1
|
Waugh S, Cameron CE. Syphilis vaccine development: Aligning vaccine design with manufacturing requirements. Hum Vaccin Immunother 2024; 20:2399915. [PMID: 39262177 PMCID: PMC11404580 DOI: 10.1080/21645515.2024.2399915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Syphilis, caused by Treponema pallidum subsp. pallidum, is a global health concern with increasing rates worldwide. Current prevention strategies, including screen-and-treat approaches, are not sufficient to resolve rising infection rates, emphasizing the need for a vaccine. Developing a syphilis vaccine necessitates a range of cross-disciplinary considerations, including essential disease-specific protection, technical requirements, economic feasibility, manufacturing constraints, public acceptance, equitable vaccine access, alignment with global public vaccination programs, and identification of essential populations to be vaccinated to achieve herd immunity. Central to syphilis vaccine development is prioritization of global vaccine availability, including access in low- to middle-income settings. Various vaccine platforms, including subunit, virus-like particle (VLP), mRNA, and outer membrane vesicle (OMV) vaccines, present both advantages and challenges. The proactive consideration of both manufacturing feasibility and efficacy throughout the pre-clinical research and development stages is essential for producing an efficacious, inexpensive, and scalable syphilis vaccine to address the growing global health burden caused by this disease.
Collapse
Affiliation(s)
- Sean Waugh
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
| | - Caroline E. Cameron
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
- Department of Medicine, Division of Allergy and Infectious Disease, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Duan Z, Song J, Zhang M, Zhang Z, Li N, Fu Y, Sun Z, Lu T, Li S, Cao M, Wang Q, Sun C, Wang X. Effects of Yersinia pseudotuberculosis outer membrane vesicles on Pseudomonas aeruginosa antigens immune response. PLoS One 2024; 19:e0310652. [PMID: 39705292 DOI: 10.1371/journal.pone.0310652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/04/2024] [Indexed: 12/22/2024] Open
Abstract
Outer membrane vesicles (OMVs) are immunogenic self-adjuvanting vesicles produced by Gram-negative bacteria such as Pseudomonas aeruginosa and Yersinia pseudotuberculosis. While the effects of OMVs on different antigens immune stimulation are not clear. In this study, we constructed recombinant Yersinia pseudotuberculosis ΔlpxL strain,with pBlue-PcrV and pBlue-OprF/I, and then purified ΔlpxL rOMVPcrV (rOMVyp2P)and ΔlpxL rOMVOprF/I (rOMVyp2F) and analyzed its effect on immune response and protection against Pseudomonas aeruginosa PAO1 infection. The results showed that OMV assists in eliciting similar humoral immune responses to PcrV and OprF/I antigens. ΔlpxL rOMVPcrV and ΔlpxL rOMVOprF/I elicited Th1/Th2 balanced immune response, and higher IgM and IgA antibodies.However, there are differences in immune protection for the pulmonary. The survival rate of mice in ΔlpxL rOMVPcrV group was 20%, which was significantly better than that in ΔlpxL rOMVOprF/I group. ΔlpxL OMVPcrV is better cooperation for Pseudomonas immune protection in lung.
Collapse
Affiliation(s)
- Zhongxu Duan
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
- State Key Laboratory of Black Soils Conservation and Utilization, Key Laboratory of Wetland Ecology and Environment Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, China
| | - Jingqi Song
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Mingru Zhang
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Zhe Zhang
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Nan Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuqin Fu
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Zhe Sun
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Tiancheng Lu
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Siyuan Li
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Mingyue Cao
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Qingyu Wang
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Chunhui Sun
- Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Xiuran Wang
- Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun, China
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Huaiyin District, Jinan, China
| |
Collapse
|
3
|
Lusta KA, Churov AV, Beloyartsev DF, Golovyuk AL, Lee AA, Sukhorukov VN, Orekhov AN. The two coin sides of bacterial extracellular membrane nanovesicles: atherosclerosis trigger or remedy. DISCOVER NANO 2024; 19:179. [PMID: 39532781 PMCID: PMC11557815 DOI: 10.1186/s11671-024-04149-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Among the numerous driving forces that cause the atherosclerotic cardiovascular disease (ASCVD), pathogenic bacterial extracellular membrane nanovesicles (BEMNs) containing toxins and virulence factors appear to be the key trigger of inflammation and atherogenesis, the major processes involved in the pathogenesis of ASCVD. Since BEMNs are the carriers of nanosized biomolecules to distant sites, they are now being considered as a novel drug delivery system. Nowadays, many therapeutic strategies are used to treat ASCVD. However, the conventional anti-atherosclerotic therapies are not effective enough. This primarily due to the inefficiency of non-targeted drug delivery systems to tissue affected areas, which, in turn, leads to numerous side effects, as well as faulty pharmacokinetics. In this regard, nanomedicine methods using nanoparticles (NPs) as targeted drug delivery vehicles proved to be extremely useful. Bioengineered BEMNs equipped with disease-specific ligand moieties and loaded with corresponding drugs represent a promising tool in nanomedicine, which can be used as a novel drug delivery system for a successful therapy of ASCVD. In this review, we outline the involvement of pathogenic BEMNs in the triggering of ASCVD, the conventional therapeutic strategies for the treatment of ASCVD, and the recent trends in nanomedicine using BEMNs and NPs as a vehicle for targeted drug delivery.
Collapse
Affiliation(s)
- Konstantin A Lusta
- Institute for Atherosclerosis Research, Ltd, Osennyaya Street 4-1-207, Moscow, Russia, 121609.
| | - Alexey V Churov
- Institute on Aging Research, Russian Gerontology Clinical Research Center, Pirogov Russian National Research Medical University, Moscow, Russia, 129226
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| | - Dmitry F Beloyartsev
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Alexander L Golovyuk
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Arthur A Lee
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
| | - Vasily N Sukhorukov
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| | - Alexander N Orekhov
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| |
Collapse
|
4
|
Li Y, Quan Y, Chen P, Zhuge X, Qin T, Chen S, Peng D, Liu X. Development of High-Production Bacterial Biomimetic Vesicles for Inducing Mucosal Immunity Against Avian Pathogenic Escherichia coli. Int J Mol Sci 2024; 25:12055. [PMID: 39596124 PMCID: PMC11593933 DOI: 10.3390/ijms252212055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
To evaluate the immunoprotective effect of bacterial biomimetic vesicles (BBVs) against avian pathogenic Escherichia coli (APEC), a ΔtolA J11 mutant strain was generated by deleting the tolA gene in the low pathogenic O78 serotype J11 strain. The total protein content of outer membrane vesicles (OMVs) derived from the ΔtolA J11 strain exhibited a sevenfold increase compared to the wild-type strain. Additionally, high-pressure homogenization technology was employed to produce BBVs, resulting in a sixfold increase in total protein content compared to spontaneously secreted OMVs from ΔtolA J11. The immunogenicity of both OMVs and BBVs was assessed through intranasal or intramuscular immunization in specific pathogen-free (SPF) chickens. Results demonstrated that intranasal immunization with OMVs or BBVs in chickens elicited specific IgY antibodies against APEC outer membrane proteins and specific sIgA antibodies in the nasal cavity and trachea, as well as a significant increase in the proliferation response of chicken peripheral blood lymphocytes. The bacterial load in the blood and various organs of the challenged chickens were significantly reduced, resulting in a 66.67% and 58.30% survival rate against a high pathogenic serotype O78 strain challenge, while the control group exhibited only a 16.67% survival rate. The intramuscular immunization with OMVs or BBVs in chickens only induced specific IgY antibodies, with a survival rate of only 33.33% for challenged chickens during the same period. Therefore, intranasal vaccination of the highly productive BBVs is capable of eliciting an immune response similar to that of OMVs and providing protection against APEC infection, thus offering innovative insights for the advancement of APEC vaccines.
