1
|
Lin Y, Wang J, Bu F, Zhang R, Wang J, Wang Y, Huang M, Huang Y, Zheng L, Wang Q, Hu X. Bacterial extracellular vesicles in the initiation, progression and treatment of atherosclerosis. Gut Microbes 2025; 17:2452229. [PMID: 39840620 DOI: 10.1080/19490976.2025.2452229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. However, current anti-atherosclerosis drugs have shown conflicting therapeutic outcomes, thereby spurring the search for novel and effective treatments. Recent research indicates the crucial involvement of oral and gastrointestinal microbiota in atherosclerosis. While gut microbiota metabolites, such as choline derivatives, have been extensively studied and reviewed, emerging evidence suggests that bacterial extracellular vesicles (BEVs), which are membrane-derived lipid bilayers secreted by bacteria, also play a significant role in this process. However, the role of BEVs in host-microbiota interactions remains insufficiently explored. This review aims to elucidate the complex communication mediated by BEVs along the gut-heart axis. In this review, we summarize current knowledge on BEVs, with a specific focus on how pathogen-derived BEVs contribute to the promotion of atherosclerosis, as well as how BEVs from gut symbionts and probiotics may mitigate its progression. We also explore the potential and challenges associated with engineered BEVs in the prevention and treatment of atherosclerosis. Finally, we discuss the benefits and challenges of using BEVs in atherosclerosis diagnosis and treatment, and propose future research directions to address these issues.
Collapse
Affiliation(s)
- Yuling Lin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Bu
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China
| | - Ruyi Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junhui Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yubing Wang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiumei Hu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Moghaddam ZS, Dehghan A, Halimi S, Najafi F, Nokhostin A, Naeini AE, Akbarzadeh I, Ren Q. Bacterial Extracellular Vesicles: Bridging Pathogen Biology and Therapeutic Innovation. Acta Biomater 2025:S1742-7061(25)00352-6. [PMID: 40349898 DOI: 10.1016/j.actbio.2025.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/14/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
The main role of bacterial extracellular vesicles (BEVs) has been associated with various processes such as intercellular communication and host-pathogen interactions. This comprehensive review explores the multifaceted functions of BEVs across different biological domains, emphasizing their multifaceted functions as contributors both to disease and as carriers of therapeutic possibilities. We examine the intricate interactions of BEVs within bacterial communities and between bacteria and hosts, their involvement in disease development through cargo delivery mechanisms, and their beneficial impact to microbial ecology. The review places a strong emphasis on BEVs' applications in biomedical sciences, where they are revolutionizing vaccine development, targeted drug delivery, and cancer therapy. By utilizing the inherent properties of BEVs for controlled drug release, targeted antigen delivery, and immune modulation, they offer a promising frontier in precision medicine. In addition, the diagnostic potential of BEVs is explored through their biomarker capabilities, providing valuable insights into disease states and treatment efficacy. Looking forward, this review underscores the challenges and opportunities in translating BEV research to clinical practice, promoting the use of standardized methods in BEV characterization and scaling up production. The diverse abilities of BEVs, ranging from contributing to pathogen virulence to driving therapeutic innovation, highlight their potential as a cornerstone in the future of biomedical advancements. STATEMENT OF SIGNIFICANCE: Bacterial extracellular vesicles (BEVs) are emerging as pivotal players in both pathogenesis and therapeutic innovation. This review explores their dual nature as agents of disease and as promising biomaterials in biomedical applications, and provides a comprehensive survey on their involvement in disease mechanisms and microbial ecology, and their potential in biomedical applications such as vaccine development, targeted drug delivery, cancer therapy and diagnosis. It highlights the complex interactions of BEVs within bacterial communities and between bacteria and hosts. This review also addresses current advancements, challenges, and opportunities in translating BEV research into clinical practice. The insights presented here position BEVs as a cornerstone in the future of biomedical advancements, advocating for standardized methods in BEV characterization and scalable production techniques.
Collapse
Affiliation(s)
| | - Ashkan Dehghan
- W Booth School of Engineering Practice and Technology Faculty of Engineering, McMaster University Hamilton, ON, Canada, L8S 0A3
| | - Saba Halimi
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Fatemeh Najafi
- Department of Chemical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802-1503, United States
| | - Ali Nokhostin
- Medical Sciences & Technologies Faculty, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | | | - Iman Akbarzadeh
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| | - Qun Ren
- Laboratory for Biointerfaces, Empa Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland.
| |
Collapse
|
3
|
Ho MY, Liu S, Xing B. Bacteria extracellular vesicle as nanopharmaceuticals for versatile biomedical potential. NANO CONVERGENCE 2024; 11:28. [PMID: 38990415 PMCID: PMC11239649 DOI: 10.1186/s40580-024-00434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/20/2024] [Indexed: 07/12/2024]
Abstract
Bacteria extracellular vesicles (BEVs), characterized as the lipid bilayer membrane-surrounded nanoparticles filled with molecular cargo from parent cells, play fundamental roles in the bacteria growth and pathogenesis, as well as facilitating essential interaction between bacteria and host systems. Notably, benefiting from their unique biological functions, BEVs hold great promise as novel nanopharmaceuticals for diverse biomedical potential, attracting significant interest from both industry and academia. Typically, BEVs are evaluated as promising drug delivery platforms, on account of their intrinsic cell-targeting capability, ease of versatile cargo engineering, and capability to penetrate physiological barriers. Moreover, attributing to considerable intrinsic immunogenicity, BEVs are able to interact with the host immune system to boost immunotherapy as the novel nanovaccine against a wide range of diseases. Towards these significant directions, in this review, we elucidate the nature of BEVs and their role in activating host immune response for a better understanding of BEV-based nanopharmaceuticals' development. Additionally, we also systematically summarize recent advances in BEVs for achieving the target delivery of genetic material, therapeutic agents, and functional materials. Furthermore, vaccination strategies using BEVs are carefully covered, illustrating their flexible therapeutic potential in combating bacterial infections, viral infections, and cancer. Finally, the current hurdles and further outlook of these BEV-based nanopharmaceuticals will also be provided.
Collapse
Affiliation(s)
- Ming Yao Ho
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, S637371, Singapore
| | - Songhan Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, S637371, Singapore
| | - Bengang Xing
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, S637371, Singapore.
| |
Collapse
|
4
|
Fan YL, Jin JX, Zhu J, Ruan HB, Huang JQ. Extracellular vesicles of Bifidobacterium longum reverse the acquired carboplatin resistance in ovarian cancer cells via p53 phosphorylation on Ser15. Kaohsiung J Med Sci 2024; 40:530-541. [PMID: 38647095 DOI: 10.1002/kjm2.12837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
We previously found that the relative abundance of Bifidobacterium was increased after chemotherapy; however, the role of Bifidobacterium longum in chemotherapeutic drug resistance in ovarian cancer (OVC) remains unclear. This study aimed to understand the potential effects and mechanism of B. longum extracellular vesicles (B. longum-EVs) on carboplatin (CBP) resistance in OVC. Eight normal and 11 ovarian tissues were collected and the expression of B. longum genomic DNA and its association with acquired CBP resistance in OVC patients was determined. After isolating EVs by ultracentrifugation from B. longum (ATCC 15707), CBP-resistant A2780 cells were treated with PBS, CBP, B. longum-EVs, or CBP + B. longum-EVs, and subsequently analyzed by CCK-8, Edu staining, Annexin V/PI double staining, wound healing, and Transwell assays to detect cell viability, proliferation, apoptosis, migration, and invasion, respectively. MRP1, ATP7A, ATP7B, and p53 expression as well as p53 phosphorylation were measured by western blot analysis. S15A mutation of p53 was assessed to examine the potential role of p53 Ser15 phosphorylation in CBP-resistant OVC. B. longum levels were elevated and positively associated with CBP resistance in OVC patients. Only high concentrations of B. longum-EVs attenuated A2780 cell proliferation, apoptosis, migration, and invasion. B. longum-EVs exposure significantly enhanced the sensitivity of CBP-resistant A2780 cells to CBP and decreased the expression of drug resistance-related proteins. The effect of B. longum-EVs on reversing CBP resistance was completely inhibited by S15A mutation of p53. B. longum-EVs enhanced the sensitivity of OVC cells to CBP through p53 phosphorylation on Ser15.
Collapse
Affiliation(s)
- Yun-Long Fan
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Jia-Xi Jin
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Jun Zhu
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Hai-Bo Ruan
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Jin-Qun Huang
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| |
Collapse
|
5
|
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
|
6
|
Yu MSC, Chiang DM, Reithmair M, Meidert A, Brandes F, Schelling G, Ludwig C, Meng C, Kirchner B, Zenner C, Muller L, Pfaffl MW. The proteome of bacterial membrane vesicles in Escherichia coli-a time course comparison study in two different media. Front Microbiol 2024; 15:1361270. [PMID: 38510998 PMCID: PMC10954253 DOI: 10.3389/fmicb.2024.1361270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction Bacteria inhabit the in- and outside of the human body, such as skin, gut or the oral cavity where they play an innoxious, beneficial or even pathogenic role. It is well known that bacteria can secrete membrane vesicles (MVs) like eukaryotic cells with extracellular vesicles (EVs). Several studies indicate that bacterial membrane vesicles (bMVs) play a crucial role in microbiome-host interactions. However, the composition of such bMVs and their functionality under different culture conditions are still largely unknown. Methods To gain a better insight into bMVs, we investigated the composition and functionality of E. coli (DSM 105380) bMVs from the culture media Lysogeny broth (LB) and RPMI 1640 throughout the different phases of growth (lag-, log- and stationary-phase). bMVs from three time points (8 h, 54 h, and 168 h) and two media (LB and RPMI 1640) were isolated by ultracentrifugation and analyzed using nanoparticle tracking analysis (NTA), cryogenic electron microscopy (Cryo-EM), conventional transmission electron microscopy (TEM) and mass spectrometry-based proteomics (LC-MS/MS). Furthermore, we examined pro-inflammatory cytokines IL-1β and IL-8 in the human monocyte cell line THP-1 upon bMV treatment. Results Particle numbers increased with inoculation periods. The bMV morphologies in Cryo-EM/TEM were similar at each time point and condition. Using proteomics, we identified 140 proteins, such as the common bMV markers OmpA and GroEL, present in bMVs isolated from both media and at all time points. Additionally, we were able to detect growth-condition-specific proteins. Treatment of THP-1 cells with bMVs of all six groups lead to significantly high IL-1β and IL-8 expressions. Conclusion Our study showed that the choice of medium and the duration of culturing significantly influence both E. coli bMV numbers and protein composition. Our TEM/Cryo-EM results demonstrated the presence of intact E. coli bMVs. Common E. coli proteins, including OmpA, GroEL, and ribosome proteins, can consistently be identified across all six tested growth conditions. Furthermore, our functional assays imply that bMVs isolated from the six groups retain their function and result in comparable cytokine induction.
