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 multifaceted functions as contributors both to disease and as carriers of therapeutic possibilities. We examine the intricate interactions of BEVs within bacterial communities and between bacteria and hosts, their involvement in disease development through cargo delivery mechanisms, and their beneficial impact to microbial ecology. The review places a strong emphasis on BEVs' applications in biomedical sciences, where they are revolutionizing vaccine development, targeted drug delivery, and cancer therapy. By utilizing the inherent properties of BEVs for controlled drug release, targeted antigen delivery, and immune modulation, they offer a promising frontier in precision medicine. In addition, the diagnostic potential of BEVs is explored through their biomarker capabilities, providing valuable insights into disease states and treatment efficacy. Looking forward, this review underscores the challenges and opportunities in translating BEV research to clinical practice, promoting the use of standardized methods in BEV characterization and scaling up production. The diverse abilities of BEVs, ranging from contributing to pathogen virulence to driving therapeutic innovation, highlight their potential as a cornerstone in the future of biomedical advancements. STATEMENT OF SIGNIFICANCE: Bacterial extracellular vesicles (BEVs) are emerging as pivotal players in both pathogenesis and therapeutic innovation. This review explores their dual nature as agents of disease and as promising biomaterials in biomedical applications, and provides a comprehensive survey on their involvement in disease mechanisms and microbial ecology, and their potential in biomedical applications such as vaccine development, targeted drug delivery, cancer therapy and diagnosis. It highlights the complex interactions of BEVs within bacterial communities and between bacteria and hosts. This review also addresses current advancements, challenges, and opportunities in translating BEV research into clinical practice. The insights presented here position BEVs as a cornerstone in the future of biomedical advancements, advocating for standardized methods in BEV characterization and scalable production techniques.
Collapse
Affiliation(s)
| | - Ashkan Dehghan
- W Booth School of Engineering Practice and Technology Faculty of Engineering, McMaster University Hamilton, ON, Canada, L8S 0A3
| | - Saba Halimi
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Fatemeh Najafi
- Department of Chemical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802-1503, United States
| | - Ali Nokhostin
- Medical Sciences & Technologies Faculty, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | | | - Iman Akbarzadeh
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| | - Qun Ren
- Laboratory for Biointerfaces, Empa Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland.
| |
Collapse
|
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
|
Zhu D, Zhang Y, Wang Z, Dai J, Zhuge X. Exploiting membrane vesicles derived from avian pathogenic Escherichia coli as a cross-protective subunit vaccine candidate against avian colibacillosis. Poult Sci 2024; 103:104148. [PMID: 39142031 PMCID: PMC11379662 DOI: 10.1016/j.psj.2024.104148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/06/2024] [Accepted: 07/26/2024] [Indexed: 08/16/2024] Open
Abstract
Avian pathogenic Escherichia coli (APEC) is a notable pathogen that frequently leads to avian colibacillosis, posing a substantial risk to both the poultry industry and public health. The commercial vaccines against avian colibacillosis are primarily inactivated vaccines, but their effectiveness is limited to specific serotypes. Recent advances have highlighted bacterial membrane vesicles (MV) as a promising candidate in vaccine research. How to produce bacterial MVs vaccines on a large scale is a significant challenge for the industrialization of MVs. The msbB gene encodes an acyltransferase and has been implicated in altering the acylation pattern of lipid A, leading to a decrease in lipid A content in lipopolysaccharides (LPS). Here, we evaluated the immunoprotective efficacy of MVs derived from the LPS low-expressed APEC strain FY26ΔmsbB, which was an APEC mutant strain with a deletion of the msbB gene. The nitrogen cavitation technique was employed to extract APEC MVs, with results indicating a significant increase in MVs yield compared to that obtained under natural culture. The immunization effectiveness was assessed, revealing that FY26ΔmsbB MVs elicited an antibody response of laying hens and facilitated bacterial clearance. Protective efficacy studies demonstrated that immunization with FY26ΔmsbB MVs conferred the immune protection in chickens challenged with the wild-type APEC strain FY26. Notably, LPS low-carried MVs recovered from the mutant FY26ΔmsbB also displayed cross-protective capabilities, and effectively safeguarding against infections caused by O1, O7, O45, O78, and O101 serotypes virulent APEC strains. These findings suggest that MVs generated from the LPS low-expressed APEC strain FY26ΔmsbB represent a novel and empirically validated subunit vaccine for the prevention and control of infections by various APEC serotypes.
