1
|
Moghaddam ZS, Dehghan A, Halimi S, Najafi F, Nokhostin A, Naeini AE, Akbarzadeh I, Ren Q. Bacterial Extracellular Vesicles: Bridging Pathogen Biology and Therapeutic Innovation. Acta Biomater 2025:S1742-7061(25)00352-6. [PMID: 40349898 DOI: 10.1016/j.actbio.2025.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/14/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
The main role of bacterial extracellular vesicles (BEVs) has been associated with various processes such as intercellular communication and host-pathogen interactions. This comprehensive review explores the multifaceted functions of BEVs across different biological domains, emphasizing their dual nature as contributors to disease and potential vehicles for therapeutic intervention. We examine the intricate interactions of BEVs within bacterial communities and between bacteria and hosts, their involvement in disease development through cargo delivery mechanisms, and their beneficial impact on microbial ecology. The review also highlights BEVs' applications in biomedical field, where they are revolutionizing vaccine development, targeted drug delivery, and cancer therapy. By utilizing the inherent properties of BEVs for controlled drug release, targeted antigen delivery, and immune modulation, they offer a promising frontier in precision medicine. In addition, the diagnostic potential of BEVs is explored through their utility as biomarkers, providing valuable insights into disease states and treatment efficacy. Looking forward, this review underscores the challenges and opportunities in translating BEV research to clinical practice, promoting the need of standardized methods in BEV characterization and scaling up production. The diverse abilities of BEVs, ranging from contributing to pathogen virulence to driving therapeutic innovation, highlight their potential as a cornerstone in the future of biomedical advancements. STATEMENT OF SIGNIFICANCE: Bacterial extracellular vesicles (BEVs) are emerging as pivotal players in both pathogenesis and therapeutic innovation. This review explores their dual nature as agents of disease and as promising biomaterials for biomedical applications, and provides a comprehensive survey on their involvement in disease mechanisms and microbial ecology, and their potential in biomedical applications such as vaccine development, targeted drug delivery, cancer therapy, and diagnosis. It highlights the complex interactions of BEVs within bacterial communities and between bacteria and hosts. This review also addresses current advancements, challenges, and opportunities in translating BEV research into clinical practice. The insights presented here position BEVs as a cornerstone in the future of biomedical advancements, advocating for standardized methods in BEV characterization and scalable production techniques.
Collapse
Affiliation(s)
| | - Ashkan Dehghan
- W Booth School of Engineering Practice and Technology Faculty of Engineering, McMaster University Hamilton, ON, Canada, L8S 0A3
| | - Saba Halimi
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Fatemeh Najafi
- Department of Chemical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802-1503, United States
| | - Ali Nokhostin
- Medical Sciences & Technologies Faculty, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | | | - Iman Akbarzadeh
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| | - Qun Ren
- Laboratory for Biointerfaces, Empa Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland.
| |
Collapse
|
2
|
Portilho AI, Hermes Monteiro da Costa H, Grando Guereschi M, Prudencio CR, De Gaspari E. Hybrid response to SARS-CoV-2 and Neisseria meningitidis C after an OMV-adjuvanted immunization in mice and their offspring. Hum Vaccin Immunother 2024; 20:2346963. [PMID: 38745461 PMCID: PMC11789737 DOI: 10.1080/21645515.2024.2346963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/16/2024] Open
Abstract
COVID-19, caused by SARS-CoV-2, and meningococcal disease, caused by Neisseria meningitidis, are relevant infectious diseases, preventable through vaccination. Outer membrane vesicles (OMVs), released from Gram-negative bacteria, such as N. meningitidis, present adjuvant characteristics and may confer protection against meningococcal disease. Here, we evaluated in mice the humoral and cellular immune response to different doses of receptor binding domain (RBD) of SARS-CoV-2 adjuvanted by N. meningitidis C:2a:P1.5 OMVs and aluminum hydroxide, as a combined preparation for these pathogens. The immunization induced IgG antibodies of high avidity for RBD and OMVs, besides IgG that recognized the Omicron BA.2 variant of SARS-CoV-2 with intermediary avidity. Cellular immunity showed IFN-γ and IL-4 secretion in response to RBD and OMV stimuli, demonstrating immunologic memory and a mixed Th1/Th2 response. Offspring presented transferred IgG of similar levels and avidity as their mothers. Humoral immunity did not point to the superiority of any RBD dose, but the group immunized with a lower antigenic dose (0.5 μg) had the better cellular response. Overall, OMVs enhanced RBD immunogenicity and conferred an immune response directed to N. meningitidis too.