Collapse
Affiliation(s)
- Mia S. C. Yu
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich (TUM), Freising, Germany
| | - Dapi Menglin Chiang
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich (TUM), Freising, Germany
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marlene Reithmair
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany
| | - Agnes Meidert
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Florian Brandes
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Gustav Schelling
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Christina Ludwig
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich (TUM), Freising, Germany
| | - Chen Meng
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich (TUM), Freising, Germany
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich (TUM), Freising, Germany
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany
| | - Christian Zenner
- Intestinal Microbiome, ZIEL – Institute for Food & Health, School of Life Sciences, Technical University of Munich (TUM), Freising, Germany
| | - Laurent Muller
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital of Basel, Basel, Switzerland
| | - Michael W. Pfaffl
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich (TUM), Freising, Germany
| |
Collapse
|
7
|
Liu BD, Akbar R, Oliverio A, Thapa K, Wang X, Fan GC. BACTERIAL EXTRACELLULAR VESICLES IN THE REGULATION OF INFLAMMATORY RESPONSE AND HOST-MICROBE INTERACTIONS. Shock 2024; 61:175-188. [PMID: 37878470 PMCID: PMC10921997 DOI: 10.1097/shk.0000000000002252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
ABSTRACT Extracellular vesicles (EVs) are a new revelation in cross-kingdom communication, with increasing evidence showing the diverse roles of bacterial EVs (BEVs) in mammalian cells and host-microbe interactions. Bacterial EVs include outer membrane vesicles released by gram-negative bacteria and membrane vesicles generated from gram-positive bacteria. Recently, BEVs have drawn attention for their potential as biomarkers and therapeutic tools because they are nano-sized and can deliver bacterial cargo into host cells. Importantly, exposure to BEVs significantly affects various physiological and pathological responses in mammalian cells. Herein, we provide a comprehensive overview of the various effects of BEVs on host cells (i.e., immune cells, endothelial cells, and epithelial cells) and inflammatory/infectious diseases. First, the biogenesis and purification methods of BEVs are summarized. Next, the mechanisms and pathways identified by BEVs that stimulate either proinflammatory or anti-inflammatory responses are highlighted. In addition, we discuss the mechanisms by which BEVs regulate host-microbe interactions and their effects on the immune system. Finally, this review focuses on the contribution of BEVs to the pathogenesis of sepsis/septic shock and their therapeutic potential for the treatment of sepsis.
Collapse
Affiliation(s)
- Benjamin D. Liu
- Department of Chemistry and Biochemistry, The Ohio State University College of Arts and Sciences, Columbus, OH, 43210, USA
| | - Rubab Akbar
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Anna Oliverio
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kajol Thapa
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xiaohong Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
8
|
Tammisto H, Karvonen K. Purification of Borrelia burgdorferi Outer Membrane Vesicles. Methods Mol Biol 2024; 2742:37-45. [PMID: 38165613 DOI: 10.1007/978-1-0716-3561-2_3] [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: 01/04/2024]
Abstract
Bacterial outer membrane vesicles (OMVs) are spherical membrane constructs shed by gram-negative bacteria. OMVs produced by the Lyme disease pathogen Borrelia burgdorferi have been identified to contain such virulence factors as OspA, OspB, OspC, and genetic material. However, the function and possible pathogenicity of borrelial OMVs are still undetermined. Therefore, further research on borrelial OMVs is required, and for that a standard method for OMV purification is necessary. Here we describe a successful and reproducible purification of borrelial outer membrane vesicles using concentration, filtration, and ultracentrifugation steps.
Collapse
Affiliation(s)
- Hanna Tammisto
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Kati Karvonen
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland.
| |
Collapse
|
9
|
Kretschmer M, Müller J, Henke P, Otto V, Rodriguez AA, Müsken M, Jahn D, Borrero-de Acuña JM, Neumann-Schaal M, Wegner A. Isolation and Quantification of Bacterial Membrane Vesicles for Quantitative Metabolic Studies Using Mammalian Cell Cultures. Cells 2023; 12:2674. [PMID: 38067103 PMCID: PMC10705164 DOI: 10.3390/cells12232674] [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: 09/25/2023] [Revised: 11/10/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Bacterial membrane vesicles (BMVs) are produced by most bacteria and participate in various cellular processes, such as intercellular communication, nutrient exchange, and pathogenesis. Notably, these vesicles can contain virulence factors, including toxic proteins, DNA, and RNA. Such factors can contribute to the harmful effects of bacterial pathogens on host cells and tissues. Although the general effects of BMVs on host cellular physiology are well known, the underlying molecular mechanisms are less understood. In this study, we introduce a vesicle quantification method, leveraging the membrane dye FM4-64. We utilize a linear regression model to analyze the fluorescence emitted by stained vesicle membranes to ensure consistent and reproducible vesicle-host interaction studies using cultured cells. This method is particularly valuable for identifying host cellular processes impacted by vesicles and their specific cargo. Moreover, it outcompetes unreliable protein concentration-based methods. We (1) show a linear correlation between the number of vesicles and the fluorescence signal emitted from the FM4-64 dye; (2) introduce the "vesicle load" as a new semi-quantitative unit, facilitating more reproducible vesicle-cell culture interaction experiments; (3) show that a stable vesicle load yields consistent host responses when studying vesicles from Pseudomonas aeruginosa mutants; (4) demonstrate that typical vesicle isolation contaminants, such as flagella, do not significantly skew the metabolic response of lung epithelial cells to P. aeruginosa vesicles; and (5) identify inositol monophosphatase 1 (SuhB) as a pivotal regulator in the vesicle-mediated pathogenesis of P. aeruginosa.
Collapse
Affiliation(s)
- Marcel Kretschmer
- Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
- Department of Bioinformatics and Biochemistry, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Julia Müller
- Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
- Department of Bioinformatics and Biochemistry, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Petra Henke
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Inhoffenstraße 7 B, 38124 Braunschweig, Germany
| | - Viktoria Otto
- Institute for Microbiology, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | | | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Dieter Jahn
- Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
- Institute for Microbiology, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - José Manuel Borrero-de Acuña
- Department of Microbiology, Facultad de Biología, University of Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| | - Meina Neumann-Schaal
- Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Inhoffenstraße 7 B, 38124 Braunschweig, Germany
| | - Andre Wegner
- Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
- Department of Bioinformatics and Biochemistry, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| |
Collapse
|
10
|
Thapa HB, Ebenberger SP, Schild S. The Two Faces of Bacterial Membrane Vesicles: Pathophysiological Roles and Therapeutic Opportunities. Antibiotics (Basel) 2023; 12:1045. [PMID: 37370364 PMCID: PMC10295235 DOI: 10.3390/antibiotics12061045] [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: 05/30/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Bacterial membrane vesicles (MVs) are nanosized lipid particles secreted by lysis or blebbing mechanisms from Gram-negative and -positive bacteria. It is becoming increasingly evident that MVs can promote antimicrobial resistance but also provide versatile opportunities for therapeutic exploitation. As non-living facsimiles of parent bacteria, MVs can carry multiple bioactive molecules such as proteins, lipids, nucleic acids, and metabolites, which enable them to participate in intra- and interspecific communication. Although energetically costly, the release of MVs seems beneficial for bacterial fitness, especially for pathogens. In this review, we briefly discuss the current understanding of diverse MV biogenesis routes affecting MV cargo. We comprehensively highlight the physiological functions of MVs derived from human pathogens covering in vivo adaptation, colonization fitness, and effector delivery. Emphasis is given to recent findings suggesting a vicious cycle of MV biogenesis, pathophysiological function, and antibiotic therapy. We also summarize potential therapeutical applications, such as immunotherapy, vaccination, targeted delivery, and antimicrobial potency, including their experimental validation. This comparative overview identifies common and unique strategies for MV modification used along diverse applications. Thus, the review summarizes timely aspects of MV biology in a so far unprecedented combination ranging from beneficial function for bacterial pathogen survival to future medical applications.
Collapse
Affiliation(s)
- Himadri B. Thapa
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Stephan P. Ebenberger
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Stefan Schild
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
- BioTechMed Graz, 8010 Graz, Austria
- Field of Excellence Biohealth, University of Graz, 8010 Graz, Austria
| |
Collapse
|
11
|
Tian CM, Yang MF, Xu HM, Zhu MZ, Zhang Y, Yao J, Wang LS, Liang YJ, Li DF. Emerging role of bacterial outer membrane vesicle in gastrointestinal tract. Gut Pathog 2023; 15:20. [PMID: 37106359 PMCID: PMC10133921 DOI: 10.1186/s13099-023-00543-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
Bacteria form a highly complex ecosystem in the gastrointestinal (GI) tract. In recent years, mounting evidence has shown that bacteria can release nanoscale phospholipid bilayer particles that encapsulate nucleic acids, proteins, lipids, and other molecules. Extracellular vesicles (EVs) are secreted by microorganisms and can transport a variety of important factors, such as virulence factors, antibiotics, HGT, and defensive factors produced by host eukaryotic cells. In addition, these EVs are vital in facilitating communication between microbiota and the host. Therefore, bacterial EVs play a crucial role in maintaining the GI tract's health and proper functioning. In this review, we outlined the structure and composition of bacterial EVs. Additionally, we highlighted the critical role that bacterial EVs play in immune regulation and in maintaining the balance of the gut microbiota. To further elucidate progress in the field of intestinal research and to provide a reference for future EV studies, we also discussed the clinical and pharmacological potential of bacterial EVs, as well as the necessary efforts required to understand the mechanisms of interaction between bacterial EVs and gut pathogenesis.
Collapse
Affiliation(s)
- Cheng-Mei Tian
- Department of Emergency, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen, Guangdong, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), No.1017, Dongmen North Road, Luohu District, Shenzhen, 518020, People's Republic of China.
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), No.1017, Dongmen North Road, Luohu District, Shenzhen, 518020, People's Republic of China.
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, No.1080, Cuizu Road, Luohu District, Shenzhen, 518020, People's Republic of China.
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), No.1017, Dongmen North Road, Luohu District, Shenzhen, 518020, People's Republic of China.
| |
Collapse
|
12
|
Chen Y, Zhu M, Huang B, Jiang Y, Su J. Advances in cell membrane-coated nanoparticles and their applications for bone therapy. BIOMATERIALS ADVANCES 2023; 144:213232. [PMID: 36502750 DOI: 10.1016/j.bioadv.2022.213232] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
Due to the specific structure of natural bone, most of the therapeutics are incapable to be delivered into the targeted site with effective concentrations. Nanotechnology has provided a good way to improve this issue, cell membrane mimetic nanoparticles (NPs) have been emerging as an ideal nanomaterial which integrates the advantages of natural cell membranes with synthetic NPs to significantly improve the biocompatibility as well as achieving long-lasting circulation and targeted delivery. In addition, functionalized modifications of the cell membrane facilitate more precise targeting and therapy. Here, an overview of the preparation of cell membrane-coated NPs and the properties of cell membranes from different cell sources has been given to expatiate their function and potential applications. Strategies for functionalized modification of cell membranes are also briefly described. The application of cell membrane-coated NPs for bone therapy is then presented according to the function of cell membranes. Moreover, the prospects and challenges of cell membrane-coated NPs for translational medicine have also been discussed.