Collapse
Affiliation(s)
- Yue Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou 225009, China; (Y.L.); (Y.Q.); (P.C.); (T.Q.); (X.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou 225009, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou 225009, China
| | - Yuji Quan
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou 225009, China; (Y.L.); (Y.Q.); (P.C.); (T.Q.); (X.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou 225009, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou 225009, China
| | - Peng Chen
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou 225009, China; (Y.L.); (Y.Q.); (P.C.); (T.Q.); (X.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou 225009, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou 225009, China
| | - Xiangkai Zhuge
- School of Public Health, Nantong University, Nantong 226019, China;
| | - Tao Qin
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou 225009, China; (Y.L.); (Y.Q.); (P.C.); (T.Q.); (X.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou 225009, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou 225009, China
| | - Sujuan Chen
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou 225009, China; (Y.L.); (Y.Q.); (P.C.); (T.Q.); (X.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou 225009, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou 225009, China
| | - Daxin Peng
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou 225009, China; (Y.L.); (Y.Q.); (P.C.); (T.Q.); (X.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou 225009, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou 225009, China
| | - Xiufan Liu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou 225009, China; (Y.L.); (Y.Q.); (P.C.); (T.Q.); (X.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou 225009, China
- Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou 225009, China
| |
Collapse
|
5
|
Henriquez T, Santoro F, Medaglini D, Pallecchi L, Clemente I, Bonechi C, Magnani A, Paccagnini E, Gentile M, Lupetti P, Marvasi M, Pini A, Bracci L, Falciani C. Analysis of the utility of a rapid vesicle isolation method for clinical strains of Pseudomonas aeruginosa. Microbiol Spectr 2024; 12:e0064924. [PMID: 39248554 PMCID: PMC11448148 DOI: 10.1128/spectrum.00649-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024] Open
Abstract
Pseudomonas aeruginosa, a pathogen capable of causing diseases ranging from mild to life-threatening, has a large arsenal of virulence factors. Notably, extracellular vesicles have emerged as significant players in the pathogenesis of this organism. However, the full range of their functions is still being studied, and difficulties related to vesicle purification (long protocols, low yields, and specialized instruments) have become a major obstacle for their characterization. In this context, the utility of rapid new methods of vesicle isolation from clinical strains is still unknown. Here, we analyze the utility of the ExoBacteria OMV isolation kit for a collection of clinical strains of P. aeruginosa. We first phenotypically characterized 15 P. aeruginosa strains to ensure that our samples were heterogeneous. We then determined the best conditions for purifying vesicles from P. aeruginosa PAO1 reference strain by the rapid method and used them to isolate vesicles from clinical strains. Our results indicated that M9 minimal medium is the best for obtaining high purity with the rapid isolation kit. Although we were able to isolate vesicles from at least four strains, the low yield and the large number of strains with unpurifiable vesicles showed that the kit was not practical or convenient for clinical strains. Our findings suggest that although fast procedures for vesicle purification can be of great utility for Escherichia coli, the more complex phenotypes of clinical isolates of P. aeruginosa are a challenge for these protocols and new alternatives/optimizations need to be developed.IMPORTANCEPseudomonas aeruginosa is recognized as an opportunistic pathogen in humans and animals. It can effectively colonize various environments thanks to a large set of virulence factors that include extracellular vesicles. Different methods were recently developed to reduce the time and effort associated with vesicle purification. However, the utility of rapid vesicle isolation methods for clinical strains of P. aeruginosa (which are recognized as being highly diverse) is not yet known. In this context, we analyzed the utility of the ExoBacteria OMV Isolation kit for vesicle purification in P. aeruginosa clinical strains. Our findings showed that the kit does not seem to be convenient for research on clinical strains due to low vesicle recovery. Our results underscore the importance of developing new rapid vesicle purification protocols/techniques for specific clinical phenotypes.
Collapse
Affiliation(s)
- Tania Henriquez
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesco Santoro
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Donata Medaglini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Lucia Pallecchi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ilaria Clemente
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Claudia Bonechi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Agnese Magnani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | | | - Pietro Lupetti
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Alessandro Pini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Luisa Bracci
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Chiara Falciani
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
6
|
Sauvat L, Verhoeven PO, Gagnaire J, Berthelot P, Paul S, Botelho-Nevers E, Gagneux-Brunon A. Vaccines and monoclonal antibodies to prevent healthcare-associated bacterial infections. Clin Microbiol Rev 2024; 37:e0016022. [PMID: 39120140 PMCID: PMC11391692 DOI: 10.1128/cmr.00160-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
SUMMARYHealthcare-associated infections (HAIs) represent a burden for public health with a high prevalence and high death rates associated with them. Pathogens with a high potential for antimicrobial resistance, such as ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) and Clostridioides difficile, are responsible for most HAIs. Despite the implementation of infection prevention and control intervention, globally, HAIs prevalence is stable and they are mainly due to endogenous pathogens. It is undeniable that complementary to infection prevention and control measures, prophylactic approaches by active or passive immunization are needed. Specific groups at-risk (elderly people, chronic condition as immunocompromised) and also healthcare workers are key targets. Medical procedures and specific interventions are known to be at risk of HAIs, in addition to hospital environmental exposure. Vaccines or monoclonal antibodies can be seen as attractive preventive approaches for HAIs. In this review, we present an overview of the vaccines and monoclonal antibodies in clinical development for prevention of the major bacterial HAIs pathogens. Based on the current state of knowledge, we look at the challenges and future perspectives to improve prevention by these means.
Collapse
Affiliation(s)
- Léo Sauvat
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Paul O Verhoeven
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Agents and Hygiene, University-Hospital of Saint-Etienne, Saint-Etienne, France
| | - Julie Gagnaire
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Philippe Berthelot
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Amandine Gagneux-Brunon
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
7
|
Baker S, Krishna A, Higham S, Naydenova P, O'Leary S, Scott JB, Harcourt K, Forrest S, Goulding D, Thi Nguyen TN, Toan ND, Alekseeva E, Zhou Q, Andreozzi I, Sobotic B, Craig H, Wong V, Forrest-Owen N, Sanchez DM, Pearce C, Roberts L, Watson S, Clare S, Torok ME, Dougan G, Kellam P, Tregoning JS, Reece ST. Exploiting human immune repertoire transgenic mice for protective monoclonal antibodies against antimicrobial resistant Acinetobacter baumannii. Nat Commun 2024; 15:7979. [PMID: 39266557 PMCID: PMC11392949 DOI: 10.1038/s41467-024-52357-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 09/04/2024] [Indexed: 09/14/2024] Open
Abstract
The use of monoclonal antibodies for the control of drug resistant nosocomial bacteria may alleviate a reliance on broad spectrum antimicrobials for treatment of infection. We identify monoclonal antibodies that may prevent infection caused by carbapenem resistant Acinetobacter baumannii. We use human immune repertoire mice (Kymouse platform mice) as a surrogate for human B cell interrogation to establish an unbiased strategy to probe the antibody-accessible target landscape of clinically relevant A. baumannii. After immunisation of the Kymouse platform mice with A. baumannii derived outer membrane vesicles (OMV) we identify 297 antibodies and analyse 26 of these for functional potential. These antibodies target lipooligosaccharide (OCL1), the Oxa-23 protein, and the KL49 capsular polysaccharide. We identify a single monoclonal antibody (mAb1416) recognising KL49 capsular polysaccharide to demonstrate prophylactic in vivo protection against a carbapenem resistant A. baumannii lineage associated with neonatal sepsis mortality in Asia. Our end-to-end approach identifies functional monoclonal antibodies with prophylactic potential against major lineages of drug resistant bacteria accounting for phylogenetic diversity and clinical relevance without existing knowledge of a specific target antigen. Such an approach might be scaled for a additional clinically important bacterial pathogens in the post-antimicrobial era.
Collapse
Affiliation(s)
- Stephen Baker
- University of Cambridge School of Clinical Medicine Cambridge Biomedical Campus, Cambridge, UK
- IAVI, Chelsea and Westminster Hospital, London, UK
| | | | - Sophie Higham
- Department of Infectious Disease, Imperial College London, St Marys Campus, Norfolk Place, London, UK
| | - Plamena Naydenova
- University of Cambridge School of Clinical Medicine Cambridge Biomedical Campus, Cambridge, UK
| | - Siobhan O'Leary
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK
| | | | - Katherine Harcourt
- University of Cambridge School of Clinical Medicine Cambridge Biomedical Campus, Cambridge, UK
| | - Sally Forrest
- University of Cambridge School of Clinical Medicine Cambridge Biomedical Campus, Cambridge, UK
| | - David Goulding
- Pathogens and Microbes Programme, Wellcome Sanger Institute, Cambridge, UK
| | - To Nguyen Thi Nguyen
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Nguyen Duc Toan
- Neonatal Intensive Care Unit, Children's Hospital 1, Ho Chi Minh City, Vietnam
| | | | - Qingqing Zhou
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK
| | - Ilaria Andreozzi
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK
| | - Barbara Sobotic
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK
| | - Hannah Craig
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK
| | - Vivian Wong
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK
| | | | | | - Claire Pearce
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK
| | - Leah Roberts
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Simon Watson
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK
| | - Simon Clare
- University of Cambridge School of Clinical Medicine Cambridge Biomedical Campus, Cambridge, UK
| | - Mili Estee Torok
- University of Cambridge School of Clinical Medicine Cambridge Biomedical Campus, Cambridge, UK
| | - Gordon Dougan
- University of Cambridge School of Clinical Medicine Cambridge Biomedical Campus, Cambridge, UK
| | - Paul Kellam
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK
- Department of Infectious Disease, Imperial College London, St Marys Campus, Norfolk Place, London, UK
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, St Marys Campus, Norfolk Place, London, UK
| | - Stephen T Reece
- Kymab, a Sanofi Company, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
8
|
Honorato L, Bonilla JJA, Valdez AF, Frases S, Araújo GRDS, Sabino ALRDN, da Silva NM, Ribeiro L, Ferreira MDS, Kornetz J, Rodrigues ML, Cunningham I, Gow NAR, Gacser A, Guimarães AJ, Dutra FF, Nimrichter L. Toll-like receptor 4 (TLR4) is the major pattern recognition receptor triggering the protective effect of a Candida albicans extracellular vesicle-based vaccine prototype in murine systemic candidiasis. mSphere 2024; 9:e0046724. [PMID: 39037263 PMCID: PMC11351041 DOI: 10.1128/msphere.00467-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024] Open
Abstract
Systemic candidiasis remains a significant public health concern worldwide, with high mortality rates despite available antifungal drugs. Drug-resistant strains add to the urgency for alternative therapies. In this context, vaccination has reemerged as a prominent immune-based strategy. Extracellular vesicles (EVs), nanosized lipid bilayer particles, carry a diverse array of native fungal antigens, including proteins, nucleic acids, lipids, and glycans. Previous studies from our laboratory demonstrated that Candida albicans EVs triggered the innate immune response, activating bone marrow-derived dendritic cells (BMDCs) and potentially acting as a bridge between innate and adaptive immunity. Vaccination with C. albicans EVs induced the production of specific antibodies, modulated cytokine production, and provided protection in immunosuppressed mice infected with lethal C. albicans inoculum. To elucidate the mechanisms underlying EV-induced immune activation, our study investigated pathogen-associated molecular patterns (PAMPs) and pattern recognition receptors (PRRs) involved in EVs-phagocyte engagement. EVs from wild-type and mutant C. albicans strains with truncated mannoproteins were compared for their ability to stimulate BMDCs. Our findings revealed that EV decoration with O- and N-linked mannans and the presence of β-1,3-glucans and chitin oligomers may modulate the activation of specific PRRs, in particular Toll-like receptor 4 (TLR4) and dectin-1. The protective effect of vaccination with wild-type EVs was found to be dependent on TLR4. These results suggest that fungal EVs can be harnessed in vaccine formulations to selectively activate PRRs in phagocytes, offering potential avenues for combating or preventing candidiasis.IMPORTANCESystemic candidiasis is a serious global health concern with high mortality rates and growing drug resistance. Vaccination offers a promising solution. A unique approach involves using tiny lipid-coated particles called extracellular vesicles (EVs), which carry various fungal components. Previous studies found that Candida albicans EVs activate the immune response and may bridge the gap between innate and adaptive immunity. To understand this better, we investigated how these EVs activate immune cells. We demonstrated that specific components on EV surfaces, such as mannans and glucans, interact with receptors on immune cells, including Toll-like receptor 4 (TLR4) and dectin-1. Moreover, vaccinating with these EVs led to strong immune responses and full protection in mice infected with Candida. This work shows how harnessing fungal EVs might lead to effective vaccines against candidiasis.