Collapse
Affiliation(s)
- Dongyu Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226019, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuting Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226019, China
| | - Zhongxing Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226019, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jianjun Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangkai Zhuge
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu 226019, China.
| |
Collapse
|
4
|
Effah CY, Ding X, Drokow EK, Li X, Tong R, Sun T. Bacteria-derived extracellular vesicles: endogenous roles, therapeutic potentials and their biomimetics for the treatment and prevention of sepsis. Front Immunol 2024; 15:1296061. [PMID: 38420121 PMCID: PMC10899385 DOI: 10.3389/fimmu.2024.1296061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Sepsis is one of the medical conditions with a high mortality rate and lacks specific treatment despite several years of extensive research. Bacterial extracellular vesicles (bEVs) are emerging as a focal target in the pathophysiology and treatment of sepsis. Extracellular vesicles (EVs) derived from pathogenic microorganisms carry pathogenic factors such as carbohydrates, proteins, lipids, nucleic acids, and virulence factors and are regarded as "long-range weapons" to trigger an inflammatory response. In particular, the small size of bEVs can cross the blood-brain and placental barriers that are difficult for pathogens to cross, deliver pathogenic agents to host cells, activate the host immune system, and possibly accelerate the bacterial infection process and subsequent sepsis. Over the years, research into host-derived EVs has increased, leading to breakthroughs in cancer and sepsis treatments. However, related approaches to the role and use of bacterial-derived EVs are still rare in the treatment of sepsis. Herein, this review looked at the dual nature of bEVs in sepsis by highlighting their inherent functions and emphasizing their therapeutic characteristics and potential. Various biomimetics of bEVs for the treatment and prevention of sepsis have also been reviewed. Finally, the latest progress and various obstacles in the clinical application of bEVs have been highlighted.
Collapse
Affiliation(s)
- Clement Yaw Effah
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Xianfei Ding
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Emmanuel Kwateng Drokow
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Department of Epidemiology and Biostatistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Xiang Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Ran Tong
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| | - Tongwen Sun
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou, China
| |
Collapse
|
5
|
Abstract
Outer membrane vesicles (OMVs) are spontaneously released by many gram-negative bacteria during their growth and constitute an important virulence factor for bacteria, helping them to survive through harsh environmental conditions. Native OMVs, naturally-released from bacteria, are produced at a level too low for vaccine manufacturing, requiring chemical treatment (detergent-extracted) or genetic manipulation, resulting in generalized modules for membrane antigens (GMMAs). Over the years, the nature and properties of OMVs have made them a viable platform for vaccine development. There are a few licensed OMV vaccines mainly for the prevention of meningitis caused by Neisseria meningitidis serogroup B (MenB) and Haemophilus influenzae type b (Hib). There are several candidates in clinical development against other gram-negative organisms from which the OMVs are derived, but also against heterologous targets in which the OMVs are used as carriers (e.g. coronavirus disease 2019 [COVID-19]). The use of OMVs for targets other than those from which they are derived is a major advancement in OMV technology, improving its versatility by being able to deliver protein or polysaccharide antigens. Other advances include the range of genetic modifications that can be made to improve their safety, reduce reactogenicity, and increase immunogenicity and protective efficacy. However, significant challenges remain, such as identification of general tools for high-content surface expression of heterologous proteins on the OMV surface. Here, we outline the progress of OMV vaccines to date, particularly discussing licensed OMV-based vaccines and candidates in clinical development. Recent trends in preclinical research are described, mainly focused on genetic manipulation and chemical conjugation for the use of OMVs as carriers for heterologous protein and polysaccharide antigens. Remaining challenges with the use of OMVs and directions for future research are also discussed.
Collapse
Affiliation(s)
- Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy.
| | | | - Usman Nakakana
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
6
|
Ko HJ, Kim YJ. Antigen Delivery Systems: Past, Present, and Future. Biomol Ther (Seoul) 2023; 31:370-387. [PMID: 37072288 PMCID: PMC10315343 DOI: 10.4062/biomolther.2023.006] [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: 01/09/2023] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 04/20/2023] Open
Abstract
The COVID-19 pandemic has increased demand for safe and effective vaccines. Research to develop vaccines against diseases including Middle East respiratory syndrome, Ebolavirus, human immunodeficiency virus, and various cancers would also contribute to global well-being. For successful vaccine development, the advancement of technologies such as antigen (Ag) screening, Ag delivery systems and adjuvants, and manufacturing processes is essential. Ag delivery systems are required not only to deliver a sufficient amount of Ag for vaccination, but also to enhance immune response. In addition, Ag types and their delivery systems determine the manufacturing processes of the vaccine product. Here, we analyze the characteristics of various Ag delivery systems: plasmids, viral vectors, bacterial vectors, nanoparticles, self-assembled particles, natural and artificial cells, and extracellular vesicles. This review provides insight into the current vaccine landscape and highlights promising avenues of research for the development and improvement of Ag delivery systems.