Collapse
Affiliation(s)
- Yutong Chen
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China; School of Medicine, Shanghai University, Shanghai 200444, PR China; School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Mengru Zhu
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China; School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Biaotong Huang
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China; Wenzhou Institute of Shanghai University, Wenzhou 325000, PR China.
| | - Yingying Jiang
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China.
| | - Jiacan Su
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
13
|
Castillo-Romero KF, Santacruz A, González-Valdez J. Production and purification of bacterial membrane vesicles for biotechnology applications: Challenges and opportunities. Electrophoresis 2023; 44:107-124. [PMID: 36398478 DOI: 10.1002/elps.202200133] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/17/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022]
Abstract
Bacterial membrane vesicles (BMVs) are bi-layered nanostructures derived from Gram-negative and Gram-positive bacteria. Among other pathophysiological roles, BMVs are critical messengers in intercellular communication. As a result, BMVs are emerging as a promising technology for the development of numerous therapeutic applications. Despite the remarkable progress in unveiling BMV biology and functions in recent years, their successful isolation and purification have been limited. Several challenges related to vesicle purity, yield, and scalability severely hamper the further development of BMVs for biotechnology and clinical applications. This review focuses on the current technologies and methodologies used in BMV production and purification, such as ultracentrifugation, density-gradient centrifugation, size-exclusion chromatography, ultrafiltration, and precipitation. We also discuss the current challenges related to BMV isolation, large-scale production, storage, and stability that limit their application. More importantly, the present work explains the most recent strategies proposed for overcoming those challenges. Finally, we summarize the ongoing applications of BMVs in the biotechnological field.
Collapse
Affiliation(s)
- Keshia F Castillo-Romero
- School of Engineering and Science, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo León, Mexico
| | - Arlette Santacruz
- School of Engineering and Science, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo León, Mexico
| | - José González-Valdez
- School of Engineering and Science, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo León, Mexico
| |
Collapse
|
14
|
Deng DK, Zhang JJ, Gan D, Zou JK, Wu RX, Tian Y, Yin Y, Li X, Chen FM, He XT. Roles of extracellular vesicles in periodontal homeostasis and their therapeutic potential. J Nanobiotechnology 2022; 20:545. [PMID: 36585740 PMCID: PMC9801622 DOI: 10.1186/s12951-022-01757-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
Periodontal tissue is a highly dynamic and frequently stimulated area where homeostasis is easily destroyed, leading to proinflammatory periodontal diseases. Bacteria-bacteria and cell-bacteria interactions play pivotal roles in periodontal homeostasis and disease progression. Several reviews have comprehensively summarized the roles of bacteria and stem cells in periodontal homeostasis. However, they did not describe the roles of extracellular vesicles (EVs) from bacteria and cells. As communication mediators evolutionarily conserved from bacteria to eukaryotic cells, EVs secreted by bacteria or cells can mediate interactions between bacteria and their hosts, thereby offering great promise for the maintenance of periodontal homeostasis. This review offers an overview of EV biogenesis, the effects of EVs on periodontal homeostasis, and recent advances in EV-based periodontal regenerative strategies. Specifically, we document the pathogenic roles of bacteria-derived EVs (BEVs) in periodontal dyshomeostasis, focusing on plaque biofilm formation, immune evasion, inflammatory pathway activation and tissue destruction. Moreover, we summarize recent advancements in cell-derived EVs (CEVs) in periodontal homeostasis, emphasizing the multifunctional biological effects of CEVs on periodontal tissue regeneration. Finally, we discuss future challenges and practical perspectives for the clinical translation of EV-based therapies for periodontitis.
Collapse
Affiliation(s)
- Dao-Kun Deng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jiu-Jiu Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Dian Gan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jie-Kang Zou
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Rui-Xin Wu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yi Tian
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xuan Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.
| | - Xiao-Tao He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.
| |
Collapse
|
15
|
Kikuchi Y, Toyofuku M, Ichinaka Y, Kiyokawa T, Obana N, Nomura N, Taoka A. Physical Properties and Shifting of the Extracellular Membrane Vesicles Attached to Living Bacterial Cell Surfaces. Microbiol Spectr 2022; 10:e0216522. [PMID: 36383005 PMCID: PMC9769862 DOI: 10.1128/spectrum.02165-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022] Open
Abstract
Bacterial cells release nanometer-sized extracellular membrane vesicles (MVs) to deliver cargo molecules for use in mediating various biological processes. However, the detailed processes of transporting these cargos from MVs to recipient cells remain unclear because of the lack of imaging techniques to image nanometer-sized fragile vesicles in a living bacterial cell surface. Herein, we quantitatively demonstrated that the direct binding of MV to the cell surface significantly promotes hydrophobic quorum-sensing signal (C16-HSL) transportation to the recipient cells. Moreover, we analyzed the MV-binding process in the Paracoccus denitrificans cell surface using high-speed atomic force microscopy phase imaging. Although MV shapes were unaltered after binding to the cell surface, the physical properties of a group of single MV particles were shifted. Additionally, the phase shift values of MVs were higher than that of the cell's surfaces upon binding, whereas the phase shift values of the group of MVs were decreased during observation. The shifting physical properties occurred irreversibly only once for each MV during the observations. The decreasing phase shift values indicated alterations of chemical components in the MVs as well, thereby suggesting the dynamic process in which single MV particles deliver their hydrophobic cargo into the recipient cell. IMPORTANCE Compared to the increasing knowledge about MV release mechanisms from donor cells, the mechanism by which recipient cells receive cargo from MVs remains unknown. Herein, we have successfully imaged single MV-binding processes in living bacterial cell surfaces. Accordingly, we confirmed the shift in the MV hydrophobic properties after landing on the cell surface. Our results showed the detailed states and the attaching process of a single MV into the cell surface and can aid the development of a new model for MV reception into Gram-negative bacterial cell surfaces. The insight provided by this study is significant for understanding MV-mediated cell-cell communication mechanisms. Moreover, the AFM technique presented for nanometer-scaled mapping of dynamic physical properties alteration on a living cell could be applied for the analyses of various biological phenomena occurring on the cell surface, and it gives us a new view into the understanding of the phenotypes of the bacterial cell surface.
Collapse
Affiliation(s)
- Yousuke Kikuchi
- Institute of Science and Engineering, Kanazawa University, Kakuma, Kanazawa, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma, Kanazawa, Japan
| | - Masanori Toyofuku
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennodai, Tsukuba, Japan
- Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, Tennodai, Tsukuba, Japan
- Suntory Rising Stars Encouragement Program in Life Sciences (SunRiSE), Seika, Kyoto, Japan
| | - Yuki Ichinaka
- Institute of Science and Engineering, Kanazawa University, Kakuma, Kanazawa, Japan
| | - Tatsunori Kiyokawa
- Graduate of Life and Environmental Sciences, University of Tsukuba, Tennodai, Tsukuba, Japan
| | - Nozomu Obana
- Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, Tennodai, Tsukuba, Japan
- Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tennodai, Tsukuba, Japan
| | - Nobuhiko Nomura
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennodai, Tsukuba, Japan
- Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, Tennodai, Tsukuba, Japan
| | - Azuma Taoka
- Institute of Science and Engineering, Kanazawa University, Kakuma, Kanazawa, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma, Kanazawa, Japan
| |
Collapse
|
16
|
Tian C, Wang K, Zhao M, Cong S, Di X, Li R. Extracellular vesicles participate in the pathogenesis of sepsis. Front Cell Infect Microbiol 2022; 12:1018692. [PMID: 36579343 PMCID: PMC9791067 DOI: 10.3389/fcimb.2022.1018692] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Sepsis is one of the leading causes of mortality worldwide and is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. The early diagnosis and effective treatment of sepsis still face challenges due to its rapid progression, dynamic changes, and strong heterogeneity among different individuals. To develop novel strategies to control sepsis, a better understanding of the complex mechanisms of sepsis is vital. Extracellular vesicles (EVs) are membrane vesicles released from cells through different mechanisms. In the disease state, the number of EVs produced by activated or apoptotic cells and the cargoes they carry were altered. They regulated the function of local or distant host cells in autocrine or paracrine ways. Current studies have found that EVs are involved in the occurrence and development of sepsis through multiple pathways. In this review, we focus on changes in the cargoes of EVs in sepsis, the regulatory roles of EVs derived from host cells and bacteria, and how EVs are involved in multiple pathological processes and organ dysfunction in sepsis. Overall, EVs have great application prospects in sepsis, such as early diagnosis of sepsis, dynamic monitoring of disease, precise therapeutic targets, and prevention of sepsis as a vaccine platform.
Collapse
Affiliation(s)
- Chang Tian
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Min Zhao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Shan Cong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xin Di
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ranwei Li
- Department of Urinary Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China,*Correspondence: Ranwei Li,
| |
Collapse
|
17
|
Involvement of Bacterial Extracellular Membrane Nanovesicles in Infectious Diseases and Their Application in Medicine. Pharmaceutics 2022; 14:pharmaceutics14122597. [PMID: 36559091 PMCID: PMC9784355 DOI: 10.3390/pharmaceutics14122597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/02/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Bacterial extracellular membrane nanovesicles (EMNs) are attracting the attention of scientists more and more every year. These formations are involved in the pathogenesis of numerous diseases, among which, of course, the leading role is occupied by infectious diseases, the causative agents of which are a range of Gram-positive and Gram-negative bacteria. A separate field for the study of the role of EMN is cancer. Extracellular membrane nanovesicles nowadays have a practical application as vaccine carriers for immunization against many infectious diseases. At present, the most essential point is their role in stimulating immune response to bacterial infections and tumor cells. The possibility of nanovesicles' practical use in several disease treatments is being evaluated. In our review, we listed diseases, focusing on their multitude and diversity, for which EMNs are essential, and also considered in detail the possibilities of using EMNs in the therapy and prevention of various pathologies.