Collapse
MESH Headings
- Animals
- Mice
- Immunoglobulin G/blood
- Neisseria meningitidis/immunology
- Female
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- Adjuvants, Immunologic/administration & dosage
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Immunity, Cellular
- Immunity, Humoral
- Mice, Inbred BALB C
- Meningococcal Infections/prevention & control
- Meningococcal Infections/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Adjuvants, Vaccine/administration & dosage
- Aluminum Hydroxide/administration & dosage
- Aluminum Hydroxide/immunology
- Immunization/methods
- Antibody Affinity
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Meningococcal Vaccines/immunology
- Meningococcal Vaccines/administration & dosage
- Immunologic Memory
- Th1 Cells/immunology
Collapse
Affiliation(s)
- Amanda Izeli Portilho
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | - Hernan Hermes Monteiro da Costa
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | | | - Carlos Roberto Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | - Elizabeth De Gaspari
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Bernardes BG, Moura AD, Guarnieri JPDO, Silva CFMD, Costa HHMD, Silva IGMD, Cordeiro KBB, Báo SN, Prudêncio CR, Lancellotti M. Vaccination with outer membrane vesicles from Neisseria Meningitidis and SBa15, SBa16 mesoporous silica associated with SARS-CoV-2 induces protective humoral and cellular response against COVID-19 in mice. Braz J Infect Dis 2024; 28:104479. [PMID: 39547005 DOI: 10.1016/j.bjid.2024.104479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/13/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
The global impact of the SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) pandemic in 2019-2020 has led to significant changes in worldwide vaccination and immune prophylactic approaches. In this study, our research delves into a new immunization strategy that does not involve the use of additional adjuvants or preservatives, focusing on the effects of virus fusion with a bacterial nanostructure. The experimental procedures outlined in this paper involved the cultivation of SARS-CoV-2, the production, extraction, and nanocharacterization of outer membrane vesicles (OMV) from Neisseria meningitidis, immunization of mice with two doses of OMV combined with SARS-CoV-2, and the use of mesoporous silica SBa15 and SBa16 adsorbed to the same virus. The immune response was assessed through an indirect elisa method, analysis of cytokine expression profiles, and seroneutralization of the SARS-CoV-2 strain. The characterizations of associated OMV - SARS-CoV-2 and adsorption SBa15 and SBa16 were performed using Nanosight Tracking Analysis (NTA), which showed a high density of particles in the formulation. mice were then immunized, resulting in an immune response that produced high levels of neutralizing antibodies in IgG and IgG1 mouse immunoglobulins. In addition, expressions of IL-2, IL-4, and IL-23 in spleen cells were reinforced after the vaccination process. The comparative study of these three vaccine formulations has shown that the development of new vaccines for SARS-CoV-2 should take into consideration the production of neutralizing antibodies and the maintenance of immunological memory.
Collapse
Affiliation(s)
- Bruno Gaia Bernardes
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil
| | - Andrew Douglas Moura
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil; Instituto Adolfo Lutz, Centro de Imunologia, São Paulo, SP, Brazil
| | - João Paulo de Oliveira Guarnieri
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil
| | - Carlos Fernando Macedo da Silva
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil
| | | | - Ingrid Gracielle Martins da Silva
- Universidade de Brasília, Campus Universitário Darcy Ribeiro, Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório de Microscopia e Microanálise (LMM), Brasília, DF, Brazil
| | - Karine Brenda Barros Cordeiro
- Universidade de Brasília, Campus Universitário Darcy Ribeiro, Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório de Microscopia e Microanálise (LMM), Brasília, DF, Brazil
| | - Sônia Nair Báo
- Universidade de Brasília, Campus Universitário Darcy Ribeiro, Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório de Microscopia e Microanálise (LMM), Brasília, DF, Brazil
| | | | - Marcelo Lancellotti
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil.
| |
Collapse
|
4
|
Ji L, Ruan H, Fu Y, Xiong S. A study of antigen selection by extracellular vesicles as vaccine candidates against Mycobacterium tuberculosis infection. J Med Microbiol 2024; 73. [PMID: 39133547 DOI: 10.1099/jmm.0.001865] [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: 08/13/2024] Open
Abstract
Introduction. Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (M. tb), remains a significant global public health concern. It is crucial to develop more effective vaccines for TB in order to achieve global control of the disease. Extracellular vesicles (EVs) are spherical membrane-bound structures released by pathogens and host cells. During the course of an infection, both pathogen- and host-derived EVs are produced and play important roles in determining the course of the infection. EVs offer intriguing tools as potential vaccines due to their ability to deliver multiple pathogen or host antigens.Hypothesis /Gap Statement. We hypothesized that EVs derived from M. tb and EVs from M. tb-infected macrophages may serve as potential vaccine candidates against M. tb infection.Aim. This study aims to compare the immunogenicity and immune protection between M. tb EVs and M. tb-infected macrophage-derived EVs.Methodology. In this study, EVs were extracted from culture supernatants of M. tb and M. tb-infected macrophages, respectively. Mass spectrometry was employed to explore the antigen composition of H37Rv-Mφ-EVs and H37Rv-EVs. Cytokine profiling and antibody response assays were used to analyse the immunogenicity offered by EVs. Additionally, we used histological examination to evaluate and protective efficacy of the EVs.Results. Our results demonstrated that mice immunized by EVs released from M. tb-infected macrophages induced stronger inflammatory cytokine response than M. tb. Moreover, EVs from M. tb-infected macrophages reinforced T-cell activation and antibody response compared to M. tb EVs. Proteomic analysis revealed that EVs from M. tb-infected macrophages containing immunodominant cargos have stronger binding ability with major histocompatibility complex molecules, which may contribute to the protection from M. tb infection. Indeed, immunization of EVs released from M. tb-infected macrophages significantly reduced the bacterial load and better protection against M. tb infection than EVs from M. tb. Importantly, the selected antigens (Ag85B, ESAT-6 and the Rv0580c) from EVs of M. tb-infected macrophages exhibited effective immunogenicity.Conclusion. Our results suggested that EVs derived from M. tb-infected macrophages might serve as a proper antigenic library for vaccine candidates against M. tb challenge.