Collapse
Affiliation(s)
- Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yeon-Jeong Kim
- Laboratory of Microbiology and Immunology, College of Pharmacy, Inje University, Gimhae 50834, Republic of Korea
- Inje Institute of Pharmaceutical Science and Research, Inje University, Gimhae 50834, Republic of Korea
- Smart Marine Therapeutic Center, Inje University, Gimhae 50834, Republic of Korea
| |
Collapse
|
7
|
Thapa HB, Ebenberger SP, Schild S. The Two Faces of Bacterial Membrane Vesicles: Pathophysiological Roles and Therapeutic Opportunities. Antibiotics (Basel) 2023; 12:1045. [PMID: 37370364 PMCID: PMC10295235 DOI: 10.3390/antibiotics12061045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Bacterial membrane vesicles (MVs) are nanosized lipid particles secreted by lysis or blebbing mechanisms from Gram-negative and -positive bacteria. It is becoming increasingly evident that MVs can promote antimicrobial resistance but also provide versatile opportunities for therapeutic exploitation. As non-living facsimiles of parent bacteria, MVs can carry multiple bioactive molecules such as proteins, lipids, nucleic acids, and metabolites, which enable them to participate in intra- and interspecific communication. Although energetically costly, the release of MVs seems beneficial for bacterial fitness, especially for pathogens. In this review, we briefly discuss the current understanding of diverse MV biogenesis routes affecting MV cargo. We comprehensively highlight the physiological functions of MVs derived from human pathogens covering in vivo adaptation, colonization fitness, and effector delivery. Emphasis is given to recent findings suggesting a vicious cycle of MV biogenesis, pathophysiological function, and antibiotic therapy. We also summarize potential therapeutical applications, such as immunotherapy, vaccination, targeted delivery, and antimicrobial potency, including their experimental validation. This comparative overview identifies common and unique strategies for MV modification used along diverse applications. Thus, the review summarizes timely aspects of MV biology in a so far unprecedented combination ranging from beneficial function for bacterial pathogen survival to future medical applications.
Collapse
Affiliation(s)
- Himadri B. Thapa
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Stephan P. Ebenberger
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Stefan Schild
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
- BioTechMed Graz, 8010 Graz, Austria
- Field of Excellence Biohealth, University of Graz, 8010 Graz, Austria
| |
Collapse
|
8
|
Durojaye OA, Okoro NO, Odiba AS, Nwanguma BC. MasitinibL shows promise as a drug-like analog of masitinib that elicits comparable SARS-Cov-2 3CLpro inhibition with low kinase preference. Sci Rep 2023; 13:6972. [PMID: 37117213 PMCID: PMC10141821 DOI: 10.1038/s41598-023-33024-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/06/2023] [Indexed: 04/30/2023] Open
Abstract
SARS-CoV-2 infection has led to several million deaths worldwide and ravaged the economies of many countries. Hence, developing therapeutics against SARS-CoV-2 remains a core priority in the fight against COVID-19. Most of the drugs that have received emergency use authorization for treating SARS-CoV-2 infection exhibit a number of limitations, including side effects and questionable efficacy. This challenge is further compounded by reinfection after vaccination and the high likelihood of mutations, as well as the emergence of viral escape mutants that render SARS-CoV-2 spike glycoprotein-targeting vaccines ineffective. Employing de novo drug synthesis or repurposing to discover broad-spectrum antivirals that target highly conserved pathways within the viral machinery is a focus of current research. In a recent drug repurposing study, masitinib, a clinically safe drug against the human coronavirus OC43 (HCoV-OC43), was identified as an antiviral agent with effective inhibitory activity against the SARS-CoV-2 3CLpro. Masitinib is currently under clinical trial in combination with isoquercetin in hospitalized patients (NCT04622865). Nevertheless, masitinib has kinase-related side effects; hence, the development of masitinib analogs with lower anti-tyrosine kinase activity becomes necessary. In this study, in an attempt to address this limitation, we executed a comprehensive virtual workflow in silico to discover drug-like compounds matching selected pharmacophore features in the SARS-CoV-2 3CLpro-bound state of masitinib. We identified a novel lead compound, "masitinibL", a drug-like analog of masitinib that demonstrated strong inhibitory properties against the SARS-CoV-2 3CLpro. In addition, masitinibL further displayed low selectivity for tyrosine kinases, which strongly suggests that masitinibL is a highly promising therapeutic that is preferable to masitinib.