Collapse
|
18
|
Jalalvand F, Su YC, Manat G, Chernobrovkin A, Kadari M, Jonsson S, Janousková M, Rutishauser D, Semsey S, Løbner-Olesen A, Sandblad L, Flärdh K, Mengin-Lecreulx D, Zubarev RA, Riesbeck K. Protein domain-dependent vesiculation of Lipoprotein A, a protein that is important in cell wall synthesis and fitness of the human respiratory pathogen Haemophilus influenzae. Front Cell Infect Microbiol 2022; 12:984955. [PMID: 36275016 PMCID: PMC9585305 DOI: 10.3389/fcimb.2022.984955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
The human pathogen Haemophilus influenzae causes respiratory tract infections and is commonly associated with prolonged carriage in patients with chronic obstructive pulmonary disease. Production of outer membrane vesicles (OMVs) is a ubiquitous phenomenon observed in Gram-negative bacteria including H. influenzae. OMVs play an important role in various interactions with the human host; from neutralization of antibodies and complement activation to spread of antimicrobial resistance. Upon vesiculation certain proteins are found in OMVs and some proteins are retained at the cell membrane. The mechanism for this phenomenon is not fully elucidated. We employed mass spectrometry to study vesiculation and the fate of proteins in the outer membrane. Functional groups of proteins were differentially distributed on the cell surface and in OMVs. Despite its supposedly periplasmic and outer membrane location, we found that the peptidoglycan synthase-activator Lipoprotein A (LpoA) was accumulated in OMVs relative to membrane fractions. A mutant devoid of LpoA lost its fitness as revealed by growth and electron microscopy. Furthermore, high-pressure liquid chromatography disclosed a lower concentration (55%) of peptidoglycan in the LpoA-deficient H. influenzae compared to the parent wild type bacterium. Using an LpoA-mNeonGreen fusion protein and fluorescence microscopy, we observed that LpoA was enriched in “foci” in the cell envelope, and further located in the septum during cell division. To define the fate of LpoA, C-terminally truncated LpoA-variants were constructed, and we found that the LpoA C-terminal domain promoted optimal transportation to the OMVs as revealed by flow cytometry. Taken together, our study highlights the importance of LpoA for H. influenzae peptidoglycan biogenesis and provides novel insights into cell wall integrity and OMV production.
Collapse
Affiliation(s)
- Farshid Jalalvand
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Yu-Ching Su
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Guillaume Manat
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Alexey Chernobrovkin
- Physiological Chemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Mahendar Kadari
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Sandra Jonsson
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Martina Janousková
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Dorothea Rutishauser
- Physiological Chemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Szabolcs Semsey
- Centre for Bacterial Stress Response and Persistence, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Anders Løbner-Olesen
- Centre for Bacterial Stress Response and Persistence, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Klas Flärdh
- Department of Biology, Lund University, Lund, Sweden
| | - Dominique Mengin-Lecreulx
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Roman A. Zubarev
- Physiological Chemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
- *Correspondence: Kristian Riesbeck,
| |
Collapse
|
19
|
Bhar S, Zhao G, Bartel JD, Sterchele H, Del Mazo A, Emerson LE, Edelmann MJ, Jones MK. Bacterial extracellular vesicles control murine norovirus infection through modulation of antiviral immune responses. Front Immunol 2022; 13:909949. [PMID: 35990695 PMCID: PMC9386532 DOI: 10.3389/fimmu.2022.909949] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Human norovirus is the primary cause of non-bacterial gastroenteritis globally and is the second leading cause of diarrheal deaths in children in developing countries. However, effective therapeutics which prevent or clear norovirus infection are not yet available due to a lack of understanding regarding norovirus pathogenesis. Evidence shows that noroviruses can bind to the surface of commensal bacteria, and the presence of these bacteria alters both acute and persistent murine norovirus infection through the modulation of host immune responses. Interestingly, norovirus-bacterial interactions also affect the bacteria by inducing bacterial stress responses and increasing the production of bacterial extracellular vesicles. Given the established ability of these vesicles to easily cross the intestinal barriers, enter the lamina propria, and modulate host responses, we hypothesized that bacterial extracellular vesicles influence murine norovirus infection through modulation of the antiviral immune response. In this study, we show that murine norovirus can attach to purified bacterial vesicles, facilitating co-inoculation of target cells with both virus and vesicle. Furthermore, we have found that when murine noroviruses and vesicles are used to co-inoculate macrophages, viral infection is reduced compared to virus infection alone. Specifically, co-inoculation with bacterial vesicles results in higher production and release of pro-inflammatory cytokines in response to viral infection. Ultimately, given that murine norovirus infection increases bacterial vesicle production in vivo, these data indicate that bacterial vesicles may serve as a mechanism by which murine norovirus infection is ultimately controlled and limited to a short-term disease.
Collapse
|
20
|
Krishnan N, Kubiatowicz LJ, Holay M, Zhou J, Fang RH, Zhang L. Bacterial membrane vesicles for vaccine applications. Adv Drug Deliv Rev 2022; 185:114294. [PMID: 35436569 DOI: 10.1016/j.addr.2022.114294] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/13/2022] [Accepted: 04/10/2022] [Indexed: 12/11/2022]
Abstract
Vaccines have been highly successful in the management of many diseases. However, there are still numerous illnesses, both infectious and noncommunicable, for which there are no clinically approved vaccine formulations. While there are unique difficulties that must be overcome in the case of each specific disease, there are also a number of common challenges that have to be addressed for effective vaccine development. In recent years, bacterial membrane vesicles (BMVs) have received increased attention as a potent and versatile vaccine platform. BMVs are inherently immunostimulatory and are able to activate both innate and adaptive immune responses. Additionally, BMVs can be readily taken up and processed by immune cells due to their nanoscale size. Finally, BMVs can be modified in a variety of ways, including by genetic engineering, cargo loading, and nanoparticle coating, in order to create multifunctional platforms that can be leveraged against different diseases. Here, an overview of the interactions between BMVs and immune cells is provided, followed by discussion on the applications of BMV vaccine nanotechnology against bacterial infections, viral infections, and cancers.
Collapse
Affiliation(s)
- Nishta Krishnan
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Luke J Kubiatowicz
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Maya Holay
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
21
|
Chen J, Zhang H, Wang S, Du Y, Wei B, Wu Q, Wang H. Inhibitors of Bacterial Extracellular Vesicles. Front Microbiol 2022; 13:835058. [PMID: 35283837 PMCID: PMC8905621 DOI: 10.3389/fmicb.2022.835058] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/25/2022] [Indexed: 01/22/2023] Open
Abstract
Both Gram-positive and Gram-negative bacteria can secrete extracellular vesicles (EVs), which contain numerous active substances. EVs mediate bacterial interactions with their hosts or other microbes. Bacterial EVs play a double-edged role in infections through various mechanisms, including the delivery of virulence factors, modulating immune responses, mediating antibiotic resistance, and inhibiting competitive microbes. The spread of antibiotic resistance continues to represent a difficult clinical challenge. Therefore, the investigation of novel therapeutics is a valuable research endeavor for targeting antibiotic-resistant bacterial infections. As a pathogenic substance of bacteria, bacterial EVs have gained increased attention. Thus, EV inhibitors are expected to function as novel antimicrobial agents. The inhibition of EV production, EV activity, and EV-stimulated inflammation are considered potential pathways. This review primarily introduces compounds that effectively inhibit bacterial EVs and evaluates the prospects of their application.
Collapse
Affiliation(s)
- Jianwei Chen
- College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China.,State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Hongfang Zhang
- College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Siqi Wang
- College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Yujie Du
- College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Bin Wei
- College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Qiang Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Hong Wang
- College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China.,Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
22
|
Cell Membrane-Cloaked Nanotherapeutics for Targeted Drug Delivery. Int J Mol Sci 2022; 23:ijms23042223. [PMID: 35216342 PMCID: PMC8879543 DOI: 10.3390/ijms23042223] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
Cell membrane cloaking technique is bioinspired nanotechnology that takes advantage of naturally derived design cues for surface modification of nanoparticles. Unlike modification with synthetic materials, cell membranes can replicate complex physicochemical properties and biomimetic functions of the parent cell source. This technique indeed has the potential to greatly augment existing nanotherapeutic platforms. Here, we provide a comprehensive overview of engineered cell membrane-based nanotherapeutics for targeted drug delivery and biomedical applications and discuss the challenges and opportunities of cell membrane cloaking techniques for clinical translation.
Collapse
|
23
|
Karvonen K, Tammisto H, Nykky J, Gilbert L. Borrelia burgdorferi Outer Membrane Vesicles Contain Antigenic Proteins, but Do Not Induce Cell Death in Human Cells. Microorganisms 2022; 10:microorganisms10020212. [PMID: 35208666 PMCID: PMC8878412 DOI: 10.3390/microorganisms10020212] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
Like many bacterial species, Borrelia burgdorferi, the pleomorphic bacterium that causes Lyme borreliosis, produces outer membrane vesicles (OMVs). Borrelial OMVs (BbOMVs) have been identified as containing virulence factors, such as outer surface proteins (Osps) A, B, and C, as well as DNA. However, the pathogenicity of BbOMVs in disease development is still unclear. In this study, we characterized purified BbOMVs by analyzing their size and immunolabeling for known antigenic markers: OspA, OspC, p39, and peptidoglycan. In addition, BbOMVs were cocultured with human non-immune cells for cytotoxicity analysis. The results demonstrated that, on average, the vesicles were small, ranging between 11 and 108 nm in diameter. In addition, both OspA and OspC, as well as Lyme arthritis markers p39 and peptidoglycan, were detected from BbOMVs. Furthermore, BbOMVs were cocultured with non-immune cells, which did not result in cell death. Combined, these results suggested that BbOMVs could participate in the induction of infection by functioning as a decoy for the host immune system. Furthermore, BbOMVs might serve as a means for persistent antigens to remain in the host for prolonged periods of time.
Collapse
Affiliation(s)
- Kati Karvonen
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyvaskyla, P.O. Box 35, FI-40014 Jyvaskyla, Finland; (H.T.); (J.N.)
- Correspondence: (K.K.); (L.G.)
| | - Hanna Tammisto
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyvaskyla, P.O. Box 35, FI-40014 Jyvaskyla, Finland; (H.T.); (J.N.)
| | - Jonna Nykky
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyvaskyla, P.O. Box 35, FI-40014 Jyvaskyla, Finland; (H.T.); (J.N.)
| | - Leona Gilbert
- Te?ted Oy, Mattilanniemi 6-8, FI-40100 Jyvaskyla, Finland
- Correspondence: (K.K.); (L.G.)
| |
Collapse
|
24
|
Huang Y, Nieh MP, Chen W, Lei Y. Outer membrane vesicles (OMVs) enabled bio-applications: A critical review. Biotechnol Bioeng 2021; 119:34-47. [PMID: 34698385 DOI: 10.1002/bit.27965] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 07/28/2021] [Accepted: 10/10/2021] [Indexed: 11/07/2022]
Abstract
Outer membrane vesicles (OMVs) are nanoscale spherical vesicles released from Gram-negative bacteria. The lipid bilayer membrane structure of OMVs consists of similar components as bacterial membrane and thus has attracted more and more attention in exploiting OMVs' bio-applications. Although the endotoxic lipopolysaccharide on natural OMVs may impose potential limits on their clinical applications, genetic modification can reduce their endotoxicity and decorate OMVs with multiple functional proteins. These genetically engineered OMVs have been employed in various fields including vaccination, drug delivery, cancer therapy, bioimaging, biosensing, and enzyme carrier. This review will first briefly introduce the background of OMVs followed by recent advances in functionalization and various applications of engineered OMVs with an emphasis on the working principles and their performance, and then discuss about the future trends of OMVs in biomedical applications.
Collapse
Affiliation(s)
- Yikun Huang
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Mu-Ping Nieh
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Yu Lei
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
25
|
Lipoproteins Are Responsible for the Pro-Inflammatory Property of Staphylococcus aureus Extracellular Vesicles. Int J Mol Sci 2021; 22:ijms22137099. [PMID: 34281154 PMCID: PMC8268867 DOI: 10.3390/ijms22137099] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 01/08/2023] Open
Abstract
Staphylococcal aureus
(S. aureus), a Gram-positive bacteria, is known to cause various infections. Extracellular vesicles (EVs) are a heterogeneous array of membranous structures secreted by cells from all three domains of life, i.e., eukaryotes, bacteria, and archaea. Bacterial EVs are implied to be involved in both bacteria–bacteria and bacteria–host interactions during infections. It is still unclear how S. aureus EVs interact with host cells and induce inflammatory responses. In this study, EVs were isolated from S. aureus and mutant strains deficient in either prelipoprotein lipidation (Δlgt) or major surface proteins (ΔsrtAB). Their immunostimulatory capacities were assessed both in vitro and in vivo. We found that S. aureus EVs induced pro-inflammatory responses both in vitro and in vivo. However, this activity was dependent on lipidated lipoproteins (Lpp), since EVs isolated from the Δlgt showed no stimulation. On the other hand, EVs isolated from the ΔsrtAB mutant showed full immune stimulation, indicating the cell wall anchoring of surface proteins did not play a role in immune stimulation. The immune stimulation of S. aureus EVs was mediated mainly by monocytes/macrophages and was TLR2 dependent. In this study, we demonstrated that not only free Lpp but also EV-imbedded Lpp had high pro-inflammatory activity.
Collapse
|
26
|
Cai Q, He B, Wang S, Fletcher S, Niu D, Mitter N, Birch PRJ, Jin H. Message in a Bubble: Shuttling Small RNAs and Proteins Between Cells and Interacting Organisms Using Extracellular Vesicles. ANNUAL REVIEW OF PLANT BIOLOGY 2021; 72:497-524. [PMID: 34143650 PMCID: PMC8369896 DOI: 10.1146/annurev-arplant-081720-010616] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Communication between plant cells and interacting microorganisms requires the secretion and uptake of functional molecules to and from the extracellular environment and is essential for the survival of both plants and their pathogens. Extracellular vesicles (EVs) are lipid bilayer-enclosed spheres that deliver RNA, protein, and metabolite cargos from donor to recipient cells and participate in many cellular processes. Emerging evidencehas shown that both plant and microbial EVs play important roles in cross-kingdom molecular exchange between hosts and interacting microbes to modulate host immunity and pathogen virulence. Recent studies revealed that plant EVs function as a defense system by encasing and delivering small RNAs (sRNAs) into pathogens, thereby mediating cross-species and cross-kingdom RNA interference to silence virulence-related genes. This review focuses on the latest advances in our understanding of plant and microbial EVs and their roles in transporting regulatory molecules, especially sRNAs, between hosts and pathogens. EV biogenesis and secretion are also discussed, as EV function relies on these important processes.
Collapse
Affiliation(s)
- Qiang Cai
- Department of Microbiology and Plant Pathology and Center for Plant Cell Biology, Institute for Integrative Genome Biology, University of California, Riverside, California 92507, USA;
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Baoye He
- Department of Microbiology and Plant Pathology and Center for Plant Cell Biology, Institute for Integrative Genome Biology, University of California, Riverside, California 92507, USA;
| | - Shumei Wang
- Department of Microbiology and Plant Pathology and Center for Plant Cell Biology, Institute for Integrative Genome Biology, University of California, Riverside, California 92507, USA;
| | - Stephen Fletcher
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Dongdong Niu
- College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Neena Mitter
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Paul R J Birch
- Division of Plant Sciences, School of Life Science, University of Dundee at James Hutton Institute, Invergowrie, Dundee DD2 5DA, United Kingdom
- Cell and Molecular Sciences, James Hutton Institute, Invergowrie, Dundee DD2 5DA, United Kingdom
| | - Hailing Jin
- Department of Microbiology and Plant Pathology and Center for Plant Cell Biology, Institute for Integrative Genome Biology, University of California, Riverside, California 92507, USA;
| |
Collapse
|
27
|
Peng Y, Yin S, Wang M. Extracellular vesicles of bacteria as potential targets for immune interventions. Hum Vaccin Immunother 2021; 17:897-903. [PMID: 32873124 DOI: 10.1080/21645515.2020.1799667] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Bacterial infection is one of the most common and serious diseases. Extracellular vesicles (EVs) expressed by bacterial cells during infection and their biological functions have been a growing field in recent years. The study of the immune interaction mechanism between EVs and bacteria has become more significant. EVs are released into the extracellular microenvironment during bacterial infection. EVs carry various lipids, proteins, nucleic acids, and other substances of host bacteria and participate in various physiological and pathological processes. EV-based vaccines against bacterial infection are also being evaluated. This review focuses on the biological characteristics of EVs, the interaction between EVs and the host immune system, and the potential of EVs as new vaccines. A deeper understanding of the interaction between EVs and the immune system informs on the biological function and heterogeneity of EVs. This knowledge also can facilitate the development and application of EVs and their potential as vaccines.
Collapse
Affiliation(s)
- Yizhi Peng
- Department of Laboratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Sheng Yin
- Department of Laboratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Min Wang
- Department of Laboratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
28
|
Engevik MA, Danhof HA, Ruan W, Engevik AC, Chang-Graham AL, Engevik KA, Shi Z, Zhao Y, Brand CK, Krystofiak ES, Venable S, Liu X, Hirschi KD, Hyser JM, Spinler JK, Britton RA, Versalovic J. Fusobacterium nucleatum Secretes Outer Membrane Vesicles and Promotes Intestinal Inflammation. mBio 2021; 12:e02706-20. [PMID: 33653893 PMCID: PMC8092269 DOI: 10.1128/mbio.02706-20] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
Multiple studies have implicated microbes in the development of inflammation, but the mechanisms remain unknown. Bacteria in the genus Fusobacterium have been identified in the intestinal mucosa of patients with digestive diseases; thus, we hypothesized that Fusobacterium nucleatum promotes intestinal inflammation. The addition of >50 kDa F. nucleatum conditioned media, which contain outer membrane vesicles (OMVs), to colonic epithelial cells stimulated secretion of the proinflammatory cytokines interleukin-8 (IL-8) and tumor necrosis factor (TNF). In addition, purified F. nucleatum OMVs, but not compounds <50 kDa, stimulated IL-8 and TNF production; which was decreased by pharmacological inhibition of Toll-like receptor 4 (TLR4). These effects were linked to downstream effectors p-ERK, p-CREB, and NF-κB. F. nucleatum >50-kDa compounds also stimulated TNF secretion, p-ERK, p-CREB, and NF-κB activation in human colonoid monolayers. In mice harboring a human microbiota, pretreatment with antibiotics and a single oral gavage of F. nucleatum resulted in inflammation. Compared to mice receiving vehicle control, mice treated with F. nucleatum showed disruption of the colonic architecture, with increased immune cell infiltration and depleted mucus layers. Analysis of mucosal gene expression revealed increased levels of proinflammatory cytokines (KC, TNF, IL-6, IFN-γ, and MCP-1) at day 3 and day 5 in F. nucleatum-treated mice compared to controls. These proinflammatory effects were absent in mice who received F. nucleatum without pretreatment with antibiotics, suggesting that an intact microbiome is protective against F. nucleatum-mediated immune responses. These data provide evidence that F. nucleatum promotes proinflammatory signaling cascades in the context of a depleted intestinal microbiome.IMPORTANCE Several studies have identified an increased abundance of Fusobacterium in the intestinal tracts of patients with colon cancer, liver cirrhosis, primary sclerosing cholangitis, gastroesophageal reflux disease, HIV infection, and alcoholism. However, the direct mechanism(s) of action of Fusobacterium on pathophysiological within the gastrointestinal tract is unclear. These studies have identified that F. nucleatum subsp. polymorphum releases outer membrane vesicles which activate TLR4 and NF-κB to stimulate proinflammatory signals in vitro Using mice harboring a human microbiome, we demonstrate that F. nucleatum can promote inflammation, an effect which required antibiotic-mediated alterations in the gut microbiome. Collectively, these results suggest a mechanism by which F. nucleatum may contribute to intestinal inflammation.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Heather A Danhof
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Amy C Engevik
- Department of Surgical Sciences, Vanderbilt University Medical Center, Nashville Tennessee, USA
| | - Alexandra L Chang-Graham
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Kristen A Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Yanling Zhao
- Department of Pediatrics, Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas, USA
| | - Colleen K Brand
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Evan S Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Susan Venable
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Kendal D Hirschi
- Department of Pediatrics and Human and Molecular Genetics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jennifer K Spinler
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Robert A Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
29
|
Characterization and proteomic analysis of outer membrane vesicles from a commensal microbe, Enterobacter cloacae. J Proteomics 2021; 231:103994. [PMID: 33007464 DOI: 10.1016/j.jprot.2020.103994] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022]
Abstract
Outer membrane vesicles (OMVs) are membrane-enclosed spherical entities released by gram-negative bacteria and are important for bacterial survival under stress conditions. There have been numerous studies on OMVs released by gram-negative pathogenic bacteria, but an understanding of the functions and characteristics of the OMVs produced by commensal microbes is still lacking. Enterobacter cloacae is a gram-negative commensal bacterium present in the human gut microbiome, but this organism can also function as an opportunistic pathogen. Understanding the OMV-mediated communication route between bacteria-bacteria or bacteria-host is essential for the determination of the biological functions of the commensal bacterium in the gut and delineating between benign and virulent characteristics. In this study, we have described a proteome of E. cloacae OMVs, which are membrane vesicles in a size range of 20-300 nm. Proteomic analysis showed the presence of membrane-bound proteins, including transporters, receptors, signaling molecules, and protein channels. The physical and proteomic analyses also indicate this bacterium uses two mechanisms for OMV production. This study is one of the few existing descriptions of the proteomic profile of OMVs generated by a commensal Proteobacteria, and the first report of OMVs produced by E. cloacae. SIGNIFICANCE: This study prioritizes the importance of understanding the vesicular proteome of the human commensal bacterium, Enterobacter cloacae. We demonstrate for the first time that the gram-negative bacterium E. cloacae ATCC 13047 produces outer membrane vesicles (OMVs). The proteomic analysis showed enrichment of membrane-bound proteins in these vesicles. Understanding the cargo proteins of OMVs will help in exploring the physiological and functional role of these vesicles in the human microbiome and how they assist in the conversion of a bacterium from commensal to pathogen under certain conditions. While EM images reveal vesicles budding from the bacterial surface, the presence of cytoplasmic proteins and genomic DNA within the OMVs indicate that explosive cell lysis is an additional mechanism of biogenesis for these OMVs along with outer membrane blebbing. This research encourages future work on characterizing membrane vesicles produced by commensal bacterial and investigating their role in cell to cell communication.
Collapse
|
30
|
OMV Vaccines and the Role of TLR Agonists in Immune Response. Int J Mol Sci 2020; 21:ijms21124416. [PMID: 32575921 PMCID: PMC7352230 DOI: 10.3390/ijms21124416] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/26/2022] Open
Abstract
Outer Membrane Vesicles (OMVs) are bacterial nanoparticles that are spontaneously released during growth both in vitro and in vivo by Gram-negative bacteria. They are spherical, bilayered membrane nanostructures that contain many components found within the external surface of the parent bacterium. Naturally, OMVs serve the bacteria as a mechanism to deliver DNA, RNA, proteins, and toxins, as well as to promote biofilm formation and remodel the outer membrane during growth. On the other hand, as OMVs possess the optimal size to be uptaken by immune cells, and present a range of surface-exposed antigens in native conformation and Toll-like receptor (TLR) activating components, they represent an attractive and powerful vaccine platform able to induce both humoral and cell-mediated immune responses. This work reviews the TLR-agonists expressed on OMVs and their capability to trigger individual TLRs expressed on different cell types of the immune system, and then focuses on their impact on the immune responses elicited by OMVs compared to traditional vaccines.
Collapse
|
31
|
Hu R, Li J, Zhao Y, Lin H, Liang L, Wang M, Liu H, Min Y, Gao Y, Yang M. Exploiting bacterial outer membrane vesicles as a cross-protective vaccine candidate against avian pathogenic Escherichia coli (APEC). Microb Cell Fact 2020; 19:119. [PMID: 32493405 PMCID: PMC7268718 DOI: 10.1186/s12934-020-01372-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/16/2020] [Indexed: 12/15/2022] Open
Abstract
Background The well-known fact that avian pathogenic Escherichia coli (APEC) is harder to prevent due to its numerous serogroups has promoted the development of biological immunostimulatory materials as new vaccine candidates in poultry farms. Bacterial outer membrane vesicles (OMVs), known as spherical nanovesicles enriched with various immunostimulants, are naturally secreted by Gram-negative bacteria, and have gained much attention for developing effective vaccine candidates. Recent report has demonstrated that OMVs of APEC O78 can induce protective immunity in chickens. Here, a novel multi-serogroup OMVs (MOMVs) vaccine was developed to achieve cross-protection against APEC infection in broiler chickens. Results In this study, OMVs produced by three APEC strains were isolated, purified and prepared into MOMVs by mixing these three OMVs. By using SDS-PAGE and LC–MS/MS, 159 proteins were identified in MOMVs and the subcellular location and biological functions of 20 most abundant proteins were analyzed. The immunogenicity of MOMVs was evaluated, and the results showed that MOMVs could elicit innate immune responses, including internalization by chicken macrophage and production of immunomodulatory cytokines. Vaccination with MOMVs induced specific broad-spectrum antibodies as well as Th1 and Th17 immune responses. The animal experiment has confirmed that immunization with an appropriate dose of MOMVs could not cause any adverse effect and was able to reduce bacteria loads and pro-inflammatory cytokines production, thus providing effective cross-protection against lethal infections induced by multi-serogroup APEC strains in chickens. Further experiments indicated that, although vesicular proteins were able to induce stronger protective efficiency than lipopolysaccharide, both vesicular proteins and lipopolysaccharide are crucial in MOMVs-mediated protection. Conclusions The multi-serogroup nanovesicles produced by APEC strains will open up a new way for the development of next generation vaccines with low toxicity and broad protection in the treatment and control of APEC infection.
Collapse
Affiliation(s)
- Rujiu Hu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jing Li
- Department of Animal Engineering, Yangling Vocation and Technical College, Yangling, 712100, Shaanxi, China
| | - Yuezhen Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hua Lin
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Liu Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Mimi Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Haojing Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yuna Min
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yupeng Gao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Mingming Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
32
|
Zwarycz AS, Livingstone PG, Whitworth DE. Within-species variation in OMV cargo proteins: the Myxococcus xanthus OMV pan-proteome. Mol Omics 2020; 16:387-397. [PMID: 32373862 DOI: 10.1039/d0mo00027b] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Extracellular membrane vesicles are produced by all domains of life (bacteria, archaea and eukaryotes). Bacterial extracellular vesicles (outer membrane vesicles or OMVs) are produced by outer membrane blebbing, and contain proteins, nucleic acids, virulence factors, lipids and metabolites. OMV functions depend on their internal composition, therefore understanding the proteome of OMVs, and how it varies between organisms, is imperative. Here, we report a comparative proteomic profiling of OMVs from strains of Myxococcus xanthus, a predatory species of Gram-negative myxobacteria whose secretions include secondary metabolites and hydrolytic enzymes, thought to be involved in prey lysis. Ten strains were chosen for study, of which seven had genome sequences available. The remaining three strains were genome sequenced allowing definition of the core and accessory genes and genome-derived proteins found within the pan-genome and pan-proteome respectively. OMVs were isolated from each strain and proteins identified using mass spectrometry. The M. xanthus OMV pan-proteome was found to contain tens of 'core' and hundreds of 'accessory' proteins. Properties of the OMV pan-proteome were compared with those of the pan-proteome deduced from the M. xanthus pan-genome. On average, 80% of 'core' OMV proteins are encoded by genes of the core genome, yet the OMV proteomes of individual strains contain subsets of core genome-derived proteins which only partially overlap. In addition, the distribution of characteristics of vesicle proteins does not correlate with the genome-derived proteome characteristic distribution. We hypothesize that M. xanthus cells package a personalized subset of proteins whose availability is only partially dictated by the presence/absence of encoding genes within the genome.
Collapse
Affiliation(s)
- Allison S Zwarycz
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, Ceredigion SY23 4DD, UK.
| | | | | |
Collapse
|
33
|
Lindholm M, Metsäniitty M, Granström E, Oscarsson J. Outer membrane vesicle-mediated serum protection in Aggregatibacter actinomycetemcomitans. J Oral Microbiol 2020; 12:1747857. [PMID: 32363008 PMCID: PMC7178816 DOI: 10.1080/20002297.2020.1747857] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 01/23/2023] Open
Abstract
Aggregatibacter actinomycetemcomitans belongs to the HACEK group of fastidious Gram-negative organisms, a recognized cause of infective endocarditis. A. actinomycetemcomitans is also implicated in periodontitis, with rapid progress in adolescents. We recently demonstrated that the major outer membrane protein, OmpA1 was critical for serum survival of the A. actinomycetemcomitans serotype a model strain, D7SS, and that the paralogue, OmpA2 could operate as a functional homologue to OmpA1 in mediating serum resistance. In the present work, an essentially serum-sensitive ompA1 ompA2 double mutant A. actinomycetemcomitans strain derivative was exploited to elucidate if A. actinomycetemcomitans OMVs can contribute to bacterial serum resistance. Indeed, supplementation of OMVs resulted in a dose-dependent increase of the survival of the serum-sensitive strain in incubations in 50% normal human serum (NHS). Whereas neither OmpA1 nor OmpA2 was required for the OMV-mediated serum protection, OMVs and LPS from an A. actinomycetemcomitans strain lacking the LPS O-antigen polysaccharide part were significantly impaired in protecting D7SS ompA1 ompA2. Our results using a complement system screen assay support a model where A. actinomycetemcomitans OMVs can act as a decoy, which can trigger complement activation in an LPS-dependent manner, and consume complement components to protect serum-susceptible bacterial cells.
Collapse
Affiliation(s)
- Mark Lindholm
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, Sweden
| | - Marjut Metsäniitty
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, Sweden
| | | | - Jan Oscarsson
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, Sweden
| |
Collapse
|
34
|
Ansari S, Yamaoka Y. Helicobacter pylori Virulence Factors Exploiting Gastric Colonization and its Pathogenicity. Toxins (Basel) 2019; 11:677. [PMID: 31752394 PMCID: PMC6891454 DOI: 10.3390/toxins11110677] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori colonizes the gastric epithelial cells of at least half of the world's population, and it is the strongest risk factor for developing gastric complications like chronic gastritis, ulcer diseases, and gastric cancer. To successfully colonize and establish a persistent infection, the bacteria must overcome harsh gastric conditions. H. pylori has a well-developed mechanism by which it can survive in a very acidic niche. Despite bacterial factors, gastric environmental factors and host genetic constituents together play a co-operative role for gastric pathogenicity. The virulence factors include bacterial colonization factors BabA, SabA, OipA, and HopQ, and the virulence factors necessary for gastric pathogenicity include the effector proteins like CagA, VacA, HtrA, and the outer membrane vesicles. Bacterial factors are considered more important. Here, we summarize the recent information to better understand several bacterial virulence factors and their role in the pathogenic mechanism.
Collapse
Affiliation(s)
- Shamshul Ansari
- Department of Microbiology, Chitwan Medical College and Teaching Hospital, Bharatpur 44200, Chitwan, Nepal;
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
- Global Oita Medical Advanced Research Center for Health, Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, 2002 Holcombe Blvd., Houston, TX 77030, USA
- Borneo Medical and Health Research Centre, Universiti Malaysia Sabah, Kota Kinabaru, Sabah 88400, Malaysia
| |
Collapse
|
35
|
Yang G, Chen S, Zhang J. Bioinspired and Biomimetic Nanotherapies for the Treatment of Infectious Diseases. Front Pharmacol 2019; 10:751. [PMID: 31333467 PMCID: PMC6624236 DOI: 10.3389/fphar.2019.00751] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
There are still great challenges for the effective treatment of infectious diseases, although considerable achievement has been made by using antiviral and antimicrobial agents varying from small-molecule drugs, peptides/proteins, to nucleic acids. The nanomedicine approach is emerging as a new strategy capable of overcoming disadvantages of molecular therapeutics and amplifying their anti-infective activities, by localized delivery to infection sites, reducing off-target effects, and/or attenuating resistance development. Nanotechnology, in combination with bioinspired and biomimetic approaches, affords additional functions to nanoparticles derived from synthetic materials. Herein, we aim to provide a state-of-the-art review on recent progress in biomimetic and bioengineered nanotherapies for the treatment of infectious disease. Different biomimetic nanoparticles, derived from viruses, bacteria, and mammalian cells, are first described, with respect to their construction and biophysicochemical properties. Then, the applications of diverse biomimetic nanoparticles in anti-infective therapy are introduced, either by their intrinsic activity or by loading and site-specifically delivering various molecular drugs. Bioinspired and biomimetic nanovaccines for prevention and/or therapy of infectious diseases are also highlighted. At the end, major translation issues and future directions of this field are discussed.
Collapse
Affiliation(s)
- Guoyu Yang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Sheng Chen
- Department of Pediatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
36
|
Molina-Tijeras JA, Gálvez J, Rodríguez-Cabezas ME. The Immunomodulatory Properties of Extracellular Vesicles Derived from Probiotics: A Novel Approach for the Management of Gastrointestinal Diseases. Nutrients 2019; 11:E1038. [PMID: 31075872 PMCID: PMC6567093 DOI: 10.3390/nu11051038] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/05/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
Probiotics, included in functional foods, nutritional supplements, or nutraceuticals, exhibit different beneficial effects on gut function. They are extensively used to improve the digestive processes as well as reduce the symptoms and progression of different diseases. Probiotics have shown to improve dysbiosis and modulate the immune response of the host by interacting with different cell types. Probiotics and the host can interact in a direct way, but it is becoming apparent that communication occurs also through extracellular vesicles (EVs) derived from probiotics. EVs are key for bacteria-bacteria and bacteria-host interactions, since they carry a wide variety of components that can modulate different signaling pathways, including those involved in the immune response. Interestingly, EVs are recently starting to be considered as an alternative to probiotics in those cases for which the use of live bacteria could be dangerous, such as immunocompromised individuals or situations where the intestinal barrier is impaired. EVs can spread through the mucus layer and interact with the host, avoiding the risk of sepsis. This review summarizes the existing knowledge about EVs from different probiotic strains, their properties, and their potential use for the prevention or treatment of different gastrointestinal diseases.
Collapse
Affiliation(s)
- Jose Alberto Molina-Tijeras
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18071-Granada, Spain.
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada 18012, Spain.
| | - Julio Gálvez
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18071-Granada, Spain.
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada 18012, Spain.
| | - Maria Elena Rodríguez-Cabezas
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Avenida del Conocimiento s/n 18071-Granada, Spain.
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada 18012, Spain.
| |
Collapse
|
37
|
Choi JP, Jeon SG, Kim YK, Cho YS. Role of house dust mite-derived extracellular vesicles in a murine model of airway inflammation. Clin Exp Allergy 2018; 49:227-238. [PMID: 30312497 DOI: 10.1111/cea.13295] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 09/30/2018] [Accepted: 10/03/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND House dust mite (HDM) is the major source of indoor allergens that cause airway disease. Recent evidence suggests that Gram-negative/positive bacteria produce nano-sized extracellular vesicles (EVs) containing diverse components, including various immunostimulatory molecules. However, the association between bacteria-derived EVs and development of airway disease is unclear. OBJECTIVE To identify and isolate HDM-derived EVs and to evaluate their effect on the development of airway inflammation. METHODS Extracellular vesicles were isolated from crude HDM extracts by ultra-centrifugation, and their physical and immunological characteristics and roles in airway inflammation were tested in vitro and in murine models of airway inflammation. In addition, 16s metagenome analysis of nucleic acid from EVs was performed to identify their origin. RESULTS Round, bilayered vesicles measuring 80-100 nanometres and containing abundant amounts of LPS were isolated. These vesicles induced innate immune responses both in vitro and in vivo. Intranasal exposure of naïve mice to HDM EVs induced production of cytokines associated with development of Th2-mediated and mixed (Th1-/Th2-/Th17-mediated) airway inflammation to allergen. Metagenome analysis identified Bacteroidetes and Proteobacteria as the probable sources of HDM EVs. CONCLUSION House dust mite EVs originating from Gram-negative bacteria may play an important role on the development of airway inflammation.
Collapse
Affiliation(s)
- Jun-Pyo Choi
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | - You Sook Cho
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
38
|
Nice JB, Balashova NV, Kachlany SC, Koufos E, Krueger E, Lally ET, Brown AC. Aggregatibacter actinomycetemcomitans Leukotoxin Is Delivered to Host Cells in an LFA-1-Indepdendent Manner When Associated with Outer Membrane Vesicles. Toxins (Basel) 2018; 10:toxins10100414. [PMID: 30322160 PMCID: PMC6215133 DOI: 10.3390/toxins10100414] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/08/2018] [Accepted: 10/10/2018] [Indexed: 12/21/2022] Open
Abstract
The Gram-negative bacterium, Aggregatibacter actinomycetemcomitans, has been associated with localized aggressive periodontitis (LAP). In particular, highly leukotoxic strains of A. actinomycetemcomitans have been more closely associated with this disease, suggesting that LtxA is a key virulence factor for A. actinomycetemcomitans. LtxA is secreted across both the inner and outer membranes via the Type I secretion system, but has also been found to be enriched within outer membrane vesicles (OMVs), derived from the bacterial outer membrane. We have characterized the association of LtxA with OMVs produced by the highly leukotoxic strain, JP2, and investigated the interaction of these OMVs with host cells to understand how LtxA is delivered to host cells in this OMV-associated form. Our results demonstrated that a significant fraction of the secreted LtxA exists in an OMV-associated form. Furthermore, we have discovered that in this OMV-associated form, the toxin is trafficked to host cells by a cholesterol- and receptor-independent mechanism in contrast to the mechanism by which free LtxA is delivered. Because OMV-associated toxin is trafficked to host cells in an entirely different manner than free toxin, this study highlights the importance of studying both free and OMV-associated forms of LtxA to understand A. actinomycetemcomitans virulence.
Collapse
Affiliation(s)
- Justin B Nice
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA 18015, USA.
| | - Nataliya V Balashova
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA.
| | - Scott C Kachlany
- Department of Oral Biology, Rutgers University School of Dental Medicine, Newark, NJ 07101, USA.
| | - Evan Koufos
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA 18015, USA.
| | - Eric Krueger
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA 18015, USA.
| | - Edward T Lally
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA.
| | - Angela C Brown
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA 18015, USA.
| |
Collapse
|
39
|
Lin LCW, Chattopadhyay S, Lin JC, Hu CMJ. Advances and Opportunities in Nanoparticle- and Nanomaterial-Based Vaccines against Bacterial Infections. Adv Healthc Mater 2018; 7:e1701395. [PMID: 29508547 DOI: 10.1002/adhm.201701395] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/22/2018] [Indexed: 02/06/2023]
Abstract
As the dawn of the postantibiotic era we approach, antibacterial vaccines are becoming increasingly important for managing bacterial infection and reducing the need for antibiotics. Despite the success of vaccination, vaccines remain unavailable for many pressing microbial diseases, including tuberculosis, chlamydia, and staphylococcus infections. Amid continuing research efforts in antibacterial vaccine development, the advancement of nanomaterial engineering has brought forth new opportunities in vaccine designs. With increasing knowledge in antibacterial immunity and immunologic adjuvants, innovative nanoparticles are designed to elicit the appropriate immune responses for effective antimicrobial defense. Rationally designed nanoparticles are demonstrated to overcome delivery barriers to shape the adaptive immunity. This article reviews the advances in nanoparticle- and nanomaterial-based antibacterial vaccines and summarizes the development of nanoparticulate adjuvants for immune potentiation against microbial pathogens. In addition, challenges and progress in ongoing antibacterial vaccine development are discussed to highlight the opportunities for future vaccine designs.
Collapse
Affiliation(s)
- Leon Chien-Wei Lin
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| | - Saborni Chattopadhyay
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| | - Jung-Chen Lin
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| | - Che-Ming Jack Hu
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| |
Collapse
|
40
|
Jalalvand F, Riesbeck K. Update on non-typeable Haemophilus influenzae-mediated disease and vaccine development. Expert Rev Vaccines 2018; 17:503-512. [DOI: 10.1080/14760584.2018.1484286] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Farshid Jalalvand
- Centre for Bacterial Stress Response and Persistence, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
41
|
Beikzadeh B, Nikbakht Brujeni G. Protection against neonatal enteric colibacillosis employing E. Coli-derived outer membrane vesicles in formulation and without vitamin D3. BMC Res Notes 2018; 11:302. [PMID: 29769118 PMCID: PMC5956550 DOI: 10.1186/s13104-018-3442-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 05/11/2018] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Enterotoxigenic Escherichia Coli (ETEC) is the cause of diarrhea and even death in humans and offspring of animals. Outer membrane vesicles (OMVs) of the ETEC was prepared and its potential as a vaccine candidate against enteric colibacillosis in neonatal mice was evaluated. Dam mice intradermally injected with ETEC-derived OMVs and OMVs plus an active form of vitamin D3 (avD3). Mucosal and systemic immune responses in mice and passive immunity protection against ETEC lethality in their offspring was investigated. RESULTS Immunization of adult mice via ETEC-derived OMV alone and in formulation with avD3 protect offspring from ETEC-induced lethality. Nevertheless, avD3 did not indicate a positive effect on mucosal and systemic immune responses. Only the combination of OMV plus avD3 elicited a significant (P < 0.05) increase in the level of specific IgA antibodies in serum.
Collapse
Affiliation(s)
- Babak Beikzadeh
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Azadi Avenue, Tehran, Iran
| | - Gholamreza Nikbakht Brujeni
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Azadi Avenue, Tehran, Iran.
| |
Collapse
|
42
|
Yu YJ, Wang XH, Fan GC. Versatile effects of bacterium-released membrane vesicles on mammalian cells and infectious/inflammatory diseases. Acta Pharmacol Sin 2018; 39:514-533. [PMID: 28858295 PMCID: PMC5888691 DOI: 10.1038/aps.2017.82] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/10/2017] [Indexed: 01/18/2023]
Abstract
Gram-negative bacterium-released outer-membrane vesicles (OMVs) and Gram-positive bacterium-released membrane vesicles (MVs) share significant similarities with mammalian cell-derived MVs (eg, microvesicles and exosomes) in terms of structure and their biological activities. Recent studies have revealed that bacterial OMVs/MVs could (1) interact with immune cells to regulate inflammatory responses, (2) transport virulence factors (eg, enzymes, DNA and small RNAs) to host cells and result in cell injury, (3) enhance barrier function by stimulating the expression of tight junction proteins in intestinal epithelial cells, (4) upregulate the expression of endothelial cell adhesion molecules, and (5) serve as natural nanocarriers for immunogenic antigens, enzyme support and drug delivery. In addition, OMVs/MVs can enter the systemic circulation and induce a variety of immunological and metabolic responses. This review highlights the recent advances in the understanding of OMV/MV biogenesis and their compositional remodeling. In addition, interactions between OMVs/MVs and various types of mammalian cells (ie, immune cells, epithelial cells, and endothelial cells) and their pathological/preventive effects on infectious/inflammatory diseases are summarized. Finally, methods for engineering OMVs/MVs and their therapeutic potential are discussed.
Collapse
Affiliation(s)
- You-jiang Yu
- Medical College of Yangzhou Polytechnic College, Yangzhou 225009, China
| | - Xiao-hong Wang
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
43
|
Chelakkot C, Choi Y, Kim DK, Park HT, Ghim J, Kwon Y, Jeon J, Kim MS, Jee YK, Gho YS, Park HS, Kim YK, Ryu SH. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp Mol Med 2018; 50:e450. [PMID: 29472701 PMCID: PMC5903829 DOI: 10.1038/emm.2017.282] [Citation(s) in RCA: 517] [Impact Index Per Article: 73.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/09/2017] [Accepted: 09/11/2017] [Indexed: 12/18/2022] Open
Abstract
The gut microbiota has an important role in the gut barrier, inflammation and metabolic functions. Studies have identified a close association between the intestinal barrier and metabolic diseases, including obesity and type 2 diabetes (T2D). Recently, Akkermansia muciniphila has been reported as a beneficial bacterium that reduces gut barrier disruption and insulin resistance. Here we evaluated the role of A. muciniphila-derived extracellular vesicles (AmEVs) in the regulation of gut permeability. We found that there are more AmEVs in the fecal samples of healthy controls compared with those of patients with T2D. In addition, AmEV administration enhanced tight junction function, reduced body weight gain and improved glucose tolerance in high-fat diet (HFD)-induced diabetic mice. To test the direct effect of AmEVs on human epithelial cells, cultured Caco-2 cells were treated with these vesicles. AmEVs decreased the gut permeability of lipopolysaccharide-treated Caco-2 cells, whereas Escherichia coli-derived EVs had no significant effect. Interestingly, the expression of occludin was increased by AmEV treatment. Overall, these results imply that AmEVs may act as a functional moiety for controlling gut permeability and that the regulation of intestinal barrier integrity can improve metabolic functions in HFD-fed mice.
Collapse
Affiliation(s)
- Chaithanya Chelakkot
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Youngwoo Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Dae-Kyum Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hyun T Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jaewang Ghim
- NovaCell Technology Inc., Pohang, Republic of Korea
| | - Yonghoon Kwon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jinseong Jeon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Min-Seon Kim
- Asan Institute of Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Koo Jee
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Yong S Gho
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon-si, Republic of Korea
| | | | - Sung H Ryu
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea.,Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| |
Collapse
|
44
|
Abstract
Outer Membrane Vesicles (OMVs) of Gram-negative bacteria are spherical membrane-enclosed entities of endocytic origin. Reported in the consortia of different bacterial species, production of OMVs into extracellular milieu seems essential for their survival. Enriched with bioactive proteins, toxins, and virulence factors, OMVs play a critical role in the bacteria-bacteria and bacteria-host interactions. Emergence of OMVs as distinct cellular entities helps bacteria in adaptating to diverse niches, in competing with other bacteria to protect members of producer species and more importantly play a crucial role in host-pathogen interaction. Composition of OMV, their ability to modulate host immune response, along with coordinated secretion of bacterial effector proteins, endows them with the armory, which can withstand hostile environments. Study of the OMV production under natural and diverse stress conditions has broadened the horizons, and also opened new frontiers in delineating the molecular machinery involved in disease pathogenesis. Playing diverse biological and pathophysiological functions, OMVs hold a great promise in enabling resurgence of bacterial diseases, in concomitance with the steep decline in the efficiency of antibiotics. Having multifaceted role, their emergence as a causative agent for a series of infectious diseases increases the probability for their exploitation in the development of effective diagnostic tools and as vaccines against diverse pathogenic species of Gram-negative origin.
Collapse
Affiliation(s)
- Arif Tasleem Jan
- Department of Medical Biotechnology, Yeungnam UniversityGyeongsan, South Korea
| |
Collapse
|
45
|
Guerrero-Mandujano A, Hernández-Cortez C, Ibarra JA, Castro-Escarpulli G. The outer membrane vesicles: Secretion system type zero. Traffic 2017; 18:425-432. [DOI: 10.1111/tra.12488] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Andrea Guerrero-Mandujano
- Laboratorio de Bacteriología Médica, Departamento de Microbiología; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional; Ciudad de México México
| | - Cecilia Hernández-Cortez
- Laboratorio de Bioquímica Microbiana, Departamento de Microbiología; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional; Ciudad de México México
| | - Jose Antonio Ibarra
- Laboratorio de Genética Microbiana, Departamento de Microbiología; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional; Ciudad de México México
| | - Graciela Castro-Escarpulli
- Laboratorio de Bacteriología Médica, Departamento de Microbiología; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional; Ciudad de México México
| |
Collapse
|
46
|
Affiliation(s)
- Daniela F Hozbor
- a Laboratorio VacSal , Instituto de Biotecnología y Biología Molecular, Facultad de Ciencias Exactas, Universidad Nacional de La Plata , La Plata , Argentina
| |
Collapse
|
47
|
Kim OY, Lee J, Gho YS. Extracellular vesicle mimetics: Novel alternatives to extracellular vesicle-based theranostics, drug delivery, and vaccines. Semin Cell Dev Biol 2016; 67:74-82. [PMID: 27916566 DOI: 10.1016/j.semcdb.2016.12.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/21/2016] [Accepted: 12/01/2016] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles are nano-sized spherical bilayered proteolipids encasing various components. Cells of all domains of life actively release these vesicles to the surroundings including various biological fluids. These extracellular vesicles are known to play pivotal roles in numerous pathophysiological functions. Extracellular vesicles have distinct characteristics, like high biocompatibility, safety, and nano-sized diameters that allow efficient drug loading capacity and long blood circulation half-life. These characteristics of extracellular vesicles have engrossed many scientists to harness them as new tools for novel delivery systems. This review will highlight the current state of the arts and problems of such extracellular vesicle-based theranostics, drug delivery and vaccines, and introduce "extracellular vesicle mimetics" as the novel alternative of extracellular vesicles. We hope to provide insights into the potential of extracellular vesicle mimetics as superior substitute to the natural extracellular vesicles that can be applied to theranostics, drug delivery, and vaccines against various diseases.
Collapse
Affiliation(s)
- Oh Youn Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jaewook Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea.
| |
Collapse
|
48
|
Lee J, Kim OY, Gho YS. Proteomic profiling of Gram-negative bacterial outer membrane vesicles: Current perspectives. Proteomics Clin Appl 2016; 10:897-909. [PMID: 27480505 DOI: 10.1002/prca.201600032] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 06/30/2016] [Accepted: 07/29/2016] [Indexed: 12/18/2022]
Abstract
Outer membrane vesicles (OMVs) are extracellular vesicles derived from Gram-negative bacteria. Recent progress in the studies of Gram-negative bacterial extracellular vesicles implies that OMVs may function as intercellular communicasomes in bacteria-bacteria and bacteria-host interactions. Current MS-based high-throughput proteomic analyses of Gram-negative bacterial OMVs have identified thousands of vesicular proteins and provided clues to reveal the biogenesis and pathophysiological functions of Gram-negative bacterial OMVs. The future directions of proteomics of Gram-negative bacterial OMVs may include the isolation strategy of Gram-negative bacterial OMVs to thoroughly exclude nonvesicular contaminants and proteomics of Gram-negative bacterial OMVs derived from diverse conditions as well as body fluids of bacterium-infected hosts. We hope this review will shed light on future research in this emerging field of proteomics of extracellular vesicles derived from Gram-negative bacteria and contribute to the development of OMV-based diagnostic tools and effective vaccines.
Collapse
Affiliation(s)
- Jaewook Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Oh Youn Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
49
|
Duell BL, Su YC, Riesbeck K. Host-pathogen interactions of nontypeable Haemophilus influenzae: from commensal to pathogen. FEBS Lett 2016; 590:3840-3853. [PMID: 27508518 DOI: 10.1002/1873-3468.12351] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/01/2016] [Accepted: 08/05/2016] [Indexed: 11/09/2022]
Abstract
Nontypeable Haemophilus influenzae (NTHi) is a commensal microbe often isolated from the upper and lower respiratory tract. This bacterial species can cause sinusitis, acute otitis media in preschool children, exacerbations in patients suffering from chronic obstructive pulmonary disease, as well as conjunctivitis and bacteremia. Since the introduction of a vaccine against H. influenzae serotype b in the 1990s, the burden of H. influenzae-related infections has been increasingly dominated by NTHi. Understanding the ability of NTHi to cause infection is currently an expanding area of study. NTHi is able to exert differential binding to the host tissue through the use of a broad range of adhesins. NTHi survival in the host is multifaceted, that is, using virulence factors involved in complement resistance, biofilm, modified immunoglobulin responses, and, finally, formation and utilization of host proteins as a secondary strategy of increasing the adhesive ability.
Collapse
Affiliation(s)
- Benjamin Luke Duell
- Clinical Microbiology, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Yu-Ching Su
- Clinical Microbiology, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
50
|
De Geyter J, Tsirigotaki A, Orfanoudaki G, Zorzini V, Economou A, Karamanou S. Protein folding in the cell envelope of Escherichia coli. Nat Microbiol 2016; 1:16107. [PMID: 27573113 DOI: 10.1038/nmicrobiol.2016.107] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/02/2016] [Indexed: 11/09/2022]
Abstract
While the entire proteome is synthesized on cytoplasmic ribosomes, almost half associates with, localizes in or crosses the bacterial cell envelope. In Escherichia coli a variety of mechanisms are important for taking these polypeptides into or across the plasma membrane, maintaining them in soluble form, trafficking them to their correct cell envelope locations and then folding them into the right structures. The fidelity of these processes must be maintained under various environmental conditions including during stress; if this fails, proteases are called in to degrade mislocalized or aggregated proteins. Various soluble, diffusible chaperones (acting as holdases, foldases or pilotins) and folding catalysts are also utilized to restore proteostasis. These responses can be general, dealing with multiple polypeptides, with functional overlaps and operating within redundant networks. Other chaperones are specialized factors, dealing only with a few exported proteins. Several complex machineries have evolved to deal with binding to, integration in and crossing of the outer membrane. This complex protein network is responsible for fundamental cellular processes such as cell wall biogenesis; cell division; the export, uptake and degradation of molecules; and resistance against exogenous toxic factors. The underlying processes, contributing to our fundamental understanding of proteostasis, are a treasure trove for the development of novel antibiotics, biopharmaceuticals and vaccines.
Collapse
Affiliation(s)
- Jozefien De Geyter
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Bacteriology, B-3000 Leuven, Belgium
| | - Alexandra Tsirigotaki
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Bacteriology, B-3000 Leuven, Belgium
| | - Georgia Orfanoudaki
- Institute of Molecular Biology and Biotechnology, FORTH and Department of Biology, University of Crete, PO Box 1385, GR-711 10 Iraklio, Crete, Greece
| | - Valentina Zorzini
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Bacteriology, B-3000 Leuven, Belgium
| | - Anastassios Economou
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Bacteriology, B-3000 Leuven, Belgium.,Institute of Molecular Biology and Biotechnology, FORTH and Department of Biology, University of Crete, PO Box 1385, GR-711 10 Iraklio, Crete, Greece
| | - Spyridoula Karamanou
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Bacteriology, B-3000 Leuven, Belgium
| |
Collapse
|