Collapse
Affiliation(s)
- Lin Ji
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
| | - Hang Ruan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
| | - Yuxuan Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
- The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu 215123, PR China
| |
Collapse
|
5
|
da Costa HHM, Bielavsky M, Orts DJB, Araujo S, Adriani PP, Nogueira JS, Astray RM, Pandey RP, Lancellotti M, Cunha-Junior JP, Prudencio CR. Production of Recombinant Zika Virus Envelope Protein by Airlift Bioreactor as a New Subunit Vaccine Platform. Int J Mol Sci 2023; 24:13955. [PMID: 37762254 PMCID: PMC10531330 DOI: 10.3390/ijms241813955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 09/29/2023] Open
Abstract
The Zika Virus (ZIKV) is an emerging arbovirus of great public health concern, particularly in the Americas after its last outbreak in 2015. There are still major challenges regarding disease control, and there is no ZIKV vaccine currently approved for human use. Among many different vaccine platforms currently under study, the recombinant envelope protein from Zika Virus (rEZIKV) constitutes an alternative option for vaccine development and has great potential for monitoring ZIKV infection and antibody response. This study describes a method to obtain a bioactive and functional rEZIKV using an E. coli expression system, with the aid of a 5-L airlift bioreactor and following an automated fast protein liquid chromatography (FPLC) protocol, capable of obtaining high yields of approximately 20 mg of recombinant protein per liter of bacterium cultures. The purified rEZIKV presented preserved antigenicity and immunogenicity. Our results show that the use of an airlift bioreactor for the production of rEZIKV is ideal for establishing protocols and further research on ZIKV vaccines bioprocess, representing a promising system for the production of a ZIKV envelope recombinant protein-based vaccine candidate.
Collapse
Affiliation(s)
- Hernan H. M. da Costa
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Monica Bielavsky
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Diego J. B. Orts
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Laboratory of Cardiobiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, São Paulo 04023-062, Brazil
| | - Sergio Araujo
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Patrícia P. Adriani
- Skinzymes Biotechnology Ltd., São Paulo 05441-040, Brazil
- Laboratory of Nanopharmaceuticals and Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Renato M. Astray
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
- Multi-Purpose Laboratory Butantan Institute, São Paulo 05503-900, Brazil
| | - Ramendra P. Pandey
- School of Health Sciences and Technology, UPES University, Dehradun 248007, Uttarakhand, India
| | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences—FCF, University of Campinas—UNICAMP, Campinas 13083-871, Brazil
| | - Jair P. Cunha-Junior
- Laboratory of Immunochemistry and Immunotechnology, Department of Immunology, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
| | - Carlos R. Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
6
|
Aytar Çelik P, Erdogan-Gover K, Barut D, Enuh BM, Amasya G, Sengel-Türk CT, Derkus B, Çabuk A. Bacterial Membrane Vesicles as Smart Drug Delivery and Carrier Systems: A New Nanosystems Tool for Current Anticancer and Antimicrobial Therapy. Pharmaceutics 2023; 15:pharmaceutics15041052. [PMID: 37111538 PMCID: PMC10142793 DOI: 10.3390/pharmaceutics15041052] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Bacterial membrane vesicles (BMVs) are known to be critical communication tools in several pathophysiological processes between bacteria and host cells. Given this situation, BMVs for transporting and delivering exogenous therapeutic cargoes have been inspiring as promising platforms for developing smart drug delivery systems (SDDSs). In the first section of this review paper, starting with an introduction to pharmaceutical technology and nanotechnology, we delve into the design and classification of SDDSs. We discuss the characteristics of BMVs including their size, shape, charge, effective production and purification techniques, and the different methods used for cargo loading and drug encapsulation. We also shed light on the drug release mechanism, the design of BMVs as smart carriers, and recent remarkable findings on the potential of BMVs for anticancer and antimicrobial therapy. Furthermore, this review covers the safety of BMVs and the challenges that need to be overcome for clinical use. Finally, we discuss the recent advancements and prospects for BMVs as SDDSs and highlight their potential in revolutionizing the fields of nanomedicine and drug delivery. In conclusion, this review paper aims to provide a comprehensive overview of the state-of-the-art field of BMVs as SDDSs, encompassing their design, composition, fabrication, purification, and characterization, as well as the various strategies used for targeted delivery. Considering this information, the aim of this review is to provide researchers in the field with a comprehensive understanding of the current state of BMVs as SDDSs, enabling them to identify critical gaps and formulate new hypotheses to accelerate the progress of the field.