Collapse
Affiliation(s)
- Olanrewaju Ayodeji Durojaye
- MOE Key Laboratory of Membraneless Organelle and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230027, Anhui, China
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Department of Chemical Sciences, Coal City University, Emene, Enugu State, Nigeria
| | - Nkwachukwu Oziamara Okoro
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, 410001, Nigeria
| | - Arome Solomon Odiba
- Department of Molecular Genetics and Biotechnology, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria.
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria.
| | - Bennett Chima Nwanguma
- Department of Molecular Genetics and Biotechnology, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria.
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria.
| |
Collapse
|
9
|
Yehia N, Salem HM, Mahmmod Y, Said D, Samir M, Mawgod SA, Sorour HK, AbdelRahman MAA, Selim S, Saad AM, El-Saadony MT, El-Meihy RM, Abd El-Hack ME, El-Tarabily KA, Zanaty AM. Common viral and bacterial avian respiratory infections: an updated review. Poult Sci 2023; 102:102553. [PMID: 36965253 PMCID: PMC10064437 DOI: 10.1016/j.psj.2023.102553] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Many pathogens that cause chronic diseases in birds use the respiratory tract as a primary route of infection, and respiratory disorders are the main leading source of financial losses in the poultry business. Respiratory infections are a serious problem facing the poultry sector, causing severe economic losses. Avian influenza virus, Newcastle disease virus, infectious bronchitis virus, and avian pneumovirus are particularly serious viral respiratory pathogens. Mycoplasma gallisepticum, Staphylococcus, Bordetella avium, Pasteurella multocida, Riemerella anatipestifer, Chlamydophila psittaci, and Escherichia coli have been identified as the most serious bacterial respiratory pathogens in poultry. This review gives an updated summary, incorporating the latest data, about the evidence for the circulation of widespread, economically important poultry respiratory pathogens, with special reference to possible methods for the control and prevention of these pathogens.
Collapse
Affiliation(s)
- Nahed Yehia
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center, Giza 12618, Egypt
| | - Heba M Salem
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Yasser Mahmmod
- Department of Veterinary Sciences, Faculty of Health Sciences, Higher Colleges of Technology, Al Ain 17155, United Arab Emirates
| | - Dalia Said
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center, Giza 12618, Egypt
| | - Mahmoud Samir
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center, Giza 12618, Egypt
| | - Sara Abdel Mawgod
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center, Giza 12618, Egypt
| | - Hend K Sorour
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center, Giza 12618, Egypt
| | - Mona A A AbdelRahman
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center, Giza 12618, Egypt
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Ahmed M Saad
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Rasha M El-Meihy
- Department of Agricultural Microbiology, Faculty of Agriculture, Benha University, Moshtohor, Qaluybia 13736, Egypt
| | - Mohamed E Abd El-Hack
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates; Khalifa Center for Genetic Engineering and Biotechnology, United Arab Emirates University, Al Ain 15551, United Arab Emirates; Harry Butler Institute, Murdoch University, Murdoch 6150, Western Australia, Australia.
| | - Ali M Zanaty
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agriculture Research Center, Giza 12618, Egypt
| |
Collapse
|
10
|
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: 27] [Impact Index Per Article: 9.0] [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
|
11
|
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: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
12
|
Ozkocak DC, Phan TK, Poon IKH. Translating extracellular vesicle packaging into therapeutic applications. Front Immunol 2022; 13:946422. [PMID: 36045692 PMCID: PMC9420853 DOI: 10.3389/fimmu.2022.946422] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound particles released by cells in various (patho)physiological conditions. EVs can transfer effector molecules and elicit potent responses in recipient cells, making them attractive therapeutic agents and drug delivery platforms. In contrast to their tremendous potential, only a few EV-based therapies and drug delivery have been approved for clinical use, which is largely attributed to limited therapeutic loading technologies and efficiency. As EV cargo has major influence on their functionality, understanding and translating the biology underlying the packaging and transferring of biomolecule cargos (e.g. miRNAs, pathogen antigens, small molecule drugs) into EVs is key in harnessing their therapeutic potential. In this review, through recent insights into EVs’ content packaging, we discuss different mechanisms utilized by EVs during cargo packaging, and how one might therapeutically exploit this process. Apart from the well-characterized EVs like exosomes and microvesicles, we