohydrolase, RNA polymerase sigma-54 factor, inactive transglutaminase, PepSY domain-containing protein, and Rhs-family protein caused EMV overproduction. On the other hand, disruption of the genes encoding putative phosphoenolpyruvate synthase, d-hexose-6-phosphate epimerase, NAD-specific glutamate dehydrogenase, and sensory box histidine kinase/response regulator decreased EMV production. This study demonstrates the utility of a novel screening method using a curvature-sensing peptide for mutants with altered EMV productivity and provides information on the genes related to EMV production.IMPORTANCEConventional methods for isolation and quantification of extracellular membrane vesicles (EMVs) are generally time-consuming. nFAAV5-NBD can detect EMVs in the culture without separating EMVs from cells. In situ detection of EMVs using this peptide facilitated screening of the genes related to EMV production. We succeeded in identifying various genes associated with EMV production of Shewanella vesiculosa HM13, which would contribute to the elucidation of bacterial EMV formation mechanisms. Additionally, the hyper-vesiculating mutants obtained in this study would be valuable for EMV applications, such as secreting useful substances as EMV cargoes and producing artificially functionalized EMVs.
Collapse
Affiliation(s)
- Hiromu Inoue
- Institute for Chemical Research, Kyoto University, Uji, Kyoto, Japan
| | - Kenichi Kawano
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Jun Kawamoto
- Institute for Chemical Research, Kyoto University, Uji, Kyoto, Japan
| | - Takuya Ogawa
- Institute for Chemical Research, Kyoto University, Uji, Kyoto, Japan
| | - Tatsuo Kurihara
- Institute for Chemical Research, Kyoto University, Uji, Kyoto, Japan
| |
Collapse
|
3
|
Wu Z, Long W, Yin Y, Tan B, Liu C, Li H, Ge S. Outer membrane vesicles of Porphyromonas gingivalis: recent advances in pathogenicity and associated mechanisms. Front Microbiol 2025; 16:1555868. [PMID: 40256625 PMCID: PMC12007433 DOI: 10.3389/fmicb.2025.1555868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/20/2025] [Indexed: 04/22/2025] Open
Abstract
Periodontitis is a chronic infectious inflammatory disease primarily caused by periodontal pathogenic bacteria, which poses a significant threat to human health. The pathogenic mechanisms associated with Porphyromonas gingivalis (P. gingivalis), a principal causative agent of periodontitis, are particularly complex and warrant thorough investigation. The extensive array of virulence factors released by this bacterium during its growth and pathogenesis not only inflicts localized damage to periodontal tissues but is also intricately linked to the development of systemic diseases through various mechanisms. The outer membrane vesicles (OMVs) produced by P. gingivalis play a key role in this process. These OMVs serve as important mediators of communication between bacteria and host cells and other bacteria, carrying and delivering virulence factors to host cells and distant tissues, thereby damaging host cells and exacerbating inflammatory responses. The ability of these OMVs to disseminate and deliver bacterial virulence factors allows P. gingivalis to play a pathogenic role far beyond the confines of the periodontal tissue and has been closely associated with the development of a variety of systemic diseases such as cardiovascular disease, Alzheimer's disease, rheumatoid arthritis, diabetes mellitus, non-alcoholic hepatitis, and cancer. In view of this, it is of great pathophysiological and clinical significance to deeply investigate its pathogenic role and related mechanisms. This will not only help to better understand the pathogenesis of periodontitis and its related systemic diseases but also provide new ideas and more effective and precise strategies for the early diagnosis, prevention, and treatment of these diseases. However, the current research in this field is still insufficient and in-depth, and many key issues and mechanisms need to be further elucidated. This article summarizes the recent research progress on the role of P. gingivalis OMVs (P. g-OMVs) in related diseases, with the aim of providing a theoretical basis and direction for future research and revealing the pathogenic mechanism of P. g-OMVs more comprehensively.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Song Ge
- School and Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
4
|
Müller GA. The Transformation Experiment of Frederick Griffith I: Its Narrowing and Potential for the Creation of Novel Microorganisms. Bioengineering (Basel) 2025; 12:324. [PMID: 40150788 PMCID: PMC11939280 DOI: 10.3390/bioengineering12030324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
The construction of artificial microorganisms often relies on the transfer of genomes from donor to acceptor cells. This synthetic biology approach has been considerably fostered by the J. Craig Venter Institute but apparently depends on the use of microorganisms, which are very closely related. One reason for this limitation of the "creative potential" of "classical" transformation is the requirement for adequate "fitting" of newly synthesized polypeptide components, directed by the donor genome, to interacting counterparts encoded by the pre-existing acceptor genome. Transformation was introduced in 1928 by Frederick Griffith in the course of the demonstration of the instability of pneumococci and their conversion from rough, non-pathogenic into smooth, virulent variants. Subsequently, this method turned out to be critical for the identification of DNA as the sole matter of inheritance. Importantly, the initial experimental design (1.0) also considered the inheritance of both structural (e.g., plasma membranes) and cybernetic information (e.g., metabolite fluxes), which, in cooperation, determine topological and cellular heredity, as well as fusion and blending of bacterial cells. In contrast, subsequent experimental designs (1.X) were focused on the use of whole-cell homogenates and, thereafter, of soluble and water-clear fractions deprived of all information and macromolecules other than those directing protein synthesis, including outer-membrane vesicles, bacterial prions, lipopolysaccharides, lipoproteins, cytoskeletal elements, and complexes thereof. Identification of the reasons for this narrowing may be helpful in understanding the potential of transformation for the creation of novel microorganisms.
Collapse
Affiliation(s)
- Günter A. Müller
- Biology and Technology Studies Institute Munich (BITSIM), 80939 Munich, Germany; ; Tel.: +49-151-25216987
- Institute of Media Sociology, Department of Cultural Sciences, University of Paderborn, 33104 Paderborn, Germany
| |
Collapse
|
5
|
Ali-Khiavi P, Mohammadi M, Masoumi S, Saffarfar H, Kheradmand R, Mobed A, Hatefnia F. The Therapeutic Potential of Exosome Therapy in Sepsis Management: Addressing Complications and Improving Outcomes". Cell Biochem Biophys 2025; 83:307-326. [PMID: 39363035 DOI: 10.1007/s12013-024-01564-7] [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] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
Infection occurs when pathogens penetrate tissues, reproduce, and trigger a host response to both the infectious agents and their toxins. A diverse array of pathogens, including viruses and bacteria, can cause infections. The host's immune system employs several mechanisms to combat these infections, typically involving an innate inflammatory response. Inflammation is a complex biological reaction that can affect various parts of the body and is a key component of the response to harmful stimuli. Sepsis arises when the body's response to infection leads to widespread damage to tissues and organs, potentially resulting in severe outcomes or death. The initial phase of sepsis involves immune system suppression. Early identification and targeted management are crucial for improving sepsis outcomes. Common treatment approaches include antibiotics, intravenous fluids, blood cultures, and monitoring urine output. This study explores the potential of exosome therapy in enhancing the management and alleviation of sepsis symptoms.
Collapse
Affiliation(s)
- Payam Ali-Khiavi
- Medical faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahya Mohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Masoumi
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Kheradmand
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Faezeh Hatefnia
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
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
|
7
|
Wang J, Wang X, Luo H, Xie Y, Cao H, Mao L, Liu T, Yue Y, Qian H. Extracellular vesicles in Helicobacter pylori-mediated diseases: mechanisms and therapeutic potential. Cell Commun Signal 2025; 23:79. [PMID: 39934861 DOI: 10.1186/s12964-025-02074-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
Extracellular vesicles (EVs) are relevant elements for cell-to-cell communication and are considered crucial in host-pathogen interactions by transferring molecules between the pathogen and the host during infections. These structures participate in various physiological and pathological processes and are considered promising candidates as disease markers, therapeutic reagents, and drug carriers. Both H. pylori and the host epithelial cells infected by H. pylori secrete EVs, which contribute to inflammation and the development of disease phenotypes. However, many aspects of the cellular and molecular biology of EV functions remain incompletely understood due to methodological challenges in studying these small structures. This review also highlights the roles of EVs derived from H. pylori-infected cells in the pathogenesis of gastric and extragastric diseases. Understanding the specific functions of these EVs during H. pylori infections, whether are advantageous to the host or the pathogen, may help the development new therapeutic approaches to prevent disease.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, 215300, China
| | - Xiuping Wang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, 215300, China
| | - Hao Luo
- Department of Clinical Laboratory, The Second People's Hospital of Kunshan, Suzhou, Jiangsu, 215300, China
| | - Yiping Xie
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, 215300, China
| | - Hui Cao
- Department of Food and Nutrition Safety, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, 210003, China
| | - Lingxiang Mao
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, 215300, China
| | - Tingting Liu
- Science and Technology Talent Department, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, 215300, China
| | - Yushan Yue
- Department of Rehabilitative Medicine, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, Jiangsu, 215300, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhengjiang, Jiangsu, 212013, China.
| |
Collapse
|
8
|
Torabian P, Singh N, Crawford J, Gonzalez G, Burgado N, Videva M, Miller A, Perdue J, Dinu M, Pietropaoli A, Gaborski T, Michel LV. Effect of clinically relevant antibiotics on bacterial extracellular vesicle release from Escherichia coli. Int J Antimicrob Agents 2025; 65:107384. [PMID: 39542065 DOI: 10.1016/j.ijantimicag.2024.107384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Sepsis, a leading cause of death in hospitals, can be defined as a dysregulated host inflammatory response to infection, which can lead to tissue damage, organ failure and cardiovascular complications. Although there is no cure for sepsis, the condition is typically managed with broad-spectrum antibiotics to eliminate any potential bacterial source of infection. However, a potential side effect of antibiotic treatment is the enhanced release of bacterial extracellular vesicles (BEVs), membrane-bound nanoparticles containing proteins and other biological molecules from their parent bacterium. Some of the Gram-negative extracellular vesicle (EV) cargo, including peptidoglycan associated lipoprotein and outer membrane protein A, have been shown to induce both acute and chronic inflammation in host tissue. It was hypothesized that the antibiotic concentration and mechanism of action may affect the amount of released BEVs, which could potentially exacerbate the host inflammatory response. This study evaluated nine clinically relevant antibiotics for their effect on EV release from Escherichia coli. Several beta-lactam antibiotics caused significantly more EV release, while quinolone and aminoglycoside antibiotics caused less vesiculation. Further study is warranted to corroborate the correlation between an antibiotic's mechanism of action and its effect on EV release, but these results underline the importance of antibiotic choice when treating patients with sepsis.
Collapse
Affiliation(s)
- Panteha Torabian
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Navraj Singh
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, USA
| | - James Crawford
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Gabriela Gonzalez
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Nicholas Burgado
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Martina Videva
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Aidan Miller
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Janai Perdue
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Milena Dinu
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Anthony Pietropaoli
- Department of Medicine, Pulmonary Diseases and Critical Care, University of Rochester, Rochester, NY, USA
| | - Thomas Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Lea Vacca Michel
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, USA.
| |
Collapse
|
9
|
Jia X, Yuan B, Wang W, Wang K, Ling D, Wei M, Hu Y, Guo W, Chen Z, Du L, Jin Y. Gene editing tool-loaded biomimetic cationic vesicles with highly efficient bacterial internalization for in vivo eradication of pathogens. J Nanobiotechnology 2024; 22:787. [PMID: 39710679 DOI: 10.1186/s12951-024-03065-4] [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: 10/08/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
In the post-COVID-19 era, drug-resistant bacterial infections emerge as one of major death causes, where multidrug-resistant Acinetobacter baumannii (MRAB) and drug-resistant Pseudomonas aeruginosa (DRPA) represent primary pathogens. However, the classical antibiotic strategy currently faces the bottleneck of drug resistance. We develop an antimicrobial strategy that applies the selective delivery of CRISPR/Cas9 plasmids to pathogens with biomimetic cationic hybrid vesicles (BCVs), irrelevant to bacterial drug resistance. CRISPR/Cas9 plasmids were constructed, replicating in MRAB or DRPA and expressing ribonucleic proteins, leading to irreparable chromosomal lesions; however, delivering the negatively charged plasmids with extremely large molecular weight to the pathogens at the infection site became a huge challenge. We found that the BCVs integrating the bacterial out membrane vesicles and cationic lipids efficiently delivered the plasmids in vitro/in vivo to the pathogens followed by effective internalization. The BCVs were used by intratracheal or topical hydrogel application against MRAB pulmonary infection or DRPA wound infection, and both of the two pathogens were eradicated from the lung or the wound. CRISPR/Cas9 plasmid-loaded BCVs become a promising medication for drug-resistant bacteria infections.