Collapse
Affiliation(s)
- Pınar Aytar Çelik
- Environmental Protection and Control Program, Eskisehir Osmangazi University, Eskisehir 26110, Turkey
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Kubra Erdogan-Gover
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Dilan Barut
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Blaise Manga Enuh
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| | - Gülin Amasya
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Ankara 06100, Turkey
| | - Ceyda Tuba Sengel-Türk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Ankara 06100, Turkey
| | - Burak Derkus
- Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
| | - Ahmet Çabuk
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
- Department of Biology, Faculty of Science, Eskisehir Osmangazi University, Eskisehir 26040, Turkey
| |
Collapse
|
7
|
da Costa HHM, Orts DJB, Moura AD, Duarte-Neto AN, Cirqueira CS, Réssio RA, Kanamura CT, Miguita K, Ferreira JE, Santos RTM, Adriani PP, Cunha-Junior JP, Astray RM, Catarino RM, Lancelotti M, Prudencio CR. RBD and Spike DNA-Based Immunization in Rabbits Elicited IgG Avidity Maturation and High Neutralizing Antibody Responses against SARS-CoV-2. Viruses 2023; 15:555. [PMID: 36851769 PMCID: PMC9959588 DOI: 10.3390/v15020555] [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: 12/24/2022] [Revised: 01/23/2023] [Accepted: 02/11/2023] [Indexed: 02/19/2023] Open
Abstract
Neutralizing antibodies (nAbs) are a critical part of coronavirus disease 2019 (COVID-19) research as they are used to gain insight into the immune response to severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) infections. Among the technologies available for generating nAbs, DNA-based immunization methods are an alternative to conventional protocols. In this pilot study, we investigated whether DNA-based immunization by needle injection in rabbits was a viable approach to produce a functional antibody response. We demonstrated that three doses of DNA plasmid carrying the gene encoding the full-length spike protein (S) or the receptor binding domain (RBD) of SARS-CoV-2 induced a time-dependent increase in IgG antibody avidity maturation. Moreover, the IgG antibodies displayed high cross neutralization by live SARS-CoV-2 and pseudoviruses neutralization assays. Thus, we established a simple, low cost and feasible DNA-based immunization protocol in rabbits that elicited high IgG avidity maturation and nAbs production against SARS-CoV-2, highlighting the importance of DNA-based platforms for developing new immunization strategies against SARS-CoV-2 and future emerging epidemics.
Collapse
Affiliation(s)
- Hernan H. M. da Costa
- Immunology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
- Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Diego J. B. Orts
- Immunology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
| | - Andrew D. Moura
- Immunology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
| | | | | | - Rodrigo A. Réssio
- Pathology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
| | | | - Karen Miguita
- Pathology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
| | | | | | - Patricia P. Adriani
- Skinzymes Biotechnology Ltd., São Paulo 05441-040, Brazil
- Laboratory of Nanopharmaceuticals and Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Jair P. Cunha-Junior
- Laboratory of Immunochemistry and Immunotechnology, Department of Immunology, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
| | - Renato M. Astray
- Multi-Purpose Laboratory, Butantan Institute, São Paulo 05503-900, Brazil
| | | | - Marcelo Lancelotti
- Faculty of Pharmaceutical Sciences, Campinas State University, Campinas 13083-871, Brazil
| | - Carlos R. Prudencio
- Immunology Center, Institute Adolfo Lutz, São Paulo 01246-902, Brazil
- Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
8
|
Mougenot MF, Pereira VS, Costa ALR, Lancellotti M, Porcionatto MA, da Silveira JC, de la Torre LG. Biomimetic Nanovesicles—Sources, Design, Production Methods, and Applications. Pharmaceutics 2022; 14:pharmaceutics14102008. [PMID: 36297442 PMCID: PMC9610935 DOI: 10.3390/pharmaceutics14102008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Despite all the progress in the field of liposomes and nanoparticles for applications as drug and gene delivery systems, the specific targeting and immune system escape capabilities of these systems are still limited. Biomimetic nanovesicles emerged as a strategy to overcome these and other limitations associated with synthetic carriers, such as short circulation time, cytotoxicity, and difficulty in crossing biological barriers, since many of the desirable abilities of drug delivery systems are innate characteristics of biological vesicles. Thus, the question arises: would biomimetic nanovesicles be responsible for addressing these advances? It is currently known that biomimetic nanovesicles (BNV) can combine the intrinsic advantages of natural materials with the well-known production methods and controllability of synthetic systems. Besides, the development of the biotechnology and nanotechnology fields has provided a better understanding of the functionalities of biological vesicles and the means for the design and production of biomimetic nanovesicles (BNV). Based on this, this work will focus on tracking the main research on biomimetic nanovesicles (BNV) applied as drug and gene delivery systems, and for vaccines applications. In addition, it will describe the different sources of natural vesicles, the technical perspectives on obtaining them, and the possibility of their hybridization with synthetic liposomes.
Collapse
Affiliation(s)
- Marcel Franco Mougenot
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
| | - Vanessa Sousa Pereira
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
| | - Ana Letícia Rodrigues Costa
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
- Institute of Exact and Technological Sciences, Campus Florestal, Federal University of Viçosa (UFV), Florestal 35690-000, Brazil
| | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas 13083-871, Brazil
| | | | - Juliano Coelho da Silveira
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga 13635-900, Brazil
| | - Lucimara Gaziola de la Torre
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas 13083-970, Brazil
- Correspondence: ; Tel.: +55-19-3521-0397
| |
Collapse
|
9
|
Kashyap D, Panda M, Baral B, Varshney N, R S, Bhandari V, Parmar HS, Prasad A, Jha HC. Outer Membrane Vesicles: An Emerging Vaccine Platform. Vaccines (Basel) 2022; 10:1578. [PMID: 36298443 PMCID: PMC9610665 DOI: 10.3390/vaccines10101578] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 11/08/2023] Open
Abstract
Vaccine adjuvants are substances that improve the immune capacity of a recombinant vaccine to a great extent and have been in use since the early 1900s; they are primarily short-lived and initiate antigen activity, mainly an inflammatory response. With the developing technologies and innovation, early options such as alum were modified, yet the inorganic nature of major vaccine adjuvants caused several side effects. Outer membrane vesicles, which respond to the stressed environment, are small nano-sized particles secreted by gram-negative bacteria. The secretory nature of OMV gives us many benefits in terms of infection bioengineering. This article aims to provide a detailed overview of bacteria's outer membrane vesicles (OMV) and their potential usage as adjuvants in making OMV-based vaccines. The OMV adjuvant-based vaccines can be a great benefactor, and there are ongoing trials for formulating OMV adjuvant-based vaccines for SARS-CoV-2. This study emphasizes engineering the OMVs to develop better versions for safety purposes. This article will also provide a gist about the advantages and disadvantages of such vaccines, along with other aspects.
Collapse
Affiliation(s)
- Dharmendra Kashyap
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, India
| | - Mrutyunjaya Panda
- Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, India
| | - Budhadev Baral
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, India
| | - Nidhi Varshney
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, India
| | - Sajitha R
- Amity Institute of Biotechnology, Amity University Noida, Amity 201313, India
| | - Vasundhra Bhandari
- Department of Biological Science, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, India
| | | | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi 175005, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, India
| |
Collapse
|
10
|
Puricelli C, Boggio E, Gigliotti CL, Stoppa I, Sutti S, Rolla R, Dianzani U. Cutting-Edge Delivery Systems and Adjuvants in Tolerogenic Vaccines: A Review. Pharmaceutics 2022; 14:1782. [PMID: 36145531 PMCID: PMC9501480 DOI: 10.3390/pharmaceutics14091782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Conventional therapies for immune-mediated diseases, including autoimmune disorders, transplant reactions, and allergies, have undergone a radical evolution in the last few decades; however, they are still not specific enough to avoid widespread immunosuppression. The idea that vaccine usage could be extended beyond its traditional immunogenic function by encompassing the ability of vaccines to induce antigen-specific tolerance may revolutionize preventive and therapeutic strategies in several clinical fields that deal with immune-mediated disorders. This approach has been supported by improved data relating to the several mechanisms involved in controlling unwanted immune responses and allowing peripheral tolerance. Given these premises, several approaches have been developed to induce peripheral tolerance against the antigens that are involved in the pathological immune response, including allergens, autoantigens, and alloantigens. Technological innovations, such as nucleic acid manipulation and the advent of micro- and nanoparticles, have further supported these novel preventive and therapeutic approaches. This review focuses on the main strategies used in the development of tolerogenic vaccines, including the technological issues used in their design and the role of "inverse adjuvants". Even though most studies are still limited to the preclinical field, the enthusiasm generated by their results has prompted some initial clinical trials, and they show great promise for the future management of immune-mediated pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Roberta Rolla
- Department of Health Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| | | |
Collapse
|
11
|
Qin S, Xiao W, Zhou C, Pu Q, Deng X, Lan L, Liang H, Song X, Wu M. Pseudomonas aeruginosa: pathogenesis, virulence factors, antibiotic resistance, interaction with host, technology advances and emerging therapeutics. Signal Transduct Target Ther 2022; 7:199. [PMID: 35752612 PMCID: PMC9233671 DOI: 10.1038/s41392-022-01056-1] [Citation(s) in RCA: 510] [Impact Index Per Article: 170.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 06/04/2022] [Accepted: 06/08/2022] [Indexed: 02/05/2023] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a Gram-negative opportunistic pathogen that infects patients with cystic fibrosis, burn wounds, immunodeficiency, chronic obstructive pulmonary disorder (COPD), cancer, and severe infection requiring ventilation, such as COVID-19. P. aeruginosa is also a widely-used model bacterium for all biological areas. In addition to continued, intense efforts in understanding bacterial pathogenesis of P. aeruginosa including virulence factors (LPS, quorum sensing, two-component systems, 6 type secretion systems, outer membrane vesicles (OMVs), CRISPR-Cas and their regulation), rapid progress has been made in further studying host-pathogen interaction, particularly host immune networks involving autophagy, inflammasome, non-coding RNAs, cGAS, etc. Furthermore, numerous technologic advances, such as bioinformatics, metabolomics, scRNA-seq, nanoparticles, drug screening, and phage therapy, have been used to improve our understanding of P. aeruginosa pathogenesis and host defense. Nevertheless, much remains to be uncovered about interactions between P. aeruginosa and host immune responses, including mechanisms of drug resistance by known or unannotated bacterial virulence factors as well as mammalian cell signaling pathways. The widespread use of antibiotics and the slow development of effective antimicrobials present daunting challenges and necessitate new theoretical and practical platforms to screen and develop mechanism-tested novel drugs to treat intractable infections, especially those caused by multi-drug resistance strains. Benefited from has advancing in research tools and technology, dissecting this pathogen's feature has entered into molecular and mechanistic details as well as dynamic and holistic views. Herein, we comprehensively review the progress and discuss the current status of P. aeruginosa biophysical traits, behaviors, virulence factors, invasive regulators, and host defense patterns against its infection, which point out new directions for future investigation and add to the design of novel and/or alternative therapeutics to combat this clinically significant pathogen.
Collapse
Affiliation(s)
- Shugang Qin
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Xiao
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chuanmin Zhou
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan, 430071, P.R. China
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Qinqin Pu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Xin Deng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
| | - Lefu Lan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Haihua Liang
- College of Life Sciences, Northwest University, Xi'an, ShaanXi, 710069, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Min Wu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA.
| |
Collapse
|
12
|
Krishnan N, Kubiatowicz LJ, Holay M, Zhou J, Fang RH, Zhang L. Bacterial membrane vesicles for vaccine applications. Adv Drug Deliv Rev 2022; 185:114294. [PMID: 35436569 DOI: 10.1016/j.addr.2022.114294] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/13/2022] [Accepted: 04/10/2022] [Indexed: 12/11/2022]
Abstract
Vaccines have been highly successful in the management of many diseases. However, there are still numerous illnesses, both infectious and noncommunicable, for which there are no clinically approved vaccine formulations. While there are unique difficulties that must be overcome in the case of each specific disease, there are also a number of common challenges that have to be addressed for effective vaccine development. In recent years, bacterial membrane vesicles (BMVs) have received increased attention as a potent and versatile vaccine platform. BMVs are inherently immunostimulatory and are able to activate both innate and adaptive immune responses. Additionally, BMVs can be readily taken up and processed by immune cells due to their nanoscale size. Finally, BMVs can be modified in a variety of ways, including by genetic engineering, cargo loading, and nanoparticle coating, in order to create multifunctional platforms that can be leveraged against different diseases. Here, an overview of the interactions between BMVs and immune cells is provided, followed by discussion on the applications of BMV vaccine nanotechnology against bacterial infections, viral infections, and cancers.
Collapse
Affiliation(s)
- Nishta Krishnan
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Luke J Kubiatowicz
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Maya Holay
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
13
|
Liu S, Wu X, Chandra S, Lyon C, Ning B, jiang L, Fan J, Hu TY. Extracellular vesicles: Emerging tools as therapeutic agent carriers. Acta Pharm Sin B 2022; 12:3822-3842. [PMID: 36213541 PMCID: PMC9532556 DOI: 10.1016/j.apsb.2022.05.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/02/2022] [Accepted: 04/28/2022] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) are secreted by both eukaryotes and prokaryotes, and are present in all biological fluids of vertebrates, where they transfer DNA, RNA, proteins, lipids, and metabolites from donor to recipient cells in cell-to-cell communication. Some EV components can also indicate the type and biological status of their parent cells and serve as diagnostic targets for liquid biopsy. EVs can also natively carry or be modified to contain therapeutic agents (e.g., nucleic acids, proteins, polysaccharides, and small molecules) by physical, chemical, or bioengineering strategies. Due to their excellent biocompatibility and stability, EVs are ideal nanocarriers for bioactive ingredients to induce signal transduction, immunoregulation, or other therapeutic effects, which can be targeted to specific cell types. Herein, we review EV classification, intercellular communication, isolation, and characterization strategies as they apply to EV therapeutics. This review focuses on recent advances in EV applications as therapeutic carriers from in vitro research towards in vivo animal models and early clinical applications, using representative examples in the fields of cancer chemotherapeutic drug, cancer vaccine, infectious disease vaccines, regenerative medicine and gene therapy. Finally, we discuss current challenges for EV therapeutics and their future development.
Collapse
|
14
|
Bacteria and bacterial derivatives as delivery carriers for immunotherapy. Adv Drug Deliv Rev 2022; 181:114085. [PMID: 34933064 DOI: 10.1016/j.addr.2021.114085] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023]
Abstract
There is growing interest in the role of microorganisms in human health and disease, with evidence showing that new types of biotherapy using engineered bacterial therapeutics, including bacterial derivatives, can address specific mechanisms of disease. The complex interactions between microorganisms and metabolic/immunologic pathways underlie many diseases with unmet medical needs, suggesting that targeting these interactions may improve patient treatment. Using tools from synthetic biology and chemical engineering, non-pathogenic bacteria or bacterial products can be programmed and designed to sense and respond to environmental signals to deliver therapeutic effectors. This review describes current progress in biotherapy using live bacteria and their derivatives to achieve therapeutic benefits against various diseases.
Collapse
|
15
|
Baig MMFA, Dissanayaka WL, Zhang C. 2D DNA nanoporous scaffold promotes osteogenic differentiation of pre-osteoblasts. Int J Biol Macromol 2021; 188:657-669. [PMID: 34371047 DOI: 10.1016/j.ijbiomac.2021.07.198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/23/2021] [Accepted: 07/31/2021] [Indexed: 01/06/2023]
Abstract
Biofunctional materials with nanomechanical parameters similar to bone tissue may promote the adherence, migration, proliferation, and differentiation of pre-osteoblasts. In this study, deoxyribonucleic acid (DNA) nanoporous scaffold (DNA-NPS) was synthesized by the polymerization of rectangular and double-crossover (DX) DNA tiles. The diagonally precise polymerization of nanometer-sized DNA tiles (A + B) through sticky end cohesion gave rise to a micrometer-sized porous giant-sheet material. The synthesized DNA-NPS exhibited a uniformly distributed porosity with a size of 25 ± 20 nm. The morphology, dimensions, sectional profiles, 2-dimensional (2D) layer height, texture, topology, pore size, and mechanical parameters of DNA-NPS have been characterized by atomic force microscopy (AFM). The size and zeta potential of DNA-NPS have been characterized by the zeta sizer. Cell biocompatibility, proliferation, and apoptosis have been evaluated by flow cytometry. The AFM results confirmed that the fabricated DNA-NPS was interconnected and uniformly porous, with a surface roughness of 0.125 ± 0.08035 nm. The elastic modulus of the DNA-NPS was 22.45 ± 8.65 GPa, which was comparable to that of native bone tissue. DNA-NPS facilitated pre-osteoblast adhesion, proliferation, and osteogenic differentiation. These findings indicated the potential of 2D DNA-NPS in promoting bone tissue regeneration.
Collapse
Affiliation(s)
| | - Waruna Lakmal Dissanayaka
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
16
|
Nano-Microparticle Platforms in Developing Next-Generation Vaccines. Vaccines (Basel) 2021; 9:vaccines9060606. [PMID: 34198865 PMCID: PMC8228777 DOI: 10.3390/vaccines9060606] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
The first vaccines ever made were based on live-attenuated or inactivated pathogens, either whole cells or fragments. Although these vaccines required the co-administration of antigens with adjuvants to induce a strong humoral response, they could only elicit a poor CD8+ T-cell response. In contrast, next-generation nano/microparticle-based vaccines offer several advantages over traditional ones because they can induce a more potent CD8+ T-cell response and, at the same time, are ideal carriers for proteins, adjuvants, and nucleic acids. The fact that these nanocarriers can be loaded with molecules able to modulate the immune response by inducing different effector functions and regulatory activities makes them ideal tools for inverse vaccination, whose goal is to shut down the immune response in autoimmune diseases. Poly (lactic-co-glycolic acid) (PLGA) and liposomes are biocompatible materials approved by the Food and Drug Administration (FDA) for clinical use and are, therefore, suitable for nanoparticle-based vaccines. Recently, another candidate platform for innovative vaccines based on extracellular vesicles (EVs) has been shown to efficiently co-deliver antigens and adjuvants. This review will discuss the potential use of PLGA-NPs, liposomes, and EVs as carriers of peptides, adjuvants, mRNA, and DNA for the development of next-generation vaccines against endemic and emerging viruses in light of the recent COVID-19 pandemic.
Collapse
|
17
|
Duarte JL, Filippo LDD, Araujo VHS, Oliveira AEMDFM, de Araújo JTC, Silva FBDR, Pinto MC, Chorilli M. Nanotechnology as a tool for detection and treatment of arbovirus infections. Acta Trop 2021; 216:105848. [PMID: 33524384 DOI: 10.1016/j.actatropica.2021.105848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 12/19/2022]
Abstract
Arboviruses are medically important viruses that cause high rates of infection all over the world. In addition, the severity of the symptoms and the inadequate diagnostic methods represent a challenge far beyond eradicating the vector. The lack of specific treatments for arbovirus infections reflects the imminent need for new research for safe and efficient medicines to treat these infections. Nanotechnology is an innovative approach currently used as a platform for developing new treatments, thus improving the biopharmaceutical properties of drugs. It can also be applied to the development of diagnostic devices, improving their detection capacity. The purpose of this paper is to review recent research on the use of nanotechnology for developing new treatments and detection devices for arbovirus infections. Interestingly, it was found that only a few studies report on the use of nanotechnology to treat arbovirus infections and that most of these reports focus on the fabrication of diagnostic tools. Also, some papers report on the use of nanotechnology for the development of vaccines, which in association with mosquito eradication programs could effectively reduce the high rates of infections by these viruses.
Collapse
Affiliation(s)
- Jonatas Lobato Duarte
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Leonardo Delello Di Filippo
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Victor Hugo Sousa Araujo
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Anna Eliza Maciel de Faria Mota Oliveira
- Federal University of Amapá - UNIFAP, Department of Health and biological sciences, Rodovia Juscelino Kubitschek, Km 02, Jardim Marco Zero, Macapá-AP, 68903-361, Brazil
| | - Jennifer Thayanne Cavalcante de Araújo
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Flávia Benini da Rocha Silva
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Mara Cristina Pinto
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Marlus Chorilli
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil.
| |
Collapse
|
18
|
Nanoparticles as Vaccines to Prevent Arbovirus Infection: A Long Road Ahead. Pathogens 2021; 10:pathogens10010036. [PMID: 33466440 PMCID: PMC7824877 DOI: 10.3390/pathogens10010036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022] Open
Abstract
Arthropod-borne viruses (arboviruses) are a significant public health problem worldwide. Vaccination is considered one of the most effective ways to control arbovirus diseases in the human population. Nanoparticles have been widely explored as new vaccine platforms. Although nanoparticles' potential to act as new vaccines against infectious diseases has been identified, nanotechnology's impact on developing new vaccines to prevent arboviruses is unclear. Thus, we used a comprehensive bibliographic survey to integrate data concerning the use of diverse nanoparticles as vaccines against medically important arboviruses. Our analysis showed that considerable research had been conducted to develop and evaluate nanovaccines against Chikungunya virus, Dengue virus, Zika virus, Japanese encephalitis virus, and West Nile virus. The main findings indicate that nanoparticles have great potential for use as a new vaccine system against arboviruses. Most of the studies showed an increase in neutralizing antibody production after mouse immunization. Nevertheless, even with significant advances in this field, further efforts are necessary to address the nanoparticles' potential to act as a vaccine against these arboviruses. To promote advances in the field, we proposed a roadmap to help researchers better characterize and evaluate nanovaccines against medically important arboviruses.
Collapse
|
19
|
Molecular signatures associated with prostate cancer cell line (PC-3) exposure to inactivated Zika virus. Sci Rep 2019; 9:15351. [PMID: 31653965 PMCID: PMC6814752 DOI: 10.1038/s41598-019-51954-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023] Open
Abstract
The recent outbreak of Zika virus (ZIKV) infection associated with microcephaly cases has elicited much research on the mechanisms involved in ZIKV-host cell interactions. It has been described that Zika virus impairs cell growth, raising a hypothesis about its oncolytic potential against cancer cells. ZIKV tumor cell growth inhibition was later confirmed for glioblastoma. It was also demonstrated that an inactivated ZIKV prototype (ZVp) based on bacterial outer membrane vesicles has antiproliferative activity upon other cancer cell lines, such as PC-3 prostate cancer cell. This study aims at understanding the pathways that might be involved with the antiproliferative effect of Zika virus against prostate cancer cells. A metabolomic approach based on high-resolution mass spectrometry analysis led to the identification of 21 statistically relevant markers of PC-3 cells treated with ZVp. The markers were associated with metabolic alterations that trigger lipid remodeling, endoplasmic reticulum stress, inflammatory mediators, as well as disrupted porphyrin and folate metabolism. These findings highlight molecular signatures of ZVp-induced response that may be involved on cellular pathways triggered by its antiproliferative effect. To our knowledge, this is the first reported metabolomic assessment of ZIKV effect on prostate cancer cells, a promising topic for further research.
Collapse
|
20
|
A new level of complexity in parasite-host interaction: The role of extracellular vesicles. ADVANCES IN PARASITOLOGY 2019; 104:39-112. [PMID: 31030771 DOI: 10.1016/bs.apar.2019.02.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Humans and animals have co-existed with parasites in a battle of constant adaptation to one another. It is becoming increasingly clear that extracellular vesicles (EVs) play important roles in this co-existence and pathology. This chapter reviews the current research on EVs released by protozoa, nematodes, trematodes, and cestodes with a special focus on EVs in parasite life cycles. The environmental changes experienced by the parasite during its life cycle is associated with distinct changes in EV release and content. The function of these EV seems to have a significant influence on parasite pathology and survival in the host by concomitantly modulating host immune responses and triggering parasite differentiation. The role of EVs in communication between the parasites and the host adds a new level of complexity in our understanding of parasite biology, which may be a key to further understand the complexity behind host-parasite interactions and communication. This increased understanding can, in turn, open up new avenues for vaccine, diagnostic, and therapeutic development for a wide variety of diseases such as parasite infection, cancers, and immunological disorders.
Collapse
|
21
|
Ghaffar KA, Ng LFP, Renia L. Fast Tracks and Roadblocks for Zika Vaccines. Vaccines (Basel) 2018; 6:vaccines6040077. [PMID: 30469444 PMCID: PMC6313897 DOI: 10.3390/vaccines6040077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 01/07/2023] Open
Abstract
In early 2014, a relatively obscure virus, the Zika virus, made headlines worldwide following an increase in the number of congenital malformations. Since then, research on Zika virus, treatment and vaccines have progressed swiftly with various drugs being repurposed and vaccines heading into clinical trials. Nonetheless, the need for a vaccine is crucial in order to eradicate this re-emerging arthropod-borne virus which remained silent since its first discovery in 1947. In this review, we focused on how the inconspicuous virus managed to spread, the key immunological factors required for a vaccine and the various vaccine platforms that are currently being studied.
Collapse
Affiliation(s)
- Khairunnisa Abdul Ghaffar
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
- Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7BE, UK.
| | - Laurent Renia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
| |
Collapse
|
22
|
Jesus S, Soares E, Cruz MT, Borges O. Exosomes as adjuvants for the recombinant hepatitis B antigen: First report. Eur J Pharm Biopharm 2018; 133:1-11. [PMID: 30287267 DOI: 10.1016/j.ejpb.2018.09.029] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/10/2018] [Accepted: 09/28/2018] [Indexed: 01/11/2023]
Abstract
Over the past few years, exosomes, a class of extracellular vesicles (EVs), have emerged as key players for inter-cellular communication ultimately modulating the behavior of target cells with countless outcomes. Nevertheless, the potential role of exosomes as vaccine adjuvants remains largely unexplored. Herein, we hypothesized that exosomes derived from immune cells may have an immunostimulatory effect and could constitute a good target towards the development of new fine-tuned vaccine adjuvants. To accomplish this goal, exosomes isolated from lipopolysaccharide endotoxin (LPS)-stimulated human monocytic cell line (THP-1) were characterized and tested for their non-specific immunostimulatory activity when administered subcutaneously to healthy mice; additionally, exosomes' vaccine adjuvant ability was also disclosed after their inclusion in vaccine formulations. The results obtained suggested that the isolated exosomes evoked a pro-inflammatory profile in spleen cells of healthy mice through the induction of cytokines such as tumor necrosis factor alpha (TNF-α), chemokine (C-C motif) ligand 5 (CCL5, also known as RANTES) and interleukin 1 beta (IL-1β). Moreover, subcutaneous vaccination of mice with exosomes combined with a solution of hepatitis B recombinant antigen (HBsAg) or combined with a suspension containing HBsAg loaded poly-ε-caprolactone (PCL)/chitosan nanoparticles (NPs), induced a humoral immune response quite similar to the one achieved with the experimental control group (HBsAg solution without exosomes). However, exosomes triggered an immunomodulator effect on the cellular immune response, highlighted by the enhancement of IFN-γ secretion. To the best of authors knowledge, this is the first report describing extensively the role of unmodified exosomes as adjuvants and co-adjuvants for hepatitis B vaccination strategies.
Collapse
Affiliation(s)
- Sandra Jesus
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, 1st floor, 3004-504 Coimbra, Portugal
| | - Edna Soares
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, 1st floor, 3004-504 Coimbra, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, 1st floor, 3004-504 Coimbra, Portugal
| | - Olga Borges
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, 1st floor, 3004-504 Coimbra, Portugal.
| |
Collapse
|