Collapse
Affiliation(s)
- Leandro Honorato
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jhon J. Artunduaga Bonilla
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro F. Valdez
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susana Frases
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filhos (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
| | - Glauber Ribeiro de Sousa Araújo
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filhos (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Natalia Martins da Silva
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Ribeiro
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marina da Silva Ferreira
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Julio Kornetz
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcio L. Rodrigues
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Carlos Chagas (ICC), Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Brazil
| | - Iain Cunningham
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Neil A. R. Gow
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Attila Gacser
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Allan J. Guimarães
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Fabianno F. Dutra
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Nimrichter
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Meng Y, Kong C, Ma Y, Sun J, Zhang G. Bacterial outer membrane vesicles in the fight against cancer. Chin Med J (Engl) 2024:00029330-990000000-01174. [PMID: 39118214 PMCID: PMC11407815 DOI: 10.1097/cm9.0000000000003234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Indexed: 08/10/2024] Open
Abstract
ABSTRACT Bacterial outer membrane vesicles (OMVs) are diminutive vesicles naturally released by Gram-negative bacteria. These vesicles possess distinctive characteristics that attract attention for their potential use in drug administration and immunotherapy in cancer treatment. Therapeutic medicines may be delivered via OMVs directly to the tumor sites, thereby minimizing exposure to healthy cells and lowering the risk of systemic toxicity. Furthermore, the activation of the immune system by OMVs has been demonstrated to facilitate the recognition and elimination of cancer cells, which makes them a desirable tool for immunotherapy. They can also be genetically modified to carry specific antigens, immunomodulatory compounds, and small interfering RNAs, enhancing the immune response to cancerous cells and silencing genes associated with disease progression. Combining OMVs with other cancer treatments like chemotherapy and radiation has shown promising synergistic effects. This review highlights the crucial role of bacterial OMVs in cancer, emphasizing their potential as vectors for novel cancer targeted therapies. As researchers delve deeper into the complexities of these vesicles and their interactions with tumors, there is a growing sense of optimism that this avenue of study will bring positive outcomes and renewed hope to cancer patients in the foreseeable future.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Cuicui Kong
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yushu Ma
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Jing Sun
- Department of Biobank, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| |
Collapse
|
10
|
Nadalian B, Nadalian B, Zali MR, Yadegar A. Outer Membrane Vesicles Derived from Adherent-Invasive Escherichia coli Induce Inflammatory Response and Alter the Gene Expression of Junction-Associated Proteins in Human Intestinal Epithelial Cells. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:2701675. [PMID: 38826676 PMCID: PMC11142853 DOI: 10.1155/2024/2701675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/02/2024] [Accepted: 05/10/2024] [Indexed: 06/04/2024]
Abstract
Adherent-invasive Escherichia coli (AIEC) pathobionts, which are characterized by their ability to adhere to and invade intestinal epithelial cells, are associated with the etiopathogenesis of inflammatory bowel diseases (IBDs). Outer membrane vesicles (OMVs) released by AIEC strains can facilitate the interaction of these bacteria with host cells through delivering bacterial effectors. The aim of this study was to determine the ability of OMVs derived from AIEC strain LF82 to induce the host immune response, leading to production of proinflammatory cytokines and also altering the gene expression of junction-associated proteins in the human epithelial colorectal adenocarcinoma Caco-2 cell line. OMVs were extracted from AIEC strain LF82, and the cell viability of Caco-2 cells treated with these vesicles was assessed by MTT assay. The morphology and size distribution of vesicles were analyzed using transmission electron microscopy and dynamic light scattering, respectively. Gene expression of occludin, ZO-1, claudin-2, E-cadherin, TLR-2, and TLR-4 in response to OMVs was assessed in Caco-2 cells by RT-qPCR. Moreover, the secretion of IL-8 and TNF-α into the supernatant of Caco-2 cells upon treatment with OMVs was measured using ELISA. Our results demonstrated that OMVs upregulated the gene expression level of TLRs and also altered the gene expression level of junction-associated proteins. OMVs derived from AIEC may play a major role in the promotion of intestinal inflammation and epithelial barrier dysfunction. However, further investigations are needed to elucidate the putative role of OMVs in the pathogenesis of AIEC and IBD.
Collapse
Affiliation(s)
- Bahareh Nadalian
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Banafsheh Nadalian
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
David S, Sophia I, Anbazhagan S, Karikalan M, Saravanan R, Viswas KN, Thomas P, Chaudhuri P. Outer membrane vesicles as nanovaccine candidates against pathogenic Leptospira in experimental Guinea pig model. Biologicals 2024; 86:101764. [PMID: 38537360 DOI: 10.1016/j.biologicals.2024.101764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 02/15/2024] [Accepted: 03/13/2024] [Indexed: 06/11/2024] Open
Abstract
Leptospira interrogans serovar Hardjo is a long slender bacterium of size 0.1-0.3 μm × 5-50 μm. It is one of the major causes of bovine leptospirosis and is of economical importance because of the reproductive failure, still birth, abortion, and reduced productivity in cattle. It is also a zoonotic disease-causing infection in humans characterized by headaches, fever, chills, sweats and myalgia, lethargy, aching joints, pulmonary haemorrhages, and death in severe cases. Control of the disease involves antibiotic therapy, management and vaccination, of which immunization is the cheapest and effective means of disease prevention. The present study was developed to isolate and characterize the outer membrane vesicles of Leptospira interrogans serovar Hardjo and to evaluate their vaccine potential in guinea pig model. The OMVs were isolated from the culture by sonication and ultracentrifugation. In transmission electron microscopy, the isolated OMVs appeared as small spherical structures of 50-200 nm size. In Western blot and indirect ELISA, antibodies specific to OMVs were observed as indicative of a good humoral immune response elicited by L. interrogans serovar Hardjo OMV. The OMV-based Leptospira vaccine was able to prevent kidney lesions and renal colonization compared to the control and bacterin vaccinated group as proven by histopathology and PCR.
Collapse
Affiliation(s)
- S David
- Division of Veterinary Bacteriology and Mycology, ICAR-Indian Veterinary Research Institute, Bareilly, UP, 243122, India
| | - Inbaraj Sophia
- Division of Veterinary Bacteriology and Mycology, ICAR-Indian Veterinary Research Institute, Bareilly, UP, 243122, India; ICAR-NRC on Meat, Hyderabad, Telangana-92, India.
| | - Subbaiyan Anbazhagan
- ICMR - National Animal Resource Facility for Biomedical Research, Hyderabad, India
| | - M Karikalan
- Centre for Wildlife Conservation, ICAR-Indian Veterinary Research Institute, Bareilly, UP, 243122, India
| | - R Saravanan
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, UP, 243122, India
| | - K Nagaleekar Viswas
- ICAR-Indian Veterinary Research Institute, Bengaluru Campus, Karnataka, India
| | - Prasad Thomas
- Division of Veterinary Bacteriology and Mycology, ICAR-Indian Veterinary Research Institute, Bareilly, UP, 243122, India
| | - Pallab Chaudhuri
- ICAR-Indian Veterinary Research Institute, Bengaluru Campus, Karnataka, India
| |
Collapse
|
12
|
van Schaik EJ, Fratzke AP, Gregory AE, Dumaine JE, Samuel JE. Vaccine development: obligate intracellular bacteria new tools, old pathogens: the current state of vaccines against obligate intracellular bacteria. Front Cell Infect Microbiol 2024; 14:1282183. [PMID: 38567021 PMCID: PMC10985213 DOI: 10.3389/fcimb.2024.1282183] [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: 08/23/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Obligate intracellular bacteria have remained those for which effective vaccines are unavailable, mostly because protection does not solely rely on an antibody response. Effective antibody-based vaccines, however, have been developed against extracellular bacteria pathogens or toxins. Additionally, obligate intracellular bacteria have evolved many mechanisms to subvert the immune response, making vaccine development complex. Much of what we know about protective immunity for these pathogens has been determined using infection-resolved cases and animal models that mimic disease. These studies have laid the groundwork for antigen discovery, which, combined with recent advances in vaccinology, should allow for the development of safe and efficacious vaccines. Successful vaccines against obligate intracellular bacteria should elicit potent T cell memory responses, in addition to humoral responses. Furthermore, they ought to be designed to specifically induce strong cytotoxic CD8+ T cell responses for protective immunity. This review will describe what we know about the potentially protective immune responses to this group of bacteria. Additionally, we will argue that the novel delivery platforms used during the Sars-CoV-2 pandemic should be excellent candidates to produce protective immunity once antigens are discovered. We will then look more specifically into the vaccine development for Rickettsiaceae, Coxiella burnetti, and Anaplasmataceae from infancy until today. We have not included Chlamydia trachomatis in this review because of the many vaccine related reviews that have been written in recent years.
Collapse
Affiliation(s)
- E J van Schaik
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
| | - A P Fratzke
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
- Charles River Laboratories, Reno, NV, United States
| | - A E Gregory
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Jennifer E Dumaine
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
| | - J E Samuel
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
- Department of Veterinary Pathobiology, School of Veterinary Medicine, Texas A&M University (TAMU), College Station, TX, United States
| |
Collapse
|
13
|
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
|
14
|
Wang G, Wang Y, Ma F. Exploiting bacterial-origin immunostimulants for improved vaccination and immunotherapy: current insights and future directions. Cell Biosci 2024; 14:24. [PMID: 38368397 PMCID: PMC10874560 DOI: 10.1186/s13578-024-01207-7] [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: 10/10/2023] [Accepted: 02/06/2024] [Indexed: 02/19/2024] Open
Abstract
Vaccination is a valid strategy to prevent and control newly emerging and reemerging infectious diseases in humans and animals. However, synthetic and recombinant antigens are poor immunogenic to stimulate efficient and protective host immune response. Immunostimulants are indispensable factors of vaccines, which can promote to trigger fast, robust, and long-lasting immune responses. Importantly, immunotherapy with immunostimulants is increasing proved to be an effective and promising treatment of cancer, which could enhance the function of the immune system against tumor cells. Pattern recognition receptors (PRRs) play vital roles in inflammation and are central to innate and adaptive immune responses. Toll-like receptors (TLRs)-targeting immunostimulants have become one of the hotspots in adjuvant research and cancer therapy. Bacterial-origin immunoreactive molecules are usually the ligands of PRRs, which could be fast recognized by PRRs and activate immune response to eliminate pathogens. Varieties of bacterial immunoreactive molecules and bacterial component-mimicking molecules have been successfully used in vaccines and clinical therapy so far. This work provides a comprehensive review of the development, current state, mechanisms, and applications of bacterial-origin immunostimulants. The exploration of bacterial immunoreactive molecules, along with their corresponding mechanisms, holds immense significance in deepening our understanding of bacterial pathogenicity and in the development of promising immunostimulants.
Collapse
Affiliation(s)
- Guangyu Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China
| | - Yongkang Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China
| | - Fang Ma
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China.
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China.
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, 225300, China.
| |
Collapse
|
15
|
Yang YS, Chen HJ, Chen XC, Tang HJ, Chang FJ, Huang YL, Pan YL, Kesavan DK, Chen HY, Shang HS, Kuo SC, Chen TL, Chiang MH. Elizabethkingia anophelis outer membrane vesicles as a novel vaccine candidate against infection: insights into immune response and potential for passive immunity. mSphere 2023; 8:e0040023. [PMID: 38014949 PMCID: PMC10732079 DOI: 10.1128/msphere.00400-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/13/2023] [Indexed: 11/29/2023] Open
Abstract
IMPORTANCE Elizabethkingia anophelis, a Gram-negative pathogen, causes infections such as bacteraemia, pneumonia, and neonatal meningitis. The pathogen resists most antimicrobial classes, making novel approaches urgently needed. In natural settings, Gram-negative bacteria secrete outer membrane vesicles (OMVs) that carry important molecules in the bacterial life cycle. These OMVs are enriched with proteins involved in virulence, survival, and carbohydrate metabolism, making them a promising source for vaccine development against the pathogen. This study investigated the efficacy of imipenem-induced OMVs (iOMVs) as a vaccine candidate against E. anophelis infection in a mouse pneumonia model. Mice immunized with iOMVs were completely protected during lethal-dose challenges. Passive immunization with hyperimmune sera and splenocytes conferred protection against lethal pneumonia. Further investigation is needed to understand the mechanisms underlying the protective effects of iOMV-induced passive immunity, such as the action on specific antibody subclasses or T cell subsets.
Collapse
Grants
- 109-2320-B-016-002-MY2, 110-2320-B-016-014, 111-2320-B-016-015, 112-2314-B-016-023, 112-2314-B-016-039, 112-2314-B-016-024-MY2 Ministry of Science and Technology, Taiwan (MOST)
- TSGH-E-111244, TSGH-E-112253 Tri-Service General Hospital (TSGH)
- CMNDMC11108, CMNDMC11206 Chi Mei Medical Center
- MND-MAB-110-049, MND-MAB-D-111072, MND-MAB-D-112115, MND-MAB-D-113078 MOD | Medical Affairs Bureau (MAB)
Collapse
Affiliation(s)
- Ya-Sung Yang
- Department of Internal Medicine, Division of Infectious Diseases and Tropical Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Jui Chen
- Department of Infectious Diseases, Chi Mei Medical Center, Tainan, Taiwan
| | - Xiao-Chun Chen
- Department and Graduate institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Jen Tang
- Department of Infectious Diseases, Chi Mei Medical Center, Tainan, Taiwan
| | - Fang-Ju Chang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yun-Ling Huang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Ling Pan
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Dinesh Kumar Kesavan
- School of Material Science, Nanyang Technological University, Singapore, Singapore
| | - Huan-Yuan Chen
- Inflammation Core Facility, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Sheng Shang
- Department of Pathology, Division of Clinical Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Chen Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Te-Li Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hsien Chiang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
16
|
Sadeghi L, Mohit E, Moallemi S, Ahmadi FM, Bolhassani A. Recent advances in various bio-applications of bacteria-derived outer membrane vesicles. Microb Pathog 2023; 185:106440. [PMID: 37931826 DOI: 10.1016/j.micpath.2023.106440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/12/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023]
Abstract
Outer membrane vesicles (OMVs) are spherical nanoparticles released from gram-negative bacteria. OMVs were originally classified into native 'nOMVs' (produced naturally from budding of bacteria) and non-native (produced by mechanical means). nOMVs and detergent (dOMVs) are isolated from cell supernatant without any detergent cell disruption techniques and through detergent extraction, respectively. Growth stages and conditions e.g. different stress factors, including temperature, nutrition deficiency, and exposure to hazardous chemical agents can affect the yield of OMVs production and OMVs content. Because of the presence of bacterial antigens, pathogen-associated molecular patterns (PAMPs), various proteins and the vesicle structure, OMVs have been developed in many biomedical applications. OMVs due to their size can be phagocytized by APCs, enter lymph vessels, transport antigens efficiently, and induce both T and B cells immune responses. Non-engineered OMVs have been frequently used as vaccines against different bacterial and viral infections, and various cancers. OMVs can also be used in combination with different antigens as an attractive vaccine adjuvant. Indeed, foreign antigens from target microorganisms can be trapped in the lumen of nonpathogenic vesicles or can be displayed on the surface through bacterial membrane protein to increase the immunogenicity of the antigens. In this review, different factors affecting OMV production including time of cultivation, growth media, stress conditions and genetic manipulations to enhance vesiculation will be described. Furthermore, recent advances in various biological applications of OMVs such as vaccine, drug delivery, cancer therapy, and enzyme carrier are discussed. Generally, the application of OMVs as vaccine carrier in three categories (i.e., non-engineered OMVs, OMVs as an adjuvant, recombinant OMVs (rOMVs)), as delivery system for small interfering RNA and therapeutic agents, and as enzymes carrier will be discussed.
Collapse
Affiliation(s)
- Leila Sadeghi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Samaneh Moallemi
- School of Biomedical Sciences, Faculty of Medicine, UNSW Sydney, NSW, 2052, Australia
| | | | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
17
|
Saeed U, Insaf RA, Piracha ZZ, Tariq MN, Sohail A, Abbasi UA, Fida Rana MS, Gilani SS, Noor S, Noor E, Waheed Y, Wahid M, Najmi MH, Fazal I. Crisis averted: a world united against the menace of multiple drug-resistant superbugs -pioneering anti-AMR vaccines, RNA interference, nanomedicine, CRISPR-based antimicrobials, bacteriophage therapies, and clinical artificial intelligence strategies to safeguard global antimicrobial arsenal. Front Microbiol 2023; 14:1270018. [PMID: 38098671 PMCID: PMC10720626 DOI: 10.3389/fmicb.2023.1270018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/03/2023] [Indexed: 12/17/2023] Open
Abstract
The efficacy of antibiotics and other antimicrobial agents in combating bacterial infections faces a grave peril in the form of antimicrobial resistance (AMR), an exceedingly pressing global health issue. The emergence and dissemination of drug-resistant bacteria can be attributed to the rampant overuse and misuse of antibiotics, leading to dire consequences such as organ failure and sepsis. Beyond the realm of individual health, the pervasive specter of AMR casts its ominous shadow upon the economy and society at large, resulting in protracted hospital stays, elevated medical expenditures, and diminished productivity, with particularly dire consequences for vulnerable populations. It is abundantly clear that addressing this ominous threat necessitates a concerted international endeavor encompassing the optimization of antibiotic deployment, the pursuit of novel antimicrobial compounds and therapeutic strategies, the enhancement of surveillance and monitoring of resistant bacterial strains, and the assurance of universal access to efficacious treatments. In the ongoing struggle against this encroaching menace, phage-based therapies, strategically tailored to combat AMR, offer a formidable line of defense. Furthermore, an alluring pathway forward for the development of vaccines lies in the utilization of virus-like particles (VLPs), which have demonstrated their remarkable capacity to elicit a robust immune response against bacterial infections. VLP-based vaccinations, characterized by their absence of genetic material and non-infectious nature, present a markedly safer and more stable alternative to conventional immunization protocols. Encouragingly, preclinical investigations have yielded promising results in the development of VLP vaccines targeting pivotal bacteria implicated in the AMR crisis, including Salmonella, Escherichia coli, and Clostridium difficile. Notwithstanding the undeniable potential of VLP vaccines, formidable challenges persist, including the identification of suitable bacterial markers for vaccination and the formidable prospect of bacterial pathogens evolving mechanisms to thwart the immune response. Nonetheless, the prospect of VLP-based vaccines holds great promise in the relentless fight against AMR, underscoring the need for sustained research and development endeavors. In the quest to marshal more potent defenses against AMR and to pave the way for visionary innovations, cutting-edge techniques that incorporate RNA interference, nanomedicine, and the integration of artificial intelligence are currently under rigorous scrutiny.
Collapse
Affiliation(s)
- Umar Saeed
- Clinical and Biomedical Research Center (CBRC) and Multidisciplinary Laboratories (MDL), Foundation University School of Health Sciences (FUSH), Foundation University Islamabad (FUI), Islamabad, Pakistan
| | - Rawal Alies Insaf
- Regional Disease Surveillance and Response Unit Sukkur, Sukkur, Sindh, Pakistan
| | - Zahra Zahid Piracha
- International Center of Medical Sciences Research (ICMSR), Islamabad, Pakistan
| | | | - Azka Sohail
- Central Park Teaching Hospital, Lahore, Pakistan
| | | | | | | | - Seneen Noor
- International Center of Medical Sciences Research (ICMSR), Islamabad, Pakistan
| | - Elyeen Noor
- International Center of Medical Sciences Research (ICMSR), Islamabad, Pakistan
| | - Yasir Waheed
- Office of Research, Innovation, and Commercialization (ORIC), Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Maryam Wahid
- Clinical and Biomedical Research Center (CBRC) and Multidisciplinary Laboratories (MDL), Foundation University School of Health Sciences (FUSH), Foundation University Islamabad (FUI), Islamabad, Pakistan
| | - Muzammil Hasan Najmi
- Clinical and Biomedical Research Center (CBRC) and Multidisciplinary Laboratories (MDL), Foundation University School of Health Sciences (FUSH), Foundation University Islamabad (FUI), Islamabad, Pakistan
| | - Imran Fazal
- Clinical and Biomedical Research Center (CBRC) and Multidisciplinary Laboratories (MDL), Foundation University School of Health Sciences (FUSH), Foundation University Islamabad (FUI), Islamabad, Pakistan
| |
Collapse
|
18
|
Faghihkhorasani A, Ahmed HH, Mashool NM, Alwan M, Assefi M, Adab AH, Yasamineh S, Gholizadeh O, Baghani M. The potential use of bacteria and bacterial derivatives as drug delivery systems for viral infection. Virol J 2023; 20:222. [PMID: 37789431 PMCID: PMC10548687 DOI: 10.1186/s12985-023-02183-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/11/2023] [Indexed: 10/05/2023] Open
Abstract
Viral infections in humans are responsible for fatalities worldwide and contribute to the incidence of various human ailments. Controllable targeted medicine delivery against many illnesses, including viral infection, may be significantly aided by using bacteria and bacteria-derived products. They may accumulate in diseased tissues despite physical obstacles, where they can launch antiviral immunity. The ability to genetically and chemically modify them means that vaccinations against viral infections may be manufactured and delivered to affected tissues more safely and effectively. The objective of this study is to provide an overview of the latest advancements in the field of utilizing bacteria and bacterial derivatives as carriers for administering medication to treat viral diseases such as SARS-CoV-2, hepatitis B virus, hepatitis C virus, human immunodeficiency virus, human papillomavirus, influenza, and Ebola virus.
Collapse
Affiliation(s)
| | | | | | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | - Marjan Assefi
- University of North Carolina at Greensboro, Greensboro, USA
| | - Aya Hussein Adab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Omid Gholizadeh
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| | - Moein Baghani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Fan F, Wang J, Chen H, Wei L, Zhang Z. Isolation and protein MdtQ analysis of outer membrane vesicles released by carbapenem-resistant Klebsiella pneumoniae. Microb Pathog 2023; 183:106325. [PMID: 37640276 DOI: 10.1016/j.micpath.2023.106325] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CRKP) has emerged as a leading public health problem, and is increasingly being reported worldwide with resistance to a wide spectrum of antibiotics. Recent reports have demonstrated that the outer membrane vesicles (OMVs) of gram-negative bacteria are potent resistance factors, but their role in the drug resistance of CRKP has not been elucidated. In order to investigate the effects of OMV components on drug resistance and to explore the mechanism of antimicrobial resistance in CRKP, we isolated the OMVs through ultracentrifugation, separated the OMV proteins through mass spectrometry (MS), and performed bioinformatics analysis. A total of 3,192 proteins were detected by nano LC-MS/MS analysis, with 108 (61.4%) cytoplasmic proteins, 50 (28.4%) cytoplasmic membrane proteins, nine (5.1%) periplasmic proteins, six (3.4%) outer membrane proteins, two (1.1%) extracellular proteins, and one (0.6%) other protein detected in the vesicles. MdtQ was detected as the only multidrug resistance outer membrane protein. Further experiments confirmed that MdtQ included the 1440 BP sequence and had a unique three-dimensional structure. To superimpose MdtQ with KPC-2 resistant proteins, I7ACB1, I7AKP2, and Q93LQ9, the root mean square deviation (RMSD) values were calculated (0.379, 0.671, and 1.35, respectively). I7ACB1 had the lowest RMSD value, indicating that it had the best superimposition effect. Furthermore, MdtQ had 20 biological pocket structures, and the four most important pockets were evenly distributed around the inner perimeter of its three-dimensional structure. These findings may provide a theoretical basis for controlling the spread of bacterial resistance in the future.
Collapse
Affiliation(s)
- Fangfang Fan
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, 233030, China
| | - Jiaqi Wang
- Department of Clinical Laboratory, Shanghai Clinical Medical College of Qingdao University, Shanghai Deji Hospital, Shanghai, 200331, China
| | - Hong Chen
- Department of Clinical Laboratory, Shanghai Clinical Medical College of Qingdao University, Shanghai Deji Hospital, Shanghai, 200331, China
| | - Li Wei
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, 233030, China.
| | - Zhen Zhang
- Department of Clinical Laboratory, Shanghai Clinical Medical College of Qingdao University, Shanghai Deji Hospital, Shanghai, 200331, China.
| |
Collapse
|
20
|
Lu W, Lu H, Wang C, Wang G, Dong W, Tan C. Effectors of the Type VI Secretion System Have the Potential to Be Modified into Antimicrobial Peptides. Microbiol Spectr 2023; 11:e0030823. [PMID: 37470717 PMCID: PMC10434152 DOI: 10.1128/spectrum.00308-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/26/2023] [Indexed: 07/21/2023] Open
Abstract
The use of antibiotics has led to the emergence of multidrug-resistant (MDR) bacteria, and there is an urgent need to find alternative treatments to alleviate this pressure. The type VI secretion system (T6SS) is a protein delivery system present in bacterial cells that secretes effectors that participate in bacterial virulence. Given the potential for the transformation of these effectors into antimicrobial peptides (AMPs), we designed T6SS effectors into AMPs that have a membrane-disrupting effect. These effectors kill bacteria by altering the membrane potential and increasing the intracellular reactive oxygen species (ROS) content. Moreover, AMPs also have a significant therapeutic effect both in vivo and in vitro. This finding suggests that it is possible to modify bacterial components of bacteria themselves to create compounds that fight bacteria. IMPORTANCE This study first identified and modified the T6SS effector into positively charged alpha-helical peptides. These peptides have good antibacterial and bactericidal effects on G+ bacteria and G- bacteria. This study broadens the source of AMPs and makes T6SS effectors more useful.
Collapse
Affiliation(s)
- Wenjia Lu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Hao Lu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Chenchen Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Gaoyan Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Wenqi Dong
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| |
Collapse
|
21
|
Higham SL, Baker S, Flight KE, Krishna A, Kellam P, Reece ST, Tregoning JS. Intranasal immunization with outer membrane vesicles (OMV) protects against airway colonization and systemic infection with Acinetobacter baumannii. J Infect 2023; 86:563-573. [PMID: 36858180 DOI: 10.1016/j.jinf.2023.02.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/08/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023]
Abstract
OBJECTIVES The multidrug-resistant bacteria Acinetobacter baumannii is a major cause of hospital-associated infection; a vaccine could significantly reduce this burden. The aim was to develop a clinically relevant model of A. baumannii respiratory tract infection and to test the impact of different immunization routes on protective immunity provided by an outer membrane vesicle (OMV) vaccine. METHODS BALB/c mice were intranasally challenged with isolates of oxa23-positive global clone GC2 A. baumannii from the lungs of patients with ventilator-associated pneumonia. Mice were immunized with OMVs by the intramuscular, subcutaneous or intranasal routes; protection was determined by measuring local and systemic bacterial load. RESULTS Infection with A. baumannii clinical isolates led to a more disseminated infection than the prototype A. baumannii strain ATCC17978; with bacteria detectable in upper and lower airways and the spleen. Intramuscular immunization induced an antibody response but did not protect against bacterial infection. However, intranasal immunization significantly reduced airway colonization and prevented systemic bacterial dissemination. CONCLUSIONS Use of clinically relevant isolates of A. baumannii provides stringent model for vaccine development. Intranasal immunization with OMVs was an effective route for providing protection, demonstrating that local immunity is important in preventing A. baumannii infection.
Collapse
Affiliation(s)
- Sophie L Higham
- Department of Infectious Disease, Imperial College London, St Marys Campus, Norfolk Place, London W2 1PG, United Kingdom
| | - Stephen Baker
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, United Kingdom
| | - Katie E Flight
- Department of Infectious Disease, Imperial College London, St Marys Campus, Norfolk Place, London W2 1PG, United Kingdom
| | - Aishwarya Krishna
- Infectious Diseases and Vaccines, Kymab, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Paul Kellam
- Department of Infectious Disease, Imperial College London, St Marys Campus, Norfolk Place, London W2 1PG, United Kingdom; Infectious Diseases and Vaccines, Kymab, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom; RQ Biotechnology Ltd, 7-12 Tavistock Square, London WC1H 9LT, United Kingdom
| | - Stephen T Reece
- Infectious Diseases and Vaccines, Kymab, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom.
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, St Marys Campus, Norfolk Place, London W2 1PG, United Kingdom.
| |
Collapse
|
22
|
Yang J, Jia F, Qiao Y, Hai Z, Zhou X. Correlation between bacterial extracellular vesicles and antibiotics: A potentially antibacterial strategy. Microb Pathog 2023:106167. [PMID: 37224984 DOI: 10.1016/j.micpath.2023.106167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 05/26/2023]
Abstract
Bacterial extracellular vesicles (BEVs) are proteoliposome nanoparticles that are secreted by both Gram-negative (G-) and Gram-positive (G+) bacteria. BEVs have significant roles in various physiological processes of bacteria, including driving inflammatory responses, regulating bacterial pathogenesis, and promoting bacterial survival in diverse environments. Recently, there has been increasing interest in the use of BEVs as a potential solution to antibiotic resistance. BEVs have shown great promise as a new approach to antibiotics, as well as a drug-delivery tool in antimicrobial strategies. In this review, we provide a summary of recent scientific advances in BEVs and antibiotics, including BEV biogenesis, ability to kill bacteria, potential for delivering antibiotics, and their role in the development of vaccines or as immune adjuvants. We propose that BEVs provide a novel antimicrobial strategy that would be beneficial against the increasing threat of antibiotic resistance.
Collapse
Affiliation(s)
- Jiangliu Yang
- College of Life Science, Ningxia University, Yinchuan, 750021, China; Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, 750021, China
| | - Fang Jia
- Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot, 010058, China
| | - Yarui Qiao
- College of Life Science, Ningxia University, Yinchuan, 750021, China; Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, 750021, China
| | - Zhenzhen Hai
- College of Life Science, Ningxia University, Yinchuan, 750021, China; Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, 750021, China
| | - Xuezhang Zhou
- College of Life Science, Ningxia University, Yinchuan, 750021, China; Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, 750021, China.
| |
Collapse
|
23
|
Amalia L, Tsai SL. Functionalization of OMVs for Biocatalytic Applications. MEMBRANES 2023; 13:membranes13050459. [PMID: 37233521 DOI: 10.3390/membranes13050459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023]
Abstract
Outer membrane vesicles (OMVs) are miniature versions of gram-negative bacteria that contain almost the same content as their parent cells, particularly in terms of membrane composition. Using OMVs as biocatalysts is a promising approach due to their potential benefits, including their ability to be handled similarly to bacteria while lacking potentially pathogenic organisms. To employ OMVs as biocatalysts, they must be functionalized with immobilized enzymes to the OMV platform. Various enzyme immobilization techniques are available, including surface display and encapsulation, each with advantages and disadvantages depending on the objectives. This review provides a concise yet comprehensive overview of these immobilization techniques and their applications in utilizing OMVs as biocatalysts. Specifically, we discuss the use of OMVs in catalyzing the conversion of chemical compounds, their role in polymer degradation, and their performance in bioremediation.
Collapse
Affiliation(s)
- Lita Amalia
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City 10607, Taiwan
| | - Shen-Long Tsai
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City 10607, Taiwan
| |
Collapse
|
24
|
Kumar M, Silori R, Mazumder P, Shrivastava V, Loge F, Barceló D, Mahlknecht J. Wars and Pandemics: AMR Accelerators of the 21st Century? ENVIRONMENTAL SCIENCE & TECHNOLOGY LETTERS 2023; 10:289-291. [DOI: 10.1021/acs.estlett.3c00020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Manish Kumar
- School of Engineering, University of Petroleum & Energy Studies, Dehradun, Uttarakhand 248007, India
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Campus Monterrey, Monterrey 64849, Nuevo Leon, Mexico
| | - Rahul Silori
- School of Engineering, University of Petroleum & Energy Studies, Dehradun, Uttarakhand 248007, India
| | - Payal Mazumder
- School of Engineering, University of Petroleum & Energy Studies, Dehradun, Uttarakhand 248007, India
| | - Vikalp Shrivastava
- School of Engineering, University of Petroleum & Energy Studies, Dehradun, Uttarakhand 248007, India
| | - Frank Loge
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Campus Monterrey, Monterrey 64849, Nuevo Leon, Mexico
- Department of Civil and Environmental Engineering, University of California Davis, One Shields Avenue, Davis, California 95616, United States
| | - Damià Barceló
- Catalan Institute for Water Research (ICRA-CERCA), H2O Building, Scientific and Technological Park of the University of Girona, Emili Grahit 101, Girona 17003, Spain
| | - Jürgen Mahlknecht
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Campus Monterrey, Monterrey 64849, Nuevo Leon, Mexico
| |
Collapse
|
25
|
Lim Y, Kim HY, Sun-Jin An, Choi BK. Activation of bone marrow-derived dendritic cells and CD4 + T cell differentiation by outer membrane vesicles of periodontal pathogens. J Oral Microbiol 2022; 14:2123550. [PMID: 36312320 PMCID: PMC9616074 DOI: 10.1080/20002297.2022.2123550] [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] [Indexed: 11/15/2022] Open
Abstract
Outer membrane vesicles (OMVs) released from gram-negative bacteria harbor diverse molecules to communicate with host cells. In this study, we evaluated the OMVs of periodontal pathogens for their effects on the activation of dendritic cells and CD4+ T cell differentiation. OMVs of Porphyromonas gingivalis ATCC 33277, Treponema denticola ATCC 33521, and Tannerella forsythia ATCC 43037 (‘red complex’ pathogens) were isolated by density gradient ultracentrifugation. Mouse bone marrow-derived dendritic cells (BMDCs) were treated with OMVs, and OMV-primed BMDCs were cocultured with naïve CD4+ T cells to analyze the polarization of effector helper T cells. The OMVs upregulated maturation markers, including MHC class II, CD80, CD86, and CD40, on BMDCs. OMVs of P. gingivalis and T. forsythia induced the expression of the proinflammatory cytokines IL-1β, IL-6, IL-23, and IL-12p70 in BMDCs. In T. denticola OMV-primed BMDCs, proinflammatory cytokines were poorly detected, which may be attributed to posttranslational degradation due to the highly proteolytic nature of OMVs. In cocultures of naïve CD4+ T cells with OMV-primed BMDCs, OMVs of P. gingivalis and T. denticola induced the differentiation of Th17 cells, whereas T. forsythia OMVs induced Th1 cell differentiation. These results demonstrate that OMVs derived from the ‘red complex’ periodontal pathogens induce maturation of BMDCs and differentiation of naïve CD4+ T cells to Th1 or Th17 cells.
Collapse
Affiliation(s)
- Younggap Lim
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Hyun Young Kim
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | | | - Bong-Kyu Choi
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Pseudomonas aeruginosa PAO1 outer membrane vesicles-diphtheria toxoid conjugate as a vaccine candidate in a murine burn model. Sci Rep 2022; 12:22324. [PMID: 36566282 PMCID: PMC9789887 DOI: 10.1038/s41598-022-26846-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen considered a common cause of nosocomial infection with high morbidity and mortality in burn patients. Immunoprophylaxis techniques may lower the mortality rate of patients with burn wounds infected by P. aeruginosa; consequently, this may be an efficient strategy to manage infections caused by this bacterium. Several pathogenic Gram-negative bacteria like P. aeruginosa release outer membrane vesicles (OMVs), and structurally OMV consists of several antigenic components capable of generating a wide range of immune responses. Here, we evaluated the immunogenicity and efficacy of P. aeruginosa PA-OMVs (PA-OMVs) conjugated with the diphtheria toxoid (DT) formulated with alum adjuvant (PA-OMVs-DT + adj) in a mice model of burn wound infection. ELISA results showed that in the group of mice immunized with PA-OMVs-DT + adj conjugated, there was a significant increase in specific antibodies titer compared to non-conjugated PA-OMVs or control groups. In addition, the vaccination of mice with PA-OMVs-DT + adj conjugated generated greater protective effectiveness, as seen by lower bacterial loads, and eightfold decreased inflammatory cell infiltration with less tissue damage in the mice burn model compared to the control group. The opsonophagocytic killing results confirmed that humoral immune response might be critical for PA-OMVs mediated protection. These findings suggest that PA-OMV-DT conjugated might be used as a new vaccine against P. aeruginosa in burn wound infection.
Collapse
|
27
|
Lieberman LA. Outer membrane vesicles: A bacterial-derived vaccination system. Front Microbiol 2022; 13:1029146. [PMID: 36620013 PMCID: PMC9811673 DOI: 10.3389/fmicb.2022.1029146] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Outer membrane vesicles (OMVs) are non-living spherical nanostructures that derive from the cell envelope of Gram-negative bacteria. OMVs are important in bacterial pathogenesis, cell-to-cell communication, horizontal gene transfer, quorum sensing, and in maintaining bacterial fitness. These structures can be modified to express antigens of interest using glycoengineering and genetic or chemical modification. The resulting OMVs can be used to immunize individuals against the expressed homo- or heterologous antigens. Additionally, cargo can be loaded into OMVs and they could be used as a drug delivery system. OMVs are inherently immunogenic due to proteins and glycans found on Gram negative bacterial outer membranes. This review focuses on OMV manipulation to increase vesiculation and decrease antigenicity, their utility as vaccines, and novel engineering approaches to extend their application.
Collapse
|
28
|
Wo J, Lv ZY, Sun JN, Tang H, Qi N, Ye BC. Engineering probiotic-derived outer membrane vesicles as functional vaccine carriers to enhance immunity against SARS-CoV-2. iScience 2022; 26:105772. [PMID: 36510593 PMCID: PMC9729586 DOI: 10.1016/j.isci.2022.105772] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Because of the continued emergence of SARS-CoV-2 variants, there has been considerable interest in how to display multivalent antigens efficiently. Bacterial outer membrane vesicles (OMVs) can serve as an attractive vaccine delivery system because of their self-adjuvant properties and the ability to be decorated with antigens. Here we set up a bivalent antigen display platform based on engineered OMVs using mCherry and GFP and demonstrated that two different antigens of SARS-CoV-2 could be presented simultaneously in the lumen and on the surface of OMVs. Comparing immunogenicity, ClyA-NG06 fusion and the receptor-binding domain (RBD) of the spike protein in the OMV lumen elicited a stronger humoral response in mice than OMVs presenting either the ClyA-NG06 fusion or RBD alone. Taken together, we provided an efficient approach to display SARS-CoV-2 antigens in the lumen and on the surface of the same OMV and highlighted the potential of OMVs as general multi-antigen carriers.
Collapse
Affiliation(s)
- Jing Wo
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Zhao-Yong Lv
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Jia-Nan Sun
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Hao Tang
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Nan Qi
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China
| | - Bang-Ce Ye
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014 Zhejiang, China,Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China,Corresponding author
| |
Collapse
|
29
|
Srivastava P, Kim KS. Membrane Vesicles Derived from Gut Microbiota and Probiotics: Cutting-Edge Therapeutic Approaches for Multidrug-Resistant Superbugs Linked to Neurological Anomalies. Pharmaceutics 2022; 14:2370. [PMID: 36365188 PMCID: PMC9692612 DOI: 10.3390/pharmaceutics14112370] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Multidrug-resistant (MDR) superbugs can breach the blood-brain barrier (BBB), leading to a continuous barrage of pro-inflammatory modulators and induction of severe infection-related pathologies, including meningitis and brain abscess. Both broad-spectrum or species-specific antibiotics (β-lactamase inhibitors, polymyxins, vancomycin, meropenem, plazomicin, and sarecycline) and biocompatible poly (lactic-co-glycolic acid) (PLGA) nanoparticles have been used to treat these infections. However, new therapeutic platforms with a broad impact that do not exert off-target deleterious effects are needed. Membrane vesicles or extracellular vesicles (EVs) are lipid bilayer-enclosed particles with therapeutic potential owing to their ability to circumvent BBB constraints. Bacteria-derived EVs (bEVs) from gut microbiota are efficient transporters that can penetrate the central nervous system. In fact, bEVs can be remodeled via surface modification and CRISPR/Cas editing and, thus, represent a novel platform for conferring protection against infections breaching the BBB. Here, we discuss the latest scientific research related to gut microbiota- and probiotic-derived bEVs, and their therapeutic modifications, in terms of regulating neurotransmitters and inhibiting quorum sensing, for the treatment of neurodegenerative diseases, such as Parkinson's and Alzheimer's diseases. We also emphasize the benefits of probiotic-derived bEVs to human health and propose a novel direction for the development of innovative heterologous expression systems to combat BBB-crossing pathogens.
Collapse
Affiliation(s)
| | - Kwang-sun Kim
- Department of Chemistry and Chemistry Institute of Functional Materials, Pusan National University, Busan 46241, Korea
| |
Collapse
|
30
|
Addressing a Rule of Thumb: Modeling the Effects of Meteorological Conditions on Prescription of Antimicrobials in Aquaculture. Microbiol Spectr 2022; 10:e0175222. [PMID: 36122183 PMCID: PMC9603815 DOI: 10.1128/spectrum.01752-22] [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] [Indexed: 12/31/2022] Open
Abstract
Meteorological phenomena such as warm summer temperatures and increased numbers of sunlight hours have repeatedly been hypothesized to be associated with infectious diseases and an increased prescription of antimicrobial compounds in Danish aquaculture. To address this hypothesis, we prepared statistical models incorporating aquaculture production data from Danish Aquaculture, detailed records on prescription of antimicrobials from the Danish VETSTAT program, and meteorological data from 2001 to 2019 from the Danish Meteorological Institute. Separate series of models were made and refined for land-based and marine production, respectively. For both production forms, the models identify summer sunlight hours as having a significant influence on antimicrobial use. In addition to summer sunlight, spring sunlight and water temperature were integral, although not statistically significant, parameters when modeling antimicrobial use in marine production. Although the extensive availability and accuracy of relevant data are associated with Danish production, we believe the results allow for more general conclusions on the influence of meteorological parameters on outbreaks of bacterial pathogens in international aquaculture. Such insights could have a substantial impact on prophylactic strategies, fish husbandry, and our understanding of how increasing temperatures may affect future antimicrobial usage in the global aquaculture industry. IMPORTANCE Global aquaculture production has been rapidly increasing for decades and is set to play a pivotal role in feeding a growing human population. Along with the growth in aquaculture production, the annual global use of antimicrobials is estimated to increase by one-third between 2017 and 2030. Using detailed antimicrobial prescription records as a proxy for outbreaks, we were able to evaluate the effects of a variety of meteorological parameters through statistical modeling. Our results lend scientific support to informal observations from the industry, but more importantly, this study provides novel, essential information on the importance of abiotic factors that can, in turn, lead to improved prophylactic efforts and thus help to reduce antimicrobial use in global aquaculture.
Collapse
|
31
|
Outer Membrane Vesicles of Actinobacillus pleuropneumoniae Exert Immunomodulatory Effects on Porcine Alveolar Macrophages. Microbiol Spectr 2022; 10:e0181922. [PMID: 36040198 PMCID: PMC9602539 DOI: 10.1128/spectrum.01819-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Outer membrane vesicles (OMVs) are spontaneously released by Gram-negative bacteria, including Actinobacillus pleuropneumoniae, which causes contagious pleuropneumonia in pigs and leads to considerable economic losses in the swine industry worldwide. A. pleuropneumoniae OMVs have previously been demonstrated to contain Apx toxins and proteases, as well as antigenic proteins. Nevertheless, comprehensive characterizations of their contents and interactions with host immune cells have not been made. Understanding the protein compositions and immunomodulating ability of A. pleuropneumoniae OMVs could help illuminate their biological functions and facilitate the development of OMV-based applications. In the current investigation, we comprehensively characterized the proteome of native A. pleuropneumoniae OMVs. Moreover, we qualitatively and quantitatively compared the OMV proteomes of a wild-type strain and three mutant strains, in which relevant genes were disrupted to increase OMV production and/or produce OMVs devoid of superantigen PalA. Furthermore, the interaction between A. pleuropneumoniae OMVs and porcine alveolar macrophages was also characterized. Our results indicate that native OMVs spontaneously released by A. pleuropneumoniae MIDG2331 appeared to dampen the innate immune responses by porcine alveolar macrophages stimulated by either inactivated or live parent cells. The findings suggest that OMVs may play a role in manipulating the porcine defense during the initial phases of the A. pleuropneumoniae infection. IMPORTANCE Owing to their built-in adjuvanticity and antigenicity, bacterial outer membrane vesicles (OMVs) are gaining increasing attention as potential vaccines for both human and animal use. OMVs released by Actinobacillus pleuropneumoniae, an important respiratory pathogen in pigs, have also been investigated for vaccine development. Our previous studies have shown that A. pleuropneumoniae secretes OMVs containing multiple immunogenic proteins. However, immunization of pigs with these vesicles was not able to relieve the pig lung lesions induced by the challenge with A. pleuropneumoniae, implying the elusive roles that A. pleuropneumoniae OMVs play in host-pathogen interaction. Here, we showed that A. pleuropneumoniae secretes OMVs whose yield and protein content can be altered by the deletion of the nlpI and palA genes. Furthermore, we demonstrate that A. pleuropneumoniae OMVs dampen the immune responses in porcine alveolar macrophages stimulated by A. pleuropneumoniae cells, suggesting a novel mechanism that A. pleuropneumoniae might use to evade host defense.
Collapse
|
32
|
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: 41] [Impact Index Per Article: 13.7] [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
|
33
|
Allahghadry T, Bojesen AM, Whitehead BJ, Antenucci F. Clarification of large-volume bacterial cultures using a centrifuge-free protocol. J Appl Microbiol 2022; 133:870-882. [PMID: 35503033 PMCID: PMC9542984 DOI: 10.1111/jam.15608] [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: 02/11/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/30/2022]
Abstract
Aims To provide a reliable, reproducible and centrifuge‐free filtration protocol for clarification of large volumes of bacterial cultures. Methods and Results Four experiments were designed to compare different techniques enabling clarification of Escherichia coli cultures using as a benchmark the concentration and quality of bacterial outer membrane vesicles (OMVs). The experiments were designed to examine the performance of different extraction methods on large volume (≥1 L) filtrations of bacterial culture media. Performance parameters included filtration flow rates, sterility testing and characterization of the filtrates by: (i) SDS‐PAGE, (ii) cryogenic transmission electron microscopy, (iii) nanoparticle tracking analysis and (iv) Qubit protein quantification. The experiments revealed that: (i) addition of the filter aid Diatomaceous Earth to the bacterial cultures improved filtration flow rates significantly and eliminated the need for centrifugation prior to filtration; (ii) sterile filtration was successful as no bacterial passage was identified through the membrane filter; (iii) centrifuge‐free filtrates contained an increased amount of OMVs compared to centrifuged filtrates. Conclusions In comparison to conventional centrifuge‐based protocols, the clarification method presented has universal applicability for a broad range of microbial extraction procedures, regardless of the volume of culture harvested. Moreover, the decreased amount of OMVs presented in the filtrates following centrifugation step provides an additional argument in favour of a centrifuge‐free approach. Significance and Impact of the Study Sterile filtration is a universal method for the clarification of bacterial cultures. Common challenges related to filtration include filter clogging and long processing times, due to limited centrifugation capacity, which can affect product quality. The proposed protocol is likely to ensure a highly effective filtration process and could be a novel approach in improving the filtrate products without the need of centrifugation.
Collapse
Affiliation(s)
- Toloe Allahghadry
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Miki Bojesen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bradley Joseph Whitehead
- Department of Clinical Medicine, Department of Infectious Diseases, Aarhus University, Aarhus N, Denmark
| | - Fabio Antenucci
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Kroniger T, Flender D, Schlüter R, Köllner B, Trautwein-Schult A, Becher D. Proteome analysis of the Gram-positive fish pathogen Renibacterium salmoninarum reveals putative role of membrane vesicles in virulence. Sci Rep 2022; 12:3003. [PMID: 35194033 PMCID: PMC8863785 DOI: 10.1038/s41598-022-06130-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/04/2022] [Indexed: 12/11/2022] Open
Abstract
Bacterial kidney disease (BKD) is a chronic bacterial disease affecting both wild and farmed salmonids. The causative agent for BKD is the Gram-positive fish pathogen Renibacterium salmoninarum. As treatment and prevention of BKD have proven to be difficult, it is important to know and identify the key bacterial proteins that interact with the host. We used subcellular fractionation to report semi-quantitative data for the cytosolic, membrane, extracellular, and membrane vesicle (MV) proteome of R. salmoninarum. These data can aid as a backbone for more targeted experiments regarding the development of new drugs for the treatment of BKD. Further analysis was focused on the MV proteome, where both major immunosuppressive proteins P57/Msa and P22 and proteins involved in bacterial adhesion were found in high abundance. Interestingly, the P22 protein was relatively enriched only in the extracellular and MV fraction, implicating that MVs may play a role in host–pathogen interaction. Compared to the other subcellular fractions, the MVs were also relatively enriched in lipoproteins and all four cell wall hydrolases belonging to the New Lipoprotein C/Protein of 60 kDa (NlpC/P60) family were detected, suggesting an involvement in the formation of the MVs.
Collapse
Affiliation(s)
- Tobias Kroniger
- Institute of Microbiology, Department of Microbial Proteomics, Center for Functional Genomics of Microbes, University of Greifswald, 17489, Greifswald, Germany
| | - Daniel Flender
- Institute of Microbiology, Department of Microbial Proteomics, Center for Functional Genomics of Microbes, University of Greifswald, 17489, Greifswald, Germany
| | - Rabea Schlüter
- Imaging Center of the Department of Biology, University of Greifswald, 17489, Greifswald, Germany
| | - Bernd Köllner
- Institute of Immunology, Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, 17493, Greifswald - Isle of Riems, Germany
| | - Anke Trautwein-Schult
- Institute of Microbiology, Department of Microbial Proteomics, Center for Functional Genomics of Microbes, University of Greifswald, 17489, Greifswald, Germany
| | - Dörte Becher
- Institute of Microbiology, Department of Microbial Proteomics, Center for Functional Genomics of Microbes, University of Greifswald, 17489, Greifswald, Germany.
| |
Collapse
|
35
|
Xue K, Wang L, Liu J. Bacterial outer membrane vesicles and their functionalization as vehicles for bioimaging, diagnosis and therapy. MATERIALS ADVANCES 2022; 3:7185-7197. [DOI: 10.1039/d2ma00420h] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
In this review, we summarize the bioactivities of bacterial outer membrane vesicles, including biogenesis, immunogenicity, and interactions, followed by a discussion on their functionalization as nanocarriers for bioimaging, diagnosis, and therapy.
Collapse
Affiliation(s)
- Kaikai Xue
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
36
|
Toyofuku M, Kikuchi Y, Taoka A. A Single Shot of Vesicles. Microbes Environ 2022; 37. [PMID: 36504177 PMCID: PMC10037094 DOI: 10.1264/jsme2.me22083] [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: 12/13/2022] Open
Abstract
Bacteria communicate through signaling molecules that coordinate group behavior. Hydrophobic signals that do not diffuse in aqueous environments are used as signaling molecules by several bacteria. However, limited information is currently available on the mechanisms by which these molecules are transported between cells. Membrane vesicles (MVs) with diverse functions play important roles in the release and delivery of hydrophobic signaling molecules, leading to differences in the dynamics of signal transportation from those of free diffusion. Studies on Paracoccus denitrificans, which produces a hydrophobic long-chain N-acyl homoserine lactone (AHL), showed that signals were loaded into MVs at a concentration with the potential to trigger the quorum sensing (QS) response with a "single shot" to the cell. Furthermore, stimulating the formation of MVs increased the release of signals from the cell; therefore, a basic understanding of MV formation is important. Novel findings revealed the formation of MVs through different routes, resulting in the production of different types of MVs. Methods such as high-speed atomic force microscopy (AFM) phase imaging allow the physical properties of MVs to be analyzed at a nanometer resolution, revealing their heterogeneity. In this special minireview, we introduce the role of MVs in bacterial communication and highlight recent findings on MV formation and their physical heterogeneity by referring to our research. We hope that this minireview will provide basic information for understanding the functionality of MVs in ecological systems.
Collapse
Affiliation(s)
- Masanori Toyofuku
- Faculty of Life and Environmental Sciences, University of Tsukuba
- Microbiology Research Center for Sustainability (MiCS), University of Tsukuba
| | - Yousuke Kikuchi
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University
| | - Azuma Taoka
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University
- Institute of Science and Engineering, Kanazawa University
| |
Collapse
|
37
|
Çelik P, Derkuş B, Erdoğan K, Barut D, Manga EB, Yıldırım Y, Pecha S, Çabuk A. Bacterial membrane vesicle functions, laboratory methods, and applications. Biotechnol Adv 2021; 54:107869. [PMID: 34793882 DOI: 10.1016/j.biotechadv.2021.107869] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/19/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022]
Abstract
Bacterial membrane vesicles are cupped-shaped structures formed by bacteria in response to environmental stress, genetic alteration, antibiotic exposure, and others. Due to the structural similarities shared with the producer organism, they can retain certain characteristics like stimulating immune responses. They are also able to carry molecules for long distances, without changes in the concentration and integrity of the molecule. Bacteria originally secrete membrane vesicles for gene transfer, excretion, cell to cell interaction, pathogenesis, and protection against phages. These functions are unique and have several innovative applications in the pharmaceutical industry that have attracted both scientific and commercial interest.This led to the development of efficient methods to artificially stimulate vesicle production, purification, and manipulation in the lab at nanoscales. Also, for specific applications, engineering methods to impart pathogen antigens against specific diseases or customization as cargo vehicles to deliver payloads to specific cells have been reported. Many applications of bacteria membrane vesicles are in cancer drugs, vaccines, and adjuvant development with several candidates in clinical trials showing promising results. Despite this, applications in therapy and commercialization stay timid probably due to some challenges one of which is the poor understanding of biogenesis mechanisms. Nevertheless, so far, bacterial membrane vesicles seem to be a reliable and cost-efficient technology with several therapeutic applications. Research toward characterizing more membrane vesicles, genetic engineering, and nanotechnology will enable the scope of applications to widen. This might include solutions to other currently faced medical and healthcare-related challenges.
Collapse
Affiliation(s)
- PınarAytar Çelik
- Environmental Protection and Control Program, Eskişehir Osmangazi University, Eskişehir 26110, Turkey; Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Burak Derkuş
- Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey
| | - Kübra Erdoğan
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Dilan Barut
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Enuh Blaise Manga
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Yalın Yıldırım
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Simon Pecha
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Ahmet Çabuk
- Department of Biology, Faculty of Science and Letter, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| |
Collapse
|