Collapse
Affiliation(s)
- Xueli Jia
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
- Department of Pharmaceutical Science, School of Pharmacy, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China.
| | - Wanmei Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Ke Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Dandan Ling
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Meng Wei
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Yadan Hu
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Wanting Guo
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Ziyuan Chen
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Lina Du
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China.
| |
Collapse
|
10
|
Zhang L, Li F, Zhao D, Duan L, Bai W, Yan B. Research trends and focus of prosthetic joint infections from 2013 to 2023: bibliometric and visualization studies. Front Microbiol 2024; 15:1507340. [PMID: 39760080 PMCID: PMC11695429 DOI: 10.3389/fmicb.2024.1507340] [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: 10/10/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025] Open
Abstract
Background Postoperative infections in artificial joints provide considerable difficulties in the field of orthopedics, especially after joint replacement procedures. These infections rank among the most severe postoperative consequences, frequently leading to treatment ineffectiveness and reduced quality of life for surgery patients. Consequently, it is crucial to acquire knowledge about worldwide research trends in this area in order to educate clinical practices and improve therapeutic techniques. This work exploits bibliometric analysis to investigate the present state, developing patterns, and main areas of focus in research on artificial joint infection. Objective To analyze the research trends, hotspots, and international collaborations on artificial joint infections worldwide from 2013 to 2023. Methods Extractions of raw data were made from the WoSCC (Web of Science Core Collection) database. Detailed information collected includes the quantity of publications, authors, citations, publication year, h-index, references, country/region, journal, and keywords. Analysis of the data was conducted using VOSviewer version 1.6.10.0 and CiteSpace version 6.3.R1. Results A total of 1,799 articles published between 2013 and 2023 were included in this analysis, showing a steady increase in publication with the United States leading at 553 articles. Infection rates and topics such as biofilm formation and antimicrobial resistance were highly cited, with Mayo Clinic contributing 65 articles as the most prolific institution. Conclusion Research on biofilm infections, antibiotic resistance, and new biomarkers is a key focus, particularly on disrupting biofilms and enhancing diagnostics. There's growing attention in biomarkers like α-defensins and exosomal miRNAs for PJI diagnosis, pointing to new clinical uses. Studies on antimicrobial-coated prosthetics and topical agents are also gaining importance in treatment strategies.
Collapse
Affiliation(s)
- Liwen Zhang
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fei Li
- Department of Orthopedics, Affiliated Hospital of Shandong Traditional Chinese Medicine University, Jinan, China
| | - Diqian Zhao
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Duan
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenzhe Bai
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Orthopedics, Affiliated Hospital of Shandong Traditional Chinese Medicine University, Jinan, China
| | - Bing Yan
- Department of Orthopedics, Affiliated Hospital of Shandong Traditional Chinese Medicine University, Jinan, China
| |
Collapse
|
11
|
Choubey R, Chatterjee M, Pandey PK, Mishra A, Datta B. Coassembly of Cell-Penetrating Peptide Octaarginine with Acetazolamide: Emergent Interactions with E. coli. ACS OMEGA 2024; 9:46204-46216. [PMID: 39583731 PMCID: PMC11579775 DOI: 10.1021/acsomega.4c06800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
The investigation of established pharmaceutical agents for recalibrating usage strongly supplements new drug development. In this work, we have prepared coassembled complexes of acetazolamide (AZM) with the cationic peptide octaarginine (R8) in an attempt to enhance its potency and scope of use. R8 and AZM in different weight ratios coassemble into remarkable nano- and microstructures such as ribbons, sheets, and stick-like structures. A combination of FTIR, XRD, SEM, and DSC has been used to characterize the R8:AZM coassemblies. The sulfonamide SO2 and NH2 groups of AZM are associated with the guanidinium amine, free amine, and terminal carbonyl groups of R8 resulting in distinctive topologies. Treatment of Escherichia coli with the complexes results in a distinctive pattern of membrane disruption and pore formation. The R8:AZM coassemblies inhibit carbonic anhydrase and E. coli growth with greater efficiency compared to bare AZM. The 1:5 w/w complex leads to pronounced outer and inner membrane rupture and significantly restricts glucose uptake by E. coli. The ability of R8 and AZM to coassemble into a distinctive set of structures based solely on differences in their relative proportions and their engagement with E. coli as more than the sum of their parts are novel facets of R8 and AZM behavior and underscore a straightforward and elegant approach for enhancing the scope of use of small molecule drugs.
Collapse
Affiliation(s)
- Rinku Choubey
- Department
of Chemistry, Indian Institute of Technology
Gandhinagar, Palaj, Gandhinagar 382355, India
| | - Moumita Chatterjee
- Department
of Chemistry, Indian Institute of Technology
Gandhinagar, Palaj, Gandhinagar 382355, India
| | - Pramina Kumari Pandey
- Department
of Materials Engineering, Indian Institute
of Technology Gandhinagar, Palaj, Gandhinagar 382355, India
| | - Abhijit Mishra
- Department
of Materials Engineering, Indian Institute
of Technology Gandhinagar, Palaj, Gandhinagar 382355, India
| | - Bhaskar Datta
- Department
of Chemistry, Indian Institute of Technology
Gandhinagar, Palaj, Gandhinagar 382355, India
- Department
of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, India
| |
Collapse
|
12
|
Sakalauskienė GV, Radzevičienė A. Antimicrobial Resistance: What Lies Beneath This Complex Phenomenon? Diagnostics (Basel) 2024; 14:2319. [PMID: 39451642 PMCID: PMC11506786 DOI: 10.3390/diagnostics14202319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
Antimicrobial Resistance (AMR) has evolved from a mere concern into a significant global threat, with profound implications for public health, healthcare systems, and the global economy. Since the introduction of antibiotics between 1945 and 1963, their widespread and often indiscriminate use in human medicine, agriculture, and animal husbandry has led to the emergence and rapid spread of antibiotic-resistant genes. Bacteria have developed sophisticated mechanisms to evade the effects of antibiotics, including drug uptake limitation, drug degradation, target modification, efflux pumps, biofilm formation, and outer membrane vesicles production. As a result, AMR now poses a threat comparable to climate change and the COVID-19 pandemic, and projections suggest that death rates will be up to 10 million deaths annually by 2050, along with a staggering economic cost exceeding $100 trillion. Addressing AMR requires a multifaceted approach, including the development of new antibiotics, alternative therapies, and a significant shift in antibiotic usage and regulation. Enhancing global surveillance systems, increasing public awareness, and prioritizing investments in research, diagnostics, and vaccines are critical steps. By recognizing the gravity of the AMR threat and committing to collaborative action, its impact can be mitigated, and global health can be protected for future generations.
Collapse
Affiliation(s)
- Giedrė Valdonė Sakalauskienė
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania;
| | | |
Collapse
|
13
|
Henriquez T, Santoro F, Medaglini D, Pallecchi L, Clemente I, Bonechi C, Magnani A, Paccagnini E, Gentile M, Lupetti P, Marvasi M, Pini A, Bracci L, Falciani C. Analysis of the utility of a rapid vesicle isolation method for clinical strains of Pseudomonas aeruginosa. Microbiol Spectr 2024; 12:e0064924. [PMID: 39248554 PMCID: PMC11448148 DOI: 10.1128/spectrum.00649-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024] Open
Abstract
Pseudomonas aeruginosa, a pathogen capable of causing diseases ranging from mild to life-threatening, has a large arsenal of virulence factors. Notably, extracellular vesicles have emerged as significant players in the pathogenesis of this organism. However, the full range of their functions is still being studied, and difficulties related to vesicle purification (long protocols, low yields, and specialized instruments) have become a major obstacle for their characterization. In this context, the utility of rapid new methods of vesicle isolation from clinical strains is still unknown. Here, we analyze the utility of the ExoBacteria OMV isolation kit for a collection of clinical strains of P. aeruginosa. We first phenotypically characterized 15 P. aeruginosa strains to ensure that our samples were heterogeneous. We then determined the best conditions for purifying vesicles from P. aeruginosa PAO1 reference strain by the rapid method and used them to isolate vesicles from clinical strains. Our results indicated that M9 minimal medium is the best for obtaining high purity with the rapid isolation kit. Although we were able to isolate vesicles from at least four strains, the low yield and the large number of strains with unpurifiable vesicles showed that the kit was not practical or convenient for clinical strains. Our findings suggest that although fast procedures for vesicle purification can be of great utility for Escherichia coli, the more complex phenotypes of clinical isolates of P. aeruginosa are a challenge for these protocols and new alternatives/optimizations need to be developed.IMPORTANCEPseudomonas aeruginosa is recognized as an opportunistic pathogen in humans and animals. It can effectively colonize various environments thanks to a large set of virulence factors that include extracellular vesicles. Different methods were recently developed to reduce the time and effort associated with vesicle purification. However, the utility of rapid vesicle isolation methods for clinical strains of P. aeruginosa (which are recognized as being highly diverse) is not yet known. In this context, we analyzed the utility of the ExoBacteria OMV Isolation kit for vesicle purification in P. aeruginosa clinical strains. Our findings showed that the kit does not seem to be convenient for research on clinical strains due to low vesicle recovery. Our results underscore the importance of developing new rapid vesicle purification protocols/techniques for specific clinical phenotypes.
Collapse
Affiliation(s)
- Tania Henriquez
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesco Santoro
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Donata Medaglini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Lucia Pallecchi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ilaria Clemente
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Claudia Bonechi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Agnese Magnani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | | | - Pietro Lupetti
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Alessandro Pini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Luisa Bracci
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Chiara Falciani
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
14
|
Bai Z, Wang X, Liang T, Xu G, Cai J, Xu W, Yang K, Hu L, Pei P. Harnessing Bacterial Membrane Components for Tumor Vaccines: Strategies and Perspectives. Adv Healthc Mater 2024; 13:e2401615. [PMID: 38935934 DOI: 10.1002/adhm.202401615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Tumor vaccines stand at the vanguard of tumor immunotherapy, demonstrating significant potential and promise in recent years. While tumor vaccines have achieved breakthroughs in the treatment of cancer, they still encounter numerous challenges, including improving the immunogenicity of vaccines and expanding the scope of vaccine application. As natural immune activators, bacterial components offer inherent advantages in tumor vaccines. Bacterial membrane components, with their safer profile, easy extraction, purification, and engineering, along with their diverse array of immune components, activate the immune system and improve tumor vaccine efficacy. This review systematically summarizes the mechanism of action and therapeutic effects of bacterial membranes and its derivatives (including bacterial membrane vesicles and hybrid membrane biomaterials) in tumor vaccines. Subsequently, the authors delve into the preparation and advantages of tumor vaccines based on bacterial membranes and hybrid membrane biomaterials. Following this, the immune effects of tumor vaccines based on bacterial outer membrane vesicles are elucidated, and their mechanisms are explained. Moreover, their advantages in tumor combination therapy are analyzed. Last, the challenges and trends in this field are discussed. This comprehensive analysis aims to offer a more informed reference and scientific foundation for the design and implementation of bacterial membrane-based tumor vaccines.
Collapse
Affiliation(s)
- Zhenxin Bai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xuanyu Wang
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, People's Republic of China
| | - Tianming Liang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, P.R. China
| | - Guangyu Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jinzhou Cai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, P.R. China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Pei Pei
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, People's Republic of China
| |
Collapse
|
15
|
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
|
16
|
Dhurve G, Behera SR, Kodetham G, Siddavattam D. Outer membrane vesicles of Acinetobacter baumannii DS002 carry circular DNA similar to bovine meat and milk factors (BMMFs) and SPHINX 2.36 and probably play a role in interdomain lateral gene transfer. Microbiol Spectr 2024; 12:e0081724. [PMID: 39101807 PMCID: PMC11370262 DOI: 10.1128/spectrum.00817-24] [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: 04/02/2024] [Accepted: 06/20/2024] [Indexed: 08/06/2024] Open
Abstract
The discovery of Replication Competent Circular DNA molecules in mammalian cells and tissues is being linked to debilitating diseases, such as multiple sclerosis (MS), bovine spongiform encephalopathy (BSE), and colorectal cancer (CRC). These circular DNA molecules, otherwise known as bovine meat and milk factors (BMMFs) and Slow Progressive Hidden INfections of variable (X) latency (SPHINX), bear significant (80%) sequence similarity with the plasmids of Acinetobacter baumannii strains. Nanostructures, such as bacterial outer membrane vesicles (OMVs) serve as vehicles for transporting biomolecular cargo and have the potential to facilitate interkingdom lateral mobility of DNA. Strengthening the proposed hypothesis, this study demonstrates that OMVs derived from A. baumannii DS002 carrying four plasmids and genome (pTS236) of phage, AbDs1, successfully reached different parts of the body, including the central nervous system, following the injection of fluorescein isothiocyanate (FITC)-labeled OMVs into experimental mice. Out of the four OMV-associated plasmids, three (pTS4586, pTS9900, and pTS134338) were identified within the lumen, and the fourth one (pTS11291) was found on the surface of OMVs. In addition to the indigenous plasmids, the phage-encoded protein, Orf96, anchored on the surface of the OMVs by establishing a strong interaction with the OMV-associated porin, OmpA. Intriguingly, a subset of labeled OMVs, when incubated with Neuro2A cells, translocated across the membrane and reached to the cytoplasmic space of the cells. Collectively, the experimental evidence presented herein underscores the promising potential of OMVs as vehicles for delivering molecular cargo containing plasmids and phage genomes to diverse mammalian tissues and cells. IMPORTANCE Several independent studies have demonstrated the existence of replication competent circular DNA molecules of bacterial and viral origin in mammalian cells and tissues. However, studies about their origin and lateral mobility to mammalian cells are scarce. Our work describes the existence of circular DNA, similar to that of DNA molecules identified in mammalian cells, OMVs derived from soil isolate of A. baumannii DS002. Furthermore, the work also provides visual evidence that demonstrates the passage of labeled OMVs to different organs of experimental mice within hours after intravenously administering OMVs into experimental mice. Some of the labeled OMVs have even crossed the membrane of Neuro2A, suggesting the existence of interkingdom horizontal mobility between bacteria and mammals.
Collapse
Affiliation(s)
- Ganeshwari Dhurve
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Sandhya Rani Behera
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Gopinath Kodetham
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Dayananda Siddavattam
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| |
Collapse
|
17
|
Luo Z, Cheng X, Feng B, Fan D, Liu X, Xie R, Luo T, Wegner SV, Ma D, Chen F, Zeng W. Engineering Versatile Bacteria-Derived Outer Membrane Vesicles: An Adaptable Platform for Advancing Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400049. [PMID: 38952055 PMCID: PMC11434149 DOI: 10.1002/advs.202400049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 06/13/2024] [Indexed: 07/03/2024]
Abstract
In recent years, cancer immunotherapy has undergone a transformative shift toward personalized and targeted therapeutic strategies. Bacteria-derived outer membrane vesicles (OMVs) have emerged as a promising and adaptable platform for cancer immunotherapy due to their unique properties, including natural immunogenicity and the ability to be engineered for specific therapeutic purposes. In this review, a comprehensive overview is provided of state-of-the-art techniques and methodologies employed in the engineering of versatile OMVs for cancer immunotherapy. Beginning by exploring the biogenesis and composition of OMVs, unveiling their intrinsic immunogenic properties for therapeutic appeal. Subsequently, innovative approaches employed to engineer OMVs are delved into, ranging from the genetic engineering of parent bacteria to the incorporation of functional molecules. The importance of rational design strategies is highlighted to enhance the immunogenicity and specificity of OMVs, allowing tailoring for diverse cancer types. Furthermore, insights into clinical studies and potential challenges utilizing OMVs as cancer vaccines or adjuvants are also provided, offering a comprehensive assessment of the current landscape and future prospects. Overall, this review provides valuable insights for researchers involved in the rapidly evolving field of cancer immunotherapy, offering a roadmap for harnessing the full potential of OMVs as a versatile and adaptable platform for cancer treatment.
Collapse
Affiliation(s)
- Ziheng Luo
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Xiang Cheng
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Bin Feng
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Duoyang Fan
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Xiaohui Liu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Ruyan Xie
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Ting Luo
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Seraphine V. Wegner
- Institute of Physiological Chemistry and PathobiochemistryUniversity of Münster48149MünsterGermany
| | - Dayou Ma
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Fei Chen
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| |
Collapse
|
18
|
Spari D, Schmid A, Sanchez-Taltavull D, Murugan S, Keller K, Ennaciri N, Salm L, Stroka D, Beldi G. Released bacterial ATP shapes local and systemic inflammation during abdominal sepsis. eLife 2024; 13:RP96678. [PMID: 39163101 PMCID: PMC11335348 DOI: 10.7554/elife.96678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024] Open
Abstract
Sepsis causes millions of deaths per year worldwide and is a current global health priority declared by the WHO. Sepsis-related deaths are a result of dysregulated inflammatory immune responses indicating the need to develop strategies to target inflammation. An important mediator of inflammation is extracellular adenosine triphosphate (ATP) that is released by inflamed host cells and tissues, and also by bacteria in a strain-specific and growth-dependent manner. Here, we investigated the mechanisms by which bacteria release ATP. Using genetic mutant strains of Escherichia coli (E. coli), we demonstrate that ATP release is dependent on ATP synthase within the inner bacterial membrane. In addition, impaired integrity of the outer bacterial membrane notably contributes to ATP release and is associated with bacterial death. In a mouse model of abdominal sepsis, local effects of bacterial ATP were analyzed using a transformed E. coli bearing an arabinose-inducible periplasmic apyrase hydrolyzing ATP to be released. Abrogating bacterial ATP release shows that bacterial ATP suppresses local immune responses, resulting in reduced neutrophil counts and impaired survival. In addition, bacterial ATP has systemic effects via its transport in outer membrane vesicles (OMV). ATP-loaded OMV are quickly distributed throughout the body and upregulated expression of genes activating degranulation in neutrophils, potentially contributing to the exacerbation of sepsis severity. This study reveals mechanisms of bacterial ATP release and its local and systemic roles in sepsis pathogenesis.
Collapse
Affiliation(s)
- Daniel Spari
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University Hospital of BernBernSwitzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University Hospital of BernBernSwitzerland
| | - Annina Schmid
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University Hospital of BernBernSwitzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University Hospital of BernBernSwitzerland
| | - Daniel Sanchez-Taltavull
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University Hospital of BernBernSwitzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University Hospital of BernBernSwitzerland
| | - Shaira Murugan
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University Hospital of BernBernSwitzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University Hospital of BernBernSwitzerland
| | - Keely Keller
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University Hospital of BernBernSwitzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University Hospital of BernBernSwitzerland
| | - Nadia Ennaciri
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University Hospital of BernBernSwitzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University Hospital of BernBernSwitzerland
| | - Lilian Salm
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University Hospital of BernBernSwitzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University Hospital of BernBernSwitzerland
| | - Deborah Stroka
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University Hospital of BernBernSwitzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University Hospital of BernBernSwitzerland
| | - Guido Beldi
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University Hospital of BernBernSwitzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University Hospital of BernBernSwitzerland
| |
Collapse
|
19
|
Casillo A, D'Amico R, Lanzetta R, Corsaro MM. Marine Delivery Vehicles: Molecular Components and Applications of Bacterial Extracellular Vesicles. Mar Drugs 2024; 22:363. [PMID: 39195479 DOI: 10.3390/md22080363] [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: 07/04/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
In marine ecosystems, communication among microorganisms is crucial since the distance is significant if considered on a microbial scale. One of the ways to reduce this gap is through the production of extracellular vesicles, which can transport molecules to guarantee nutrients to the cells. Marine bacteria release extracellular vesicles (EVs), small membrane-bound structures of 40 nm to 1 µm diameter, into their surrounding environment. The vesicles contain various cellular compounds, including lipids, proteins, nucleic acids, and glycans. EVs may contribute to dissolved organic carbon, thus facilitating heterotroph growth. This review will focus on marine bacterial EVs, analyzing their structure, composition, functions, and applications.
Collapse
Affiliation(s)
- Angela Casillo
- Department of Chemical Sciences, University of Naples Federico II, Complesso Monte S. Angelo, 80126 Naples, Italy
| | - Raffaele D'Amico
- Department of Chemical Sciences, University of Naples Federico II, Complesso Monte S. Angelo, 80126 Naples, Italy
| | - Rosa Lanzetta
- Department of Chemical Sciences, University of Naples Federico II, Complesso Monte S. Angelo, 80126 Naples, Italy
| | - Maria Michela Corsaro
- Department of Chemical Sciences, University of Naples Federico II, Complesso Monte S. Angelo, 80126 Naples, Italy
| |
Collapse
|
20
|
Margutti P, D’Ambrosio A, Zamboni S. Microbiota-Derived Extracellular Vesicle as Emerging Actors in Host Interactions. Int J Mol Sci 2024; 25:8722. [PMID: 39201409 PMCID: PMC11354844 DOI: 10.3390/ijms25168722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
The human microbiota is an intricate micro-ecosystem comprising a diverse range of dynamic microbial populations mainly consisting of bacteria, whose interactions with hosts strongly affect several physiological and pathological processes. The gut microbiota is being increasingly recognized as a critical player in maintaining homeostasis, contributing to the main functions of the intestine and distal organs such as the brain. However, gut dysbiosis, characterized by composition and function alterations of microbiota with intestinal barrier dysfunction has been linked to the development and progression of several pathologies, including intestinal inflammatory diseases, systemic autoimmune diseases, such as rheumatic arthritis, and neurodegenerative diseases, such as Alzheimer's disease. Moreover, oral microbiota research has gained significant interest in recent years due to its potential impact on overall health. Emerging evidence on the role of microbiota-host interactions in health and disease has triggered a marked interest on the functional role of bacterial extracellular vesicles (BEVs) as mediators of inter-kingdom communication. Accumulating evidence reveals that BEVs mediate host interactions by transporting and delivering into host cells effector molecules that modulate host signaling pathways and cell processes, influencing health and disease. This review discusses the critical role of BEVs from the gut, lung, skin and oral cavity in the epithelium, immune system, and CNS interactions.
Collapse
Affiliation(s)
- Paola Margutti
- Department of Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.); (S.Z.)
| | | | | |
Collapse
|
21
|
Saad MG, Beyenal H, Dong WJ. Dual roles of the conditional extracellular vesicles derived from Pseudomonas aeruginosa biofilms: Promoting and inhibiting bacterial biofilm growth. Biofilm 2024; 7:100183. [PMID: 38380422 PMCID: PMC10876606 DOI: 10.1016/j.bioflm.2024.100183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/19/2023] [Accepted: 02/05/2024] [Indexed: 02/22/2024] Open
Abstract
Antibiotic-resistant biofilm infections have emerged as public health concerns because of their enhanced tolerance to high-dose antibiotic treatments. The biofilm life cycle involves multiple developmental stages, which are tightly regulated by active cell-cell communication via specific extracellular signal messengers such as extracellular vesicles. This study was aimed at exploring the roles of extracellular vesicles secreted by Pseudomonas aeruginosa at different developmental stages in controlling biofilm growth. Our results show that extracellular vesicles secreted by P. aeruginosa biofilms during their exponential growth phase (G-EVs) enhance biofilm growth. In contrast, extracellular vesicles secreted by P. aeruginosa biofilms during their death/survival phase (D-EVs) can effectively inhibit/eliminate P. aeruginosa PAO1 biofilms up to 4.8-log10 CFU/cm2. The inhibition effectiveness of D-EVs against P. aeruginosa biofilms grown for 96 h improved further in the presence of 10-50 μM Fe3+ ions. Proteomic analysis suggests the inhibition involves an iron-dependent ferroptosis mechanism. This study is the first to report the functional role of bacterial extracellular vesicles in bacterial growth, which depends on the developmental stage of the parent bacteria. The finding of D-EV-activated ferroptosis-based bacterial death may have significant implications for preventing antibiotic resistance in biofilms.
Collapse
Affiliation(s)
- Marwa Gamal Saad
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164, USA
| | - Haluk Beyenal
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164, USA
| | - Wen-Ji Dong
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164, USA
| |
Collapse
|
22
|
Liang R, Li P, Yang N, Xiao X, Gong J, Zhang X, Bai Y, Chen Y, Xie Z, Liao Q. Parabacteroides distasonis-Derived Outer Membrane Vesicles Enhance Antitumor Immunity Against Colon Tumors by Modulating CXCL10 and CD8 + T Cells. Drug Des Devel Ther 2024; 18:1833-1853. [PMID: 38828018 PMCID: PMC11144014 DOI: 10.2147/dddt.s457338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
Purpose Given the potent immunostimulatory effects of bacterial outer membrane vesicles (OMVs) and the significant anti-colon tumor properties of Parabacteroides distasonis (Pd), this study aimed to elucidate the role and potential mechanisms of Pd-derived OMVs (Pd-OMVs) against colon cancer. Methods This study isolated and purified Pd-OMVs from Pd cultures and assessed their characteristics. The effects of Pd-OMVs on CT26 cell uptake, proliferation, and invasion were investigated in vitro. In vivo, a CT26 colon tumor model was used to investigate the anti-colon tumor effects and underlying mechanisms of Pd-OMVs. Finally, we evaluated the biosafety of Pd-OMVs. Results Purified Pd-OMVs had a uniform cup-shaped structure with an average size of 165.5 nm and a zeta potential of approximately -9.56 mV, and their proteins were associated with pathways related to immunity and apoptosis. In vitro experiments demonstrated that CT26 cells internalized the Pd-OMVs, resulting in a significant decrease in their proliferation and invasion abilities. Further in vivo studies confirmed the accumulation of Pd-OMVs in tumor tissues, which significantly inhibited the growth of colon tumors. Mechanistically, Pd-OMVs increased the expression of CXCL10, promoting infiltration of CD8+ T cells into tumor tissues and expression of pro-inflammatory factors TNF-α, IL-1β, and IL-6. Notably, Pd-OMVs demonstrated a high level of biosafety. Conclusion This paper elucidates that Pd-OMVs can exert significant anti-colon tumor effects by upregulating the expression of the chemokine CXCL10, thereby increasing the infiltration of CD8+ T cells into tumors and enhancing antitumor immune responses. This suggests that Pd-OMVs may be developed as a novel nanoscale potent immunostimulant with great potential for application in tumor immunotherapy. As well as developed as a novel nano-delivery carrier for combination with other antitumor drugs.
Collapse
Affiliation(s)
- Rongyao Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Pei Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Na Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Xiaoyi Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Jing Gong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Xingyuan Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Yunuan Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Yanlong Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong Province, People’s Republic of China
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| |
Collapse
|
23
|
Shao Z, Lu Y, Xing A, He X, Xie H, Hu M. Effect of outer membrane vesicles of Lactobacillus pentosus on Tau phosphorylation and CDK5-Calpain pathway in mice. Exp Gerontol 2024; 189:112400. [PMID: 38484904 DOI: 10.1016/j.exger.2024.112400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/16/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Alzheimer's disease (AD) stands as a neurodegenerative disorder causing cognitive decline, posing a significant health concern for the elderly population in China. This study explored the effects of outer membrane vesicles (OMVs) from the gut microbiota of AD patients on learning and memory abilities and Tau protein phosphorylation in mice. In contrast to the OMVs from healthy controls and the PBS treatment group, mice treated with AD-OMVs exhibited notable declines in learning and memory capabilities, as evidenced by results from the Morris water maze, Y-maze, and novel object recognition tests. Immunohistochemistry and Western blot assessments unveiled elevated levels of hyperphosphorylated Tau in the cortex and hippocampus of mice treated with AD-OMVs. However, there were no alterations observed in the total Tau levels. In addition, AD-OMVs treated mice showed increased neuroinflammation indicated by elevated astrocytes and microglia. Molecular mechanism studies demonstrated that AD-OMVs could activate GSK3β, CDK5-Calpain and NF-κB pathways in mice hippocampus. These studies suggest AD patient gut microbiota derived OMVs can promote host Tau phosphorylation and improved neuroinflammation.
Collapse
Affiliation(s)
- Zhongying Shao
- Department of liver diseases, Tai'an Traditional Chinese Medicine Hospital, Tai'an City, Shandong Province, China
| | - Yanjun Lu
- Department of liver diseases, Tai'an Traditional Chinese Medicine Hospital, Tai'an City, Shandong Province, China
| | - Aihong Xing
- TCM Prevent&Health Care Dept Tai'an Traditional Chinese Medicine Hospital, Tai'an City, Shandong Province, China
| | - Xiying He
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an City, Shandong Province, China
| | - Hongyan Xie
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an City, Shandong Province, China
| | - Ming Hu
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an City, Shandong Province, China.
| |
Collapse
|
24
|
Liu C, Yazdani N, Moran CS, Salomon C, Seneviratne CJ, Ivanovski S, Han P. Unveiling clinical applications of bacterial extracellular vesicles as natural nanomaterials in disease diagnosis and therapeutics. Acta Biomater 2024; 180:18-45. [PMID: 38641182 DOI: 10.1016/j.actbio.2024.04.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/03/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are naturally occurring bioactive membrane-bound nanoparticles released by both gram-negative and gram-positive bacterial species, exhibiting a multifaceted role in mediating host-microbe interactions across various physiological conditions. Increasing evidence supports BEVs as essential mediators of cell-to-cell communicaiton, influencing bacterial pathogenicity, disease mechanisms, and modulating the host immune response. However, the extent to which these BEV-mediated actions can be leveraged to predict disease onset, guide treatment strategies, and determine clinical outcomes remains uncertain, particularly in terms of their clinical translation potentials. This review briefly describes BEV biogenesis and their internalisation by recipient cells and summarises methods for isolation and characterization, essential for understanding their composition and cargo. Further, it discusses the potential of biofluid-associated BEVs as biomarkers for various diseases, spanning both cancer and non-cancerous conditions. Following this, we outline the ongoing human clinical trials of using BEVs for vaccine development. In addition to disease diagnostics, this review explores the emerging research of using natural or engineered BEVs as smart nanomaterials for applications in anti-cancer therapy and bone regeneration. This discussion extends to key factors for unlocking the clinical potential of BEVs, such as standardization of BEV isolation and characterisation, as well as other hurdles in translating these findings to the clinical setting. We propose that addressing these hurdles through collaborative research efforts and well-designed clinical trials holds the key to fully harnessing the clinical potential of BEVs. As this field advances, this review suggests that BEV-based nanomedicine has the potential to revolutionize disease management, paving the way for innovative diagnosis, therapeutics, and personalized medicine approaches. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EVs) from both host cells and bacteria serve as multifunctional biomaterials and are emerging in the fields of biomedicine, bioengineering, and biomaterials. However, the majority of current studies focus on host-derived EVs, leaving a gap in comprehensive research on bacteria-derived EVs (BEVs). Although BEVs offer an attractive option as nanomaterials for drug delivery systems, their unique nanostructure and easy-to-modify functions make them a potential method for disease diagnosis and treatment as well as vaccine development. Our work among the pioneering studies investigating the potential of BEVs as natural nanobiomaterials plays a crucial role in both understanding the development of diseases and therapeutic interventions.
Collapse
Affiliation(s)
- Chun Liu
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia
| | - Negar Yazdani
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia
| | - Corey S Moran
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029 Australia
| | - Chaminda Jayampath Seneviratne
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia
| | - Sašo Ivanovski
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia.
| | - Pingping Han
- The University of Queensland, School of Dentistry, Centre for Oralfacial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics Nanodiagnostic and Therapeutic Group, Brisbane, QLD 4006, Australia.
| |
Collapse
|
25
|
Shi G, Yang X, Wang J, Wei W, Hu K, Huang X, Qiu Y, He Y. Isolation of Extracellular Outer Membrane Vesicles (OMVs) from Escherichia coli Using EVscore47 Beads. Molecules 2024; 29:1831. [PMID: 38675651 PMCID: PMC11054804 DOI: 10.3390/molecules29081831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Outer membrane vesicles (OMVs) are attractive for biomedical applications based on their intrinsic properties in relation to bacteria and vesicles. However, their widespread use is hampered by low yields and purities. In this study, EVscore47 multifunctional chromatography microspheres were synthesized and used to efficiently isolate functional OMVs from Escherichia coli. Through this technology, OMV loss can be kept to a minimum, and OMVs can be harvested using EVscore47 at 11-fold higher yields and ~13-fold higher purity than those achieved by means of ultracentrifugation. Based on the results presented here, we propose a novel EVscore47-based isolation of OMVs that is fast and scalable.
Collapse
Affiliation(s)
- Gongming Shi
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, Chongqing 401331, China;
| | - Xiaohong Yang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Beibei, Chongqing 400714, China; (X.Y.); (W.W.); (K.H.); (X.H.); (Y.Q.)
| | - Jikai Wang
- Nanjing Aidimai Technology Co., Ltd., 18 Zhilan Road, Jiangning, Nanjing 211100, China;
| | - Wenjing Wei
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Beibei, Chongqing 400714, China; (X.Y.); (W.W.); (K.H.); (X.H.); (Y.Q.)
| | - Kecui Hu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Beibei, Chongqing 400714, China; (X.Y.); (W.W.); (K.H.); (X.H.); (Y.Q.)
| | - Xingyue Huang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Beibei, Chongqing 400714, China; (X.Y.); (W.W.); (K.H.); (X.H.); (Y.Q.)
| | - Yanfei Qiu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Beibei, Chongqing 400714, China; (X.Y.); (W.W.); (K.H.); (X.H.); (Y.Q.)
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, Chongqing 401331, China;
| |
Collapse
|
26
|
Pschunder B, Locati L, López O, Martin Aispuro P, Zurita E, Stuible M, Durocher Y, Hozbor D. Outer membrane vesicles derived from Bordetella pertussis are potent adjuvant that drive Th1-biased response. Front Immunol 2024; 15:1387534. [PMID: 38650936 PMCID: PMC11033331 DOI: 10.3389/fimmu.2024.1387534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
For several years, we have been committed to exploring the potential of Bordetella pertussis-derived outer membrane vesicles (OMVBp) as a promising third-generation vaccine against the reemerging pertussis disease. The results of our preclinical trials not only confirm its protective capacity against B. pertussis infection but also set the stage for forthcoming human clinical trials. This study delves into the examination of OMVBp as an adjuvant. To accomplish this objective, we implemented a two-dose murine schedule to evaluate the specific immune response induced by formulations containing OMVBp combined with 3 heterologous immunogens: Tetanus toxoid (T), Diphtheria toxoid (D), and the SARS-CoV-2 Spike protein (S). The specific levels of IgG, IgG1, and IgG2a triggered by the different tested formulations were evaluated using ELISA in dose-response assays for OMVBp and the immunogens at varying levels. These assays demonstrated that OMVBp exhibits adjuvant properties even at the low concentration employed (1.5 μg of protein per dose). As this effect was notably enhanced at medium (3 μg) and high concentrations (6 μg), we chose the medium concentration to determine the minimum immunogen dose at which the OMV adjuvant properties are significantly evident. These assays demonstrated that OMVBp exhibits adjuvant properties even at the lowest concentration tested for each immunogen. In the presence of OMVBp, specific IgG levels detected for the lowest amount of antigen tested increased by 2.5 to 10 fold compared to those found in animals immunized with formulations containing adjuvant-free antigens (p<0.0001). When assessing the adjuvant properties of OMVBp compared to the widely recognized adjuvant alum, we detected similar levels of specific IgG against D, T and S for both adjuvants. Experiments with OMVs derived from E. coli (OMVE.coli) reaffirmed that the adjuvant properties of OMVs extend across different bacterial species. Nonetheless, it's crucial to highlight that OMVBp notably skewed the immune response towards a Th1 profile (p<0.05). These collective findings emphasize the dual role of OMVBp as both an adjuvant and modulator of the immune response, positioning it favorably for incorporation into combined vaccine formulations.
Collapse
Affiliation(s)
- Bernarda Pschunder
- Laboratorio Vacunas Salud (VacSal), Instituto de Biotecnología y Biología Molecular (IBBM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Centro Científico Tecnológico-Consejo Nacional de Investigaciones Científicas y Técnicas (CCT-CONICET) La Plata, La Plata, Argentina
| | - Lucia Locati
- Laboratorio Vacunas Salud (VacSal), Instituto de Biotecnología y Biología Molecular (IBBM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Centro Científico Tecnológico-Consejo Nacional de Investigaciones Científicas y Técnicas (CCT-CONICET) La Plata, La Plata, Argentina
| | - Oriana López
- Laboratorio Vacunas Salud (VacSal), Instituto de Biotecnología y Biología Molecular (IBBM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Centro Científico Tecnológico-Consejo Nacional de Investigaciones Científicas y Técnicas (CCT-CONICET) La Plata, La Plata, Argentina
| | - Pablo Martin Aispuro
- Laboratorio Vacunas Salud (VacSal), Instituto de Biotecnología y Biología Molecular (IBBM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Centro Científico Tecnológico-Consejo Nacional de Investigaciones Científicas y Técnicas (CCT-CONICET) La Plata, La Plata, Argentina
| | - Eugenia Zurita
- Laboratorio Vacunas Salud (VacSal), Instituto de Biotecnología y Biología Molecular (IBBM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Centro Científico Tecnológico-Consejo Nacional de Investigaciones Científicas y Técnicas (CCT-CONICET) La Plata, La Plata, Argentina
| | - Matthew Stuible
- Human Health Therapeutics Research Center, National Research Council Canada, Montreal, QC, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Center, National Research Council Canada, Montreal, QC, Canada
| | - Daniela Hozbor
- Laboratorio Vacunas Salud (VacSal), Instituto de Biotecnología y Biología Molecular (IBBM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Centro Científico Tecnológico-Consejo Nacional de Investigaciones Científicas y Técnicas (CCT-CONICET) La Plata, La Plata, Argentina
| |
Collapse
|
27
|
Iaconis A, De Plano LM, Caccamo A, Franco D, Conoci S. Anti-Biofilm Strategies: A Focused Review on Innovative Approaches. Microorganisms 2024; 12:639. [PMID: 38674584 PMCID: PMC11052202 DOI: 10.3390/microorganisms12040639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Biofilm (BF) can give rise to systemic infections, prolonged hospitalization times, and, in the worst case, death. This review aims to provide an overview of recent strategies for the prevention and destruction of pathogenic BFs. First, the main phases of the life cycle of BF and maturation will be described to identify potential targets for anti-BF approaches. Then, an approach acting on bacterial adhesion, quorum sensing (QS), and the extracellular polymeric substance (EPS) matrix will be introduced and discussed. Finally, bacteriophage-mediated strategies will be presented as innovative approaches against BF inhibition/destruction.
Collapse
Affiliation(s)
- Antonella Iaconis
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Laura Maria De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Domenico Franco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Sabrina Conoci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy
- URT Lab Sens Beyond Nano—CNR-DSFTM, Department of Physical Sciences and Technologies of Matter, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
28
|
Li Q, Li J, He T, Ji X, Wei R, Yu M, Wang R. Sub-MIC Antibiotics Modulate Productions of Outer Membrane Vesicles in Tigecycline-Resistant Escherichia coli. Antibiotics (Basel) 2024; 13:276. [PMID: 38534711 DOI: 10.3390/antibiotics13030276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/13/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
Antimicrobial resistance (AMR) has been recognized as one of the most important crises affecting global human health in the 21st century. Tigecycline is one of the last resort antibiotics for treating severe infections caused by multi-drug resistant Enterobacteriaceae. However, the mobile resistance gene tet(X4), which could mediate high-level tigecycline resistance, was discovered in 2019. The outer membrane vesicle (OMV) has been recognized as a new route for horizontal gene transfer; antimicrobial resistant bacteria also have the ability to secret OMVs, while little is known about the impact of antibiotics on the secretion and characteristics of OMVs from tigecycline resistant bacteria till now. This study aimed to investigate the effects of antibiotics on the production and traits of a tigecycline resistant Escherichia coli strain of 47EC. The results showed that sub-inhibitory (1/2 MIC or 1/4 MIC) concentrations of gentamicin, meropenem, ceftazidime, chloramphenicol, tigecycline, ciprofloxacin, polymycin, rifaximin and mitomycin C could significantly increase the secretion of OMVs (0.713 ± 0.05~6.333 ± 0.15 mg/mL) from E. coli 47EC compared to the respective untreated control (0.709 ± 0.03 mg/mL). In addition, the particle sizes of OMVs were generally larger, and the zeta potential were lower in the antibiotics-treated groups than those of the antibiotic-free group. The copy numbers of the tigecycline resistance gene of tet(X4) in the OMVs of most antimicrobial-treated groups were higher than that of the control group. Moreover, transcriptome analysis on ciprofloxacin-treated E. coli 47EC indicated that the SOS response and prophage activation might participate in the ciprofloxacin-induced OMV formation. In conclusion, the clinical application of antibiotics in treating bacterial infections, especially multi-drug resistant bacteria, might lead to the increased secretion of bacterial OMVs and the enrichment of antimicrobial-resistant genes in the OMVs.
Collapse
Affiliation(s)
- Qianru Li
- School of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Key Laboratory of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Key Laboratory of Agro-Product Safety Risk Evaluation (Nanjing) of Ministry of Agriculture and Rural Affairs, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jun Li
- Key Laboratory of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Key Laboratory of Agro-Product Safety Risk Evaluation (Nanjing) of Ministry of Agriculture and Rural Affairs, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Tao He
- Key Laboratory of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Key Laboratory of Agro-Product Safety Risk Evaluation (Nanjing) of Ministry of Agriculture and Rural Affairs, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Xing Ji
- Key Laboratory of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Key Laboratory of Agro-Product Safety Risk Evaluation (Nanjing) of Ministry of Agriculture and Rural Affairs, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Ruicheng Wei
- Key Laboratory of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Key Laboratory of Agro-Product Safety Risk Evaluation (Nanjing) of Ministry of Agriculture and Rural Affairs, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Meiling Yu
- School of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Ran Wang
- Key Laboratory of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Key Laboratory of Agro-Product Safety Risk Evaluation (Nanjing) of Ministry of Agriculture and Rural Affairs, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| |
Collapse
|
29
|
Liu J, You Q, Liang F, Ma L, Zhu L, Wang C, Yang Y. Ultrasound-nanovesicles interplay for theranostics. Adv Drug Deliv Rev 2024; 205:115176. [PMID: 38199256 DOI: 10.1016/j.addr.2023.115176] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/04/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Nanovesicles (NVs) are widely used in the treatment and diagnosis of diseases due to their excellent vascular permeability, good biocompatibility, high loading capacity, and easy functionalization. However, their yield and in vivo penetration depth limitations and their complex preparation processes still constrain their application and development. Ultrasound, as a fundamental external stimulus with deep tissue penetration, concentrated energy sources, and good safety, has been proven to be a patient-friendly and highly efficient strategy to overcome the restrictions of traditional clinical medicine. Recent research has shown that ultrasound can drive the generation of NVs, increase their yield, simplify their preparation process, and provide direct therapeutic effects and intelligent control to enhance the therapeutic effect of NVs. In addition, NVs, as excellent drug carriers, can enhance the targeting efficiency of ultrasound-based sonodynamic therapy or sonogenetic regulation and improve the accuracy of ultrasound imaging. This review provides a detailed introduction to the classification, generation, and modification strategies of NVs, emphasizing the impact of ultrasound on the formation of NVs and summarizing the enhanced treatment and diagnostic effects of NVs combined with ultrasound for various diseases.
Collapse
Affiliation(s)
- Jingyi Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Fuming Liang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lilusi Ma
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
30
|
Park JH, Song S, Kim S, Kim M, Kim KS. Optimizing Conditions for the Production of Bacterial Extracellular Vesicles of Vibrio vulnificus and Analysis of the Inner Small RNA Compositions. J Microbiol Biotechnol 2024; 34:29-38. [PMID: 38044684 PMCID: PMC10840491 DOI: 10.4014/jmb.2310.10002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/05/2023]
Abstract
Chemical and physical elements affecting the production of bacterial extracellular vesicles (BEVs) of the human pathogen Vibrio vulnificus were quantitatively assessed to optimize the conditions for the BEV production by using the western blot quantification for an outer membrane porin OmpU and by fluorescent dye FM4-64. When cells were cultured at 37°C in an enriched medium (2 × Luria Bertani; 2 × LB) in the presence of EDTA, they produced about 70% more BEVs. BEVs were purified from the cells cultured in the established optimal conditions by the density gradient ultracentrifugation. The dynamic light scattering measurement of the purified BEVs showed that the diameter of them ranged from approximately 25 nm to 161 nm. We hypothesized that there may be some features in nucleotide sequences specific to RNAs packaged in BEVs compared to those in cellular RNA molecules. We compared the nucleotide sequences and abundance of sRNAs between in the cellular fraction and in BEVs through next-generation sequencing (NGS). While no distinct feature was observed in the nucleotide sequences of sRNAs between two groups, the length of sRNA fragments from BEVs were significantly shorter than those in cytoplasm.
Collapse
Affiliation(s)
- Jeong Heon Park
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Suji Song
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Soyee Kim
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Minjeong Kim
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Kun-Soo Kim
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| |
Collapse
|
31
|
Orsini Delgado ML, Gamelas Magalhaes J, Morra R, Cultrone A. Muropeptides and muropeptide transporters impact on host immune response. Gut Microbes 2024; 16:2418412. [PMID: 39439228 PMCID: PMC11509177 DOI: 10.1080/19490976.2024.2418412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
In bacteria, the cell envelope is the key element surrounding and protecting the bacterial content from mechanical or osmotic damages. It allows the selective interchanges of solutes, ions, cellular debris, and drugs between the cellular compartments and the external environment, thanks to the presence of transmembrane proteins called transporters. The major component of the cell envelope is the peptidoglycan, consisting of long linear glycan strands cross-linked by short peptide stems. During cell growth or under stress conditions, peptidoglycan fragments, the muropeptides, are released by bacteria and recognized by the host Pattern Recognition Receptor, promoting the activation of their innate defense mechanisms. The review sums up the salient aspects of microbiota-host interaction with a focus on the NOD-dependent immune response to bacterial peptidoglycan and on the accountability of muropeptide transporters in the crosstalk with the host and in antibiotic resistance. Furthermore, it retraces the discoveries and applications of microorganisms-derived components such as vaccines or vaccine adjuvants.
Collapse
|
32
|
Blache A, Achouak W. Extraction and Purification of Outer Membrane Vesicles and Their Associated RNAs. Methods Mol Biol 2024; 2741:11-24. [PMID: 38217646 DOI: 10.1007/978-1-0716-3565-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
Outer membrane vesicles (OMVs), produced by Gram negative-bacteria and sRNAs, are key players in cell-to-cell communication and interactions of bacteria with the environment. OMVs act as information carriers and encapsulate various molecules such as proteins, lipids, metabolites, and RNAs. OMVs and sRNAs play a broad range of functions from pathogenesis to stress resistance, to biofilm formation and both mediate interkingdom signaling. Various studies indicate that there is a mechanism of intercellular communication mediated by OMV-derived bacterial RNAs that is conserved among certain bacterial species. Here we describe methods for the extraction and purification of vesicles produced by Gram-negative bacteria, such as Pseudomonas brassicacearum and Escherichia coli, and address methods for the extraction of OMVs-derived sRNA and techniques for the analysis of sRNAs.
Collapse
Affiliation(s)
- Anaïs Blache
- CEA, CNRS, Aix Marseille University Lab of Microbial Ecology of the Rhizosphere (LEMiRE), UMR7265 BIAM, Saint-Paul-lez-Durance, France
| | - Wafa Achouak
- CEA, CNRS, Aix Marseille University Lab of Microbial Ecology of the Rhizosphere (LEMiRE), UMR7265 BIAM, Saint-Paul-lez-Durance, France.
| |
Collapse
|
33
|
Zheng K, Feng Y, Li L, Kong F, Gao J, Kong X. Engineered bacterial outer membrane vesicles: a versatile bacteria-based weapon against gastrointestinal tumors. Theranostics 2024; 14:761-787. [PMID: 38169585 PMCID: PMC10758051 DOI: 10.7150/thno.85917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 10/01/2023] [Indexed: 01/05/2024] Open
Abstract
Outer membrane vesicles (OMVs) are nanoscale lipid bilayer structures released by gram-negative bacteria. They share membrane composition and properties with their originating cells, making them adept at traversing cellular barriers. These OMVs have demonstrated exceptional membrane stability, immunogenicity, safety, penetration, and tumor-targeting properties, which have been leveraged in developing vaccines and drug delivery systems. Recent research efforts have focused on engineering OMVs to increase production yield, reduce cytotoxicity, and improve the safety and efficacy of treatment. Notably, gastrointestinal (GI) tumors have proven resistant to several traditional oncological treatment strategies, including chemotherapy, radiotherapy, and targeted therapy. Although immune checkpoint inhibitors have demonstrated efficacy in some patients, their usage as monotherapy remains limited by tumor heterogeneity and individual variability. The immunogenic and modifiable nature of OMVs makes them an ideal design platform for the individualized treatment of GI tumors. OMV-based therapy enables combination therapy and optimization of anti-tumor effects. This review comprehensively summarizes recent advances in OMV engineering for GI tumor therapy and discusses the challenges in the clinical translation of emerging OMV-based anti-tumor therapies.
Collapse
Affiliation(s)
- Keshuang Zheng
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yongpu Feng
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lei Li
- Digestive Endoscopy Center, Shanghai Tenth People's Hospital, Shanghai, China
| | - Fanyang Kong
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jie Gao
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiangyu Kong
- National Key Laboratory of Immunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
34
|
Jones EJ, Stentz R, Parker A, Carding SR. Assessing In Vivo Bacterial Extracellular Vesicle (BEV) Biodistribution Using Fluorescent Lipophilic Membrane Stains. Methods Mol Biol 2024; 2843:239-251. [PMID: 39141304 DOI: 10.1007/978-1-0716-4055-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are nano-size vesicles containing a cargo of bioactive molecules that can play key roles in microbe-microbe and microbe-host interactions. In tracking their biodistribution in vivo, BEVs can cross several physical host barriers including the intestinal epithelium, vascular endothelium, and blood-brain-barrier (BBB) to ultimately accumulate in tissues such as the liver, lungs, spleen, and the brain. This tissue-specific dissemination has been exploited for the delivery of biomolecules such as vaccines for mucosal delivery. Although numerous strategies for labeling and tracking BEVs have been described, most have constraints that impact on interpreting in vivo bioimaging patterns. Here, we describe a general method for labeling BEVs using lipophilic fluorescent membrane stains which can be adopted by non-expert users. We also describe how the procedure can be used to overcome potential limitations. Furthermore, we outline methods of quantitative ex vivo tissue imaging that can be used to evaluate BEV organ trafficking.
Collapse
Affiliation(s)
- Emily J Jones
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich, UK
| | - Regis Stentz
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich, UK
| | - Aimée Parker
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich, UK
| | - Simon R Carding
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich, UK.
- Norwich Medical School, University of East Anglia, Norwich, UK.
| |
Collapse
|
35
|
Juodeikis R, Martins C, Saalbach G, Richardson J, Koev T, Baker DJ, Defernez M, Warren M, Carding SR. Differential temporal release and lipoprotein loading in B. thetaiotaomicron bacterial extracellular vesicles. J Extracell Vesicles 2024; 13:e12406. [PMID: 38240185 PMCID: PMC10797578 DOI: 10.1002/jev2.12406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/24/2023] [Accepted: 01/01/2024] [Indexed: 01/22/2024] Open
Abstract
Bacterial extracellular vesicles (BEVs) contribute to stress responses, quorum sensing, biofilm formation and interspecies and interkingdom communication. However, the factors that regulate their release and heterogeneity are not well understood. We set out to investigate these factors in the common gut commensal Bacteroides thetaiotaomicron by studying BEV release throughout their growth cycle. Utilising a range of methods, we demonstrate that vesicles released at different stages of growth have significantly different composition, with early vesicles enriched in specifically released outer membrane vesicles (OMVs) containing a larger proportion of lipoproteins, while late phase BEVs primarily contain lytic vesicles with enrichment of cytoplasmic proteins. Furthermore, we demonstrate that lipoproteins containing a negatively charged signal peptide are preferentially incorporated in OMVs. We use this observation to predict all Bacteroides thetaiotaomicron OMV enriched lipoproteins and analyse their function. Overall, our findings highlight the need to understand media composition and BEV release dynamics prior to functional characterisation and define the theoretical functional capacity of Bacteroides thetaiotaomicron OMVs.
Collapse
Affiliation(s)
- Rokas Juodeikis
- Food, Microbiome, and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
| | | | | | | | - Todor Koev
- Food, Microbiome, and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
- School of PharmacyUniversity of East AngliaNorwichUK
| | - Dave J. Baker
- Food, Microbiome, and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
| | - Marianne Defernez
- Food, Microbiome, and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
| | - Martin Warren
- Food, Microbiome, and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
- School of BiosciencesUniversity of KentCanterburyUK
- School of Biological SciencesUniversity of East AngliaNorwichUK
| | - Simon R. Carding
- Food, Microbiome, and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
- Norwich Medical SchoolUniversity of East AngliaNorwichUK
| |
Collapse
|
36
|
Bitzenhofer NL, Höfel C, Thies S, Weiler AJ, Eberlein C, Heipieper HJ, Batra‐Safferling R, Sundermeyer P, Heidler T, Sachse C, Busche T, Kalinowski J, Belthle T, Drepper T, Jaeger K, Loeschcke A. Exploring engineered vesiculation by Pseudomonas putida KT2440 for natural product biosynthesis. Microb Biotechnol 2024; 17:e14312. [PMID: 37435812 PMCID: PMC10832525 DOI: 10.1111/1751-7915.14312] [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: 01/04/2023] [Accepted: 06/25/2023] [Indexed: 07/13/2023] Open
Abstract
Pseudomonas species have become promising cell factories for the production of natural products due to their inherent robustness. Although these bacteria have naturally evolved strategies to cope with different kinds of stress, many biotechnological applications benefit from engineering of optimised chassis strains with specially adapted tolerance traits. Here, we explored the formation of outer membrane vesicles (OMV) of Pseudomonas putida KT2440. We found OMV production to correlate with the recombinant production of a natural compound with versatile beneficial properties, the tripyrrole prodigiosin. Further, several P. putida genes were identified, whose up- or down-regulated expression allowed controlling OMV formation. Finally, genetically triggering vesiculation in production strains of the different alkaloids prodigiosin, violacein, and phenazine-1-carboxylic acid, as well as the carotenoid zeaxanthin, resulted in up to three-fold increased product yields. Consequently, our findings suggest that the construction of robust strains by genetic manipulation of OMV formation might be developed into a useful tool which may contribute to improving limited biotechnological applications.
Collapse
Affiliation(s)
- Nora Lisa Bitzenhofer
- Institute of Molecular Enzyme Technology (IMET)Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Carolin Höfel
- Institute of Molecular Enzyme Technology (IMET)Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Stephan Thies
- Institute of Molecular Enzyme Technology (IMET)Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Andrea Jeanette Weiler
- Institute of Molecular Enzyme Technology (IMET)Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Christian Eberlein
- Department of Environmental BiotechnologyHelmholtz Centre for Environmental Research (UFZ)LeipzigGermany
| | - Hermann J. Heipieper
- Department of Environmental BiotechnologyHelmholtz Centre for Environmental Research (UFZ)LeipzigGermany
| | - Renu Batra‐Safferling
- Institute of Biological Information Processing – Structural Biochemistry (IBI‐7: Structural Biochemistry)Forschungszentrum JülichJülichGermany
| | - Pia Sundermeyer
- Ernst‐Ruska Centre for Microscopy and Spectroscopy with Electrons (ER‐C‐3/Structural Biology)Forschungszentrum JülichJülichGermany
- Institute for Biological Information Processing 6 (IBI‐6/ Structural Cellular Biology)Forschungszentrum JülichJülichGermany
| | - Thomas Heidler
- Ernst‐Ruska Centre for Microscopy and Spectroscopy with Electrons (ER‐C‐3/Structural Biology)Forschungszentrum JülichJülichGermany
- Institute for Biological Information Processing 6 (IBI‐6/ Structural Cellular Biology)Forschungszentrum JülichJülichGermany
| | - Carsten Sachse
- Ernst‐Ruska Centre for Microscopy and Spectroscopy with Electrons (ER‐C‐3/Structural Biology)Forschungszentrum JülichJülichGermany
- Institute for Biological Information Processing 6 (IBI‐6/ Structural Cellular Biology)Forschungszentrum JülichJülichGermany
- Department of BiologyHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Tobias Busche
- Center for Biotechnology (CeBiTec)Bielefeld UniversityBielefeldGermany
- Bielefeld University, Medical School East Westphalia‐LippeBielefeld UniversityBielefeldGermany
| | - Jörn Kalinowski
- Center for Biotechnology (CeBiTec)Bielefeld UniversityBielefeldGermany
| | - Thomke Belthle
- DWI─Leibniz‐Institute for Interactive MaterialsAachenGermany
- Functional and Interactive Polymers, Institute of Technical and Macromolecular ChemistryRWTH Aachen UniversityAachenGermany
| | - Thomas Drepper
- Institute of Molecular Enzyme Technology (IMET)Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Karl‐Erich Jaeger
- Institute of Molecular Enzyme Technology (IMET)Heinrich Heine University DüsseldorfDüsseldorfGermany
- Institute of Bio‐ and Geosciences IBG‐1: BiotechnologyForschungszentrum JülichJülichGermany
| | - Anita Loeschcke
- Institute of Molecular Enzyme Technology (IMET)Heinrich Heine University DüsseldorfDüsseldorfGermany
| |
Collapse
|
37
|
Weng Z, Yang N, Shi S, Xu Z, Chen Z, Liang C, Zhang X, Du X. Outer Membrane Vesicles from Acinetobacter baumannii: Biogenesis, Functions, and Vaccine Application. Vaccines (Basel) 2023; 12:49. [PMID: 38250862 PMCID: PMC10818702 DOI: 10.3390/vaccines12010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
This review focuses on Acinetobacter baumannii, a Gram-negative bacterium that causes various infections and whose multidrug resistance has become a significant challenge in clinical practices. There are multiple bacterial mechanisms in A. baumannii that participate in bacterial colonization and immune responses. It is believed that outer membrane vesicles (OMVs) budding from the bacteria play a significant role in mediating bacterial survival and the subsequent attack against the host. Most OMVs originate from the bacterial membranes and molecules are enveloped in them. Elements similar to the pathogen endow OMVs with robust virulence, which provides a new direction for exploring the pathogenicity of A. baumannii and its therapeutic pathways. Although extensive research has been carried out on the feasibility of OMV-based vaccines against pathogens, no study has yet summarized the bioactive elements, biological activity, and vaccine applicability of A. baumannii OMVs. This review summarizes the components, biogenesis, and function of OMVs that contribute to their potential as vaccine candidates and the preparation methods and future directions for their development.
Collapse
Affiliation(s)
- Zheqi Weng
- The Second Clinical Medical School, Nanjing Medical University, Nanjing 210011, China; (Z.W.); (S.S.); (Z.X.); (Z.C.); (C.L.)
| | - Ning Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China;
| | - Shujun Shi
- The Second Clinical Medical School, Nanjing Medical University, Nanjing 210011, China; (Z.W.); (S.S.); (Z.X.); (Z.C.); (C.L.)
| | - Zining Xu
- The Second Clinical Medical School, Nanjing Medical University, Nanjing 210011, China; (Z.W.); (S.S.); (Z.X.); (Z.C.); (C.L.)
| | - Zixu Chen
- The Second Clinical Medical School, Nanjing Medical University, Nanjing 210011, China; (Z.W.); (S.S.); (Z.X.); (Z.C.); (C.L.)
| | - Chen Liang
- The Second Clinical Medical School, Nanjing Medical University, Nanjing 210011, China; (Z.W.); (S.S.); (Z.X.); (Z.C.); (C.L.)
| | - Xiuwei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| | - Xingran Du
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| |
Collapse
|
38
|
Fan M, Kiefer P, Charki P, Hedberg C, Seibel J, Vorholt JA, Hilbi H. The Legionella autoinducer LAI-1 is delivered by outer membrane vesicles to promote interbacterial and interkingdom signaling. J Biol Chem 2023; 299:105376. [PMID: 37866633 PMCID: PMC10692735 DOI: 10.1016/j.jbc.2023.105376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023] Open
Abstract
Legionella pneumophila is an environmental bacterium, which replicates in amoeba but also in macrophages, and causes a life-threatening pneumonia called Legionnaires' disease. The opportunistic pathogen employs the α-hydroxy-ketone compound Legionella autoinducer-1 (LAI-1) for intraspecies and interkingdom signaling. LAI-1 is produced by the autoinducer synthase Legionella quorum sensing A (LqsA), but it is not known, how LAI-1 is released by the pathogen. Here, we use a Vibrio cholerae luminescence reporter strain and liquid chromatography-tandem mass spectrometry to detect bacteria-produced and synthetic LAI-1. Ectopic production of LqsA in Escherichia coli generated LAI-1, which partitions to outer membrane vesicles (OMVs) and increases OMV size. These E. coli OMVs trigger luminescence of the V. cholerae reporter strain and inhibit the migration of Dictyostelium discoideum amoeba. Overexpression of lqsA in L.pneumophila under the control of strong stationary phase promoters (PflaA or P6SRNA), but not under control of its endogenous promoter (PlqsA), produces LAI-1, which is detected in purified OMVs. These L. pneumophila OMVs trigger luminescence of the Vibrio reporter strain and inhibit D. discoideum migration. L. pneumophila OMVs are smaller upon overexpression of lqsA or upon addition of LAI-1 to growing bacteria, and therefore, LqsA affects OMV production. The overexpression of lqsA but not a catalytically inactive mutant promotes intracellular replication of L. pneumophila in macrophages, indicating that intracellularly produced LA1-1 modulates the interaction in favor of the pathogen. Taken together, we provide evidence that L. pneumophila LAI-1 is secreted through OMVs and promotes interbacterial communication and interactions with eukaryotic host cells.
Collapse
Affiliation(s)
- Mingzhen Fan
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Paul Charki
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany
| | - Christian Hedberg
- Institute of Chemistry and Umeå Center for Microbial Research, Umeå University, Umeå, Sweden
| | - Jürgen Seibel
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany
| | | | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
39
|
Bitzenhofer NL, Classen T, Jaeger KE, Loeschcke A. Biotransformation Of l-Tryptophan To Produce Arcyriaflavin A With Pseudomonas putida KT2440. Chembiochem 2023; 24:e202300576. [PMID: 37743253 DOI: 10.1002/cbic.202300576] [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: 08/16/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 09/26/2023]
Abstract
Natural products such as indolocarbazoles are a valuable source of highly bioactive compounds with numerous potential applications in the pharmaceutical industry. Arcyriaflavin A, isolated from marine invertebrates and slime molds, is one representative of this group and acts as a cyclin D1-cyclin-dependent kinase 4 inhibitor. To date, access to this compound has mostly relied on multi-step total synthesis. In this study, biosynthetic access to arcyriaflavin A was explored using recombinant Pseudomonas putida KT2440 based on a previously generated producer strain. We used a Design of Experiment approach to analyze four key parameters, which led to the optimization of the bioprocess. By engineering the formation of outer membrane vesicles and using an adsorbent in the culture broth, we succeeded to increase the yield of arcyriaflavin A in the cell-free supernatant, resulting in a nearly eight-fold increase in the overall production titers. Finally, we managed to scale up the bioprocess leading to a final yield of 4.7 mg arcyriaflavin A product isolated from 1 L of bacterial culture. Thus, this study showcases an integrative approach to improve biotransformation and moreover also provides starting points for further optimization of indolocarbazole production in P. putida.
Collapse
Affiliation(s)
- Nora Lisa Bitzenhofer
- Institute of Molecular Enzyme Technology (IMET), Heinrich Heine University Düsseldorf located at Forschungszentrum Jülich, Stetternicher Forst, Building 15.8, 52426, Jülich, Germany
| | - Thomas Classen
- Institute of Bio- and Geosciences (IBG-1): Biotechnology, Forschungszentrum Jülich GmbH, Stetternicher Forst, Building 15.8, 52426, Jülich, Germany
| | - Karl-Erich Jaeger
- Institute of Molecular Enzyme Technology (IMET), Heinrich Heine University Düsseldorf located at Forschungszentrum Jülich, Stetternicher Forst, Building 15.8, 52426, Jülich, Germany
- Institute of Bio- and Geosciences (IBG-1): Biotechnology, Forschungszentrum Jülich GmbH, Stetternicher Forst, Building 15.8, 52426, Jülich, Germany
| | - Anita Loeschcke
- Institute of Molecular Enzyme Technology (IMET), Heinrich Heine University Düsseldorf located at Forschungszentrum Jülich, Stetternicher Forst, Building 15.8, 52426, Jülich, Germany
| |
Collapse
|
40
|
Torabian P, Singh N, Crawford J, Gonzalez G, Burgado N, Videva M, Miller A, Perdue J, Dinu M, Pietropaoli A, Gaborski T, Michel LV. The effect of clinically relevant beta-lactam, aminoglycoside, and quinolone antibiotics on bacterial extracellular vesicle release from E. coli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568081. [PMID: 38045295 PMCID: PMC10690228 DOI: 10.1101/2023.11.22.568081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Sepsis, a leading cause of death in hospitals, can be defined as a dysregulated host inflammatory response to infection, which can lead to tissue damage, organ failure, and cardiovascular complications. Although there is no cure for sepsis, the condition is typically managed with broad spectrum antibiotics to eliminate any potential bacterial source of infection. However, a potential side-effect of antibiotic treatment is the enhanced release of bacterial extracellular vesicles (BEVs). BEVs are membrane-bound nanoparticles produced by a variety of mechanisms, one of which includes the pinching-off of the outer membrane (in Gram-negative bacteria) to enclose proteins and other biological molecules for transport and intercellular communication. Some of the Gram-negative EV cargo, including Peptidoglycan associated lipoprotein (Pal) and Outer membrane protein A (OmpA), have been shown to induce both acute and chronic inflammation in host tissue. We hypothesize that antibiotic concentration and its mechanism of action can have an effect on the amount of released BEVs, which could potentially exacerbate the host inflammatory response. In this study, we evaluated nine clinically relevant antibiotics for their effect on EV release from Escherichia coli. EVs were characterized using immunoblotting, nanoparticle tracking analysis, and transmission electron microscopy. Several beta-lactam antibiotics caused significantly more EV release, while quinolone and aminoglycosides caused relatively less vesiculation. Further study is warranted to corroborate the correlation between an antibiotic's mechanism of action and its effect on EV release, but these results underline the importance of antibiotic choice when treating sepsis patients.
Collapse
Affiliation(s)
- Panteha Torabian
- Department of Biomedical Engineering, Rochester Institute of Technology
| | - Navraj Singh
- School of Chemistry and Materials Science, Rochester Institute of Technology
| | - James Crawford
- School of Chemistry and Materials Science, Rochester Institute of Technology
| | - Gabriela Gonzalez
- School of Chemistry and Materials Science, Rochester Institute of Technology
| | - Nicholas Burgado
- School of Chemistry and Materials Science, Rochester Institute of Technology
| | - Martina Videva
- School of Chemistry and Materials Science, Rochester Institute of Technology
| | - Aidan Miller
- School of Chemistry and Materials Science, Rochester Institute of Technology
| | - Janai Perdue
- School of Chemistry and Materials Science, Rochester Institute of Technology
| | - Milena Dinu
- School of Chemistry and Materials Science, Rochester Institute of Technology
| | - Anthony Pietropaoli
- Department of Medicine, Pulmonary Diseases and Critical Care, University of Rochester
| | - Thomas Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology
| | - Lea Vacca Michel
- School of Chemistry and Materials Science, Rochester Institute of Technology
| |
Collapse
|
41
|
Chen H, Ang CJ, Crowder MK, Brieher WM, Blanke SR. Revisiting bacterial cytolethal distending toxin structure and function. Front Cell Infect Microbiol 2023; 13:1289359. [PMID: 38035327 PMCID: PMC10682658 DOI: 10.3389/fcimb.2023.1289359] [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: 09/05/2023] [Accepted: 10/19/2023] [Indexed: 12/02/2023] Open
Abstract
Cytolethal distending toxins (CDTs) are intracellular-acting bacterial genotoxins generated by a diverse group of mucocutaneous human pathogens. CDTs must successfully bind to the plasma membrane of host cells in order to exert their modulatory effects. Maximal toxin activity requires all three toxin subunits, CdtA, CdtB, and CdtC, which, based primarily on high-resolution structural data, are believed to preassemble into a tripartite complex necessary for toxin activity. However, biologically active toxin has not been experimentally demonstrated to require assembly of the three subunits into a heterotrimer. Here, we experimentally compared concentration-dependent subunit interactions and toxin cellular activity of the Campylobacter jejuni CDT (Cj-CDT). Co-immunoprecipitation and dialysis retention experiments provided evidence for the presence of heterotrimeric toxin complexes, but only at concentrations of Cj-CdtA, Cj-CdtB, and Cj-CdtC several logs higher than required for Cj-CDT-mediated arrest of the host cell cycle at the G2/M interface, which is triggered by the endonuclease activity associated with the catalytic Cj-CdtB subunit. Microscale thermophoresis confirmed that Cj-CDT subunit interactions occur with low affinity. Collectively, our data suggest that at the lowest concentrations of toxin sufficient for arrest of cell cycle progression, mixtures of Cj-CdtA, Cj-CdtB, and Cj-CdtC consist primarily of non-interacting, subunit monomers. The lack of congruence between toxin tripartite structure and cellular activity suggests that the widely accepted model that CDTs principally intoxicate host cells as preassembled heterotrimeric structures should be revisited.
Collapse
Affiliation(s)
- Henry Chen
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Claire J. Ang
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Molly K. Crowder
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - William M. Brieher
- Department of Cellular and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Steven R. Blanke
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Biomedical and Translational Sciences Department, Carle-Illinois College of Medicine, University of Illinois, Urbana, IL, United States
| |
Collapse
|
42
|
Sun D, Chen P, Xi Y, Sheng J. From trash to treasure: the role of bacterial extracellular vesicles in gut health and disease. Front Immunol 2023; 14:1274295. [PMID: 37841244 PMCID: PMC10570811 DOI: 10.3389/fimmu.2023.1274295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
Bacterial extracellular vesicles (BEVs) have emerged as critical factors involved in gut health regulation, transcending their traditional roles as byproducts of bacterial metabolism. These vesicles function as cargo carriers and contribute to various aspects of intestinal homeostasis, including microbial balance, antimicrobial peptide secretion, physical barrier integrity, and immune system activation. Therefore, any imbalance in BEV production can cause several gut-related issues including intestinal infection, inflammatory bowel disease, metabolic dysregulation, and even cancer. BEVs derived from beneficial or commensal bacteria can act as potent immune regulators and have been implicated in maintaining gut health. They also show promise for future clinical applications in vaccine development and tumor immunotherapy. This review examines the multifaceted role of BEVs in gut health and disease, and also delves into future research directions and potential applications.
Collapse
Affiliation(s)
- Desen Sun
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Pan Chen
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Yang Xi
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Jinghao Sheng
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
43
|
Baldwin L, Jones EJ, Iles A, Carding SR, Pamme N, Dyer CE, Greenman J. Development of a dual-flow tissue perfusion device for modeling the gastrointestinal tract-brain axis. BIOMICROFLUIDICS 2023; 17:054104. [PMID: 37840538 PMCID: PMC10569815 DOI: 10.1063/5.0168953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023]
Abstract
Despite the large number of microfluidic devices that have been described over the past decade for the study of tissues and organs, few have become widely adopted. There are many reasons for this lack of adoption, primarily that devices are constructed for a single purpose or because they are highly complex and require relatively expensive investment in facilities and training. Here, we describe a microphysiological system (MPS) that is simple to use and provides fluid channels above and below cells, or tissue biopsies, maintained on a disposable, poly(methyl methacrylate), carrier held between polycarbonate outer plates. All other fittings are standard Luer sizes for ease of adoption. The carrier can be coated with cells on both sides to generate membrane barriers, and the devices can be established in series to allow medium to flow from one cell layer to another. Furthermore, the carrier containing cells can be easily removed after treatment on the device and the cells can be visualized or recovered for additional off-chip analysis. A 0.4 μm membrane with cell monolayers proved most effective in maintaining separate fluid flows, allowing apical and basal surfaces to be perfused independently. A panel of different cell lines (Caco-2, HT29-MTX-E12, SH-SY5Y, and HUVEC) were successfully maintained in the MPS for up to 7 days, either alone or on devices connected in series. The presence of tight junctions and mucin was expressed as expected by Caco-2 and HT-29-MTX-E12, with Concanavalin A showing uniform staining. Addition of Annexin V and PI showed viability of these cells to be >80% at 7 days. Bacterial extracellular vesicles (BEVs) produced by Bacteroides thetaiotaomicron and labeled with 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbo-cyanine perchlorate (DiD) were used as a model component of the human colonic microbiota and were visualized translocating from an apical surface containing Caco-2 cells to differentiated SH-SY5Y neuronal cells cultured on the basal surface of connected devices. The newly described MPS can be easily adapted, by changing the carrier to maintain spheroids, pieces, or slices of biopsy tissue and joined in series to study a variety of cell and tissue processes. The cell layers can be made more complex through the addition of multiple cell types and/or different patterning of extracellular matrix and the ability to culture cells adjacent to one another to allow study of cell:cell transfer, e.g., passive or active drug transfer, virus or bacterial entry or BEV uptake and transfer.
Collapse
Affiliation(s)
- Lydia Baldwin
- Centre of Biomedical Sciences, Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, United Kingdom
| | - Emily J. Jones
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
| | - Alexander Iles
- Department of Materials and Environmental Chemistry, Stockholm University, 106 91 Stockholm, Sweden
| | | | - Nicole Pamme
- Department of Materials and Environmental Chemistry, Stockholm University, 106 91 Stockholm, Sweden
| | - Charlotte E. Dyer
- Centre of Biomedical Sciences, Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, United Kingdom
| | - John Greenman
- Centre of Biomedical Sciences, Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, United Kingdom
| |
Collapse
|
44
|
Gan Y, Zhao G, Wang Z, Zhang X, Wu MX, Lu M. Bacterial Membrane Vesicles: Physiological Roles, Infection Immunology, and Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301357. [PMID: 37357142 PMCID: PMC10477901 DOI: 10.1002/advs.202301357] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/19/2023] [Indexed: 06/27/2023]
Abstract
Bacterial or fungal membrane vesicles, traditionally considered as microbial metabolic wastes, are secreted mainly from the outer membrane or cell membrane of microorganisms. However, recent studies have shown that these vesicles play essential roles in direct or indirect communications among microorganisms and between microorganisms and hosts. This review aims to provide an updated understanding of the physiological functions and emerging applications of bacterial membrane vesicles, with a focus on their biogenesis, mechanisms of adsorption and invasion into host cells, immune stimulatory effects, and roles in the much-concerned problem of bacterial resistance. Additionally, the potential applications of these vesicles as biomarkers, vaccine candidates, and drug delivery platforms are discussed.
Collapse
Affiliation(s)
- Yixiao Gan
- Department of Transfusion MedicineHuashan HospitalFudan UniversityShanghai200040P. R. China
| | - Gang Zhao
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200240P. R. China
| | - Zhicheng Wang
- Department of Transfusion MedicineHuashan HospitalFudan UniversityShanghai200040P. R. China
| | - Xingcai Zhang
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
| | - Mei X. Wu
- Wellman Center for PhotomedicineMassachusetts General HospitalDepartment of DermatologyHarvard Medical School, 50 Blossom StreetBostonMA02114USA
| | - Min Lu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200240P. R. China
| |
Collapse
|
45
|
Modasia AA, Jones EJ, Martel LM, Louvel H, Couraud P, Blackshaw LA, Carding SR. The use of a multicellular in vitro model to investigate uptake and migration of bacterial extracellular vesicles derived from the human gut commensal Bacteroides thetaiotaomicron. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e93. [PMID: 38939073 PMCID: PMC11080816 DOI: 10.1002/jex2.93] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 06/29/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are increasingly seen as key signalling mediators between the gut microbiota and the host. Recent studies have provided evidence of BEVs ability to transmigrate across cellular barriers to elicit responses in other tissues, such as the central nervous system (CNS). Here we use a combination of single-, two- and three-cell culture systems to demonstrate the transmigration of Bacteroides thetaiotaomicron derived BEVs (Bt-BEVs) across gut epithelium and blood brain barrier (BBB) endothelium, and their subsequent acquisition and downstream effects in neuronal cells. Bt-BEVs were shown to traffic to the CNS in vivo after intravenous administration to mice, and in multi-cell in vitro culture systems to transmigrate across gut epithelial and BBB endothelial cell barriers, where they were acquired by both microglia and immature neuronal cells. No significant activation/inflammatory effects were induced in non-differentiated neurons, in contrast to that observed in microglia cells, although this was notably less than that induced by lipopolysaccharide (LPS). Overall, our findings provide evidence for transmigration of Bt-BEVs across gut-epithelial and BBB endothelial cell barriers in vivo and in vitro, and their downstream responses in neural cells. This study sheds light onto how commensal bacteria-derived BEV transport across the gut-brain axis and can be exploited for the development of targeted drug delivery.
Collapse
Affiliation(s)
- Amisha A. Modasia
- Quadram Institute BioscienceRosalind Franklin RoadNorwich Research ParkNorwichUK
| | - Emily J. Jones
- Quadram Institute BioscienceRosalind Franklin RoadNorwich Research ParkNorwichUK
| | | | - Hélène Louvel
- National Institute of Health and Medical Research (INSERM)6 PlaceTristan BernardParisFrance
| | - Pierre‐Olivier Couraud
- National Institute of Health and Medical Research (INSERM)6 PlaceTristan BernardParisFrance
| | - L. Ashley Blackshaw
- Quadram Institute BioscienceRosalind Franklin RoadNorwich Research ParkNorwichUK
| | - Simon R. Carding
- Quadram Institute BioscienceRosalind Franklin RoadNorwich Research ParkNorwichUK
- Norwich Medical SchoolNorwich Research ParkUniversity of East AngliaNorwichUK
| |
Collapse
|
46
|
Olivo-Martínez Y, Bosch M, Badia J, Baldomà L. Modulation of the Intestinal Barrier Integrity and Repair by Microbiota Extracellular Vesicles through the Differential Regulation of Trefoil Factor 3 in LS174T Goblet Cells. Nutrients 2023; 15:nu15112437. [PMID: 37299399 DOI: 10.3390/nu15112437] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Trefoil factor 3 (TFF3) plays a key role in the maintenance and repair of intestinal mucosa. TFF3 expression is upregulated by the microbiota through TLR2. At the posttranscriptional level, TFF3 is downregulated by miR-7-5p. Reduced TFF3 levels have been detected in the damaged tissue of IBD patients. Here, we investigate the regulation of TFF3 expression by microbiota extracellular vesicles (EVs) in LS174T goblet cells using RT-qPCR and inhibitors of the TLR2 or PI3K pathways. To evaluate the subsequent impact on epithelial barrier function, conditioned media from control and vesicle-stimulated LS174T cells were used to treat Caco-2 monolayers. The barrier-strengthening effects were evaluated by analysing the expression and subcellular distribution of tight junction proteins, and the repairing effects were assessed using wound-healing assays. The results showed a differential regulation of TFF3 in LS174T via EVs from the probiotic EcN and the commensal ECOR12. EcN EVs activated the TFF3 production through TLR2 and downregulated miR7-5-p through PI3K. Consistently, high levels of secreted TFF3 reinforced the tight junctions and stimulated wound healing in the Caco-2 cells. ECOR12 EVs did not cause these effects. TFF3 is a potential therapeutic target in IBD. This study contributes to understanding the molecular players (microbiota EVs) connecting gut microbes to health and may help in designing better nutritional interventions based on microbiota bioactive compounds.
Collapse
Affiliation(s)
- Yenifer Olivo-Martínez
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| | - Manel Bosch
- Unitat de Microscòpia Òptica Avançada, Centres Científics i Tecnològics, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Josefa Badia
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| | - Laura Baldomà
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Barcelona, Spain
| |
Collapse
|
47
|
Henriquez T, Falciani C. Extracellular Vesicles of Pseudomonas: Friends and Foes. Antibiotics (Basel) 2023; 12:antibiotics12040703. [PMID: 37107065 PMCID: PMC10135156 DOI: 10.3390/antibiotics12040703] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/31/2023] [Accepted: 04/02/2023] [Indexed: 04/07/2023] Open
Abstract
Extracellular vesicles (Evs) are small spherical vesicles capable of transporting molecules (such as proteins, nucleic acids and lipids) from one cell to another. They have been implicated in processes such as cell-to-cell communication, pathogenicity, biofilm formation and metabolism. In parallel, Evs have been proposed as interesting biotechnological tools. In recent years, antibiotic resistance has become a major problem for human health worldwide. A pathogen singled out as among the most lethal antibiotic-resistant organisms is Pseudomonas aeruginosa, an important Gram-negative bacterium that has been extensively studied for the production and characterization of Evs. Here, we describe the advances made in the last decade regarding understanding of the role of Evs in the pathogenicity of Pseudomonas. We also examine the potential of Evs for the development of new treatment strategies.
Collapse
Affiliation(s)
- Tania Henriquez
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Chiara Falciani
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| |
Collapse
|
48
|
Huynh DT, Jong WSP, Oudejans MAH, van den Berg van Saparoea HB, Luirink J, van Ulsen P. Heterologous Display of Chlamydia trachomatis PmpD Passenger at the Surface of Salmonella OMVs. MEMBRANES 2023; 13:366. [PMID: 37103793 PMCID: PMC10145130 DOI: 10.3390/membranes13040366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 06/19/2023]
Abstract
Chlamydia trachomatis is the bacterial pathogen that causes most cases of sexually transmitted diseases annually. To combat the global spread of asymptomatic infection, development of effective (mucosal) vaccines that offer both systemic and local immune responses is considered a high priority. In this study, we explored the expression of C. trachomatis full-length (FL) PmpD, as well as truncated PmpD passenger constructs fused to a "display" autotransporter (AT) hemoglobin protease (HbpD) and studied their inclusion into outer membrane vesicles (OMVs) of Escherichia coli and Salmonella Typhimurium. OMVs are considered safe vaccine vectors well-suited for mucosal delivery. By using E. coli AT HbpD-fusions of chimeric constructs we improved surface display and successfully generated Salmonella OMVs decorated with a secreted and immunogenic PmpD passenger fragment (aa68-629) to 13% of the total protein content. Next, we investigated whether a similar chimeric surface display strategy could be applied to other AT antigens, i.e., secreted fragments of Prn (aa35-350) of Bordetella pertussis and VacA (aa65-377) of Helicobacter pylori. The data provided information on the complexity of heterologous expression of AT antigens at the OMV surface and suggested that optimal expression strategies should be developed on an antigen-to-antigen basis.
Collapse
Affiliation(s)
- Dung T. Huynh
- Abera Bioscience AB, 750 26 Uppsala, Sweden
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | | | - Manon A. H. Oudejans
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | | | - Joen Luirink
- Abera Bioscience AB, 750 26 Uppsala, Sweden
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - Peter van Ulsen
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
49
|
Fonseca S, Carvalho AL, Miquel-Clopés A, Jones EJ, Juodeikis R, Stentz R, Carding SR. Extracellular vesicles produced by the human gut commensal bacterium Bacteroides thetaiotaomicron elicit anti-inflammatory responses from innate immune cells. Front Microbiol 2022; 13:1050271. [PMID: 36439842 PMCID: PMC9684339 DOI: 10.3389/fmicb.2022.1050271] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/25/2022] [Indexed: 07/24/2023] Open
Abstract
Bacterial extracellular vesicles (BEVs) produced by gut commensal bacteria have been proposed to play an important role in maintaining host homeostasis via interactions with the immune system. Details of the mediators and pathways of BEV-immune cell interactions are however incomplete. In this study, we provide evidence for the anti-inflammatory and immunomodulatory properties of extracellular vesicles produced by the prominent human gut commensal bacterium Bacteroides thetaiotaomicron (Bt BEVs) and identify the molecular mechanisms underlying their interaction with innate immune cells. Administration of Bt BEVs to mice treated with colitis-inducing dextran sodium sulfate (DSS) ameliorates the symptoms of intestinal inflammation, improving survival rate and reducing weight loss and disease activity index scores, in association with upregulation of IL-10 production in colonic tissue and in splenocytes. Pre-treatment (conditioning) of murine bone marrow derived monocytes (BMDM) with Bt BEVs resulted in higher ratio of IL-10/TNFα production after an LPS challenge when compared to LPS pre-conditioned or non-conditioned BMDM. Using the THP-1 monocytic cell line the interactions between Bt BEVs and monocytes/macrophages were shown to be mediated primarily by TLR2. Histone (H3K4me1) methylation analysis showed that Bt BEVs induced epigenetic reprogramming which persisted after infectious challenge, as revealed by increased levels of H3K4me1 in Bt BEV-conditioned LPS-challenged BMDM. Collectively, our findings highlight the important role of Bt BEVs in maintaining host immune homeostasis and raise the promising possibility of considering their use in immune therapies.
Collapse
Affiliation(s)
- Sonia Fonseca
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Ana L. Carvalho
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, United Kingdom
- Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | - Emily J. Jones
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Rokas Juodeikis
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Régis Stentz
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Simon R. Carding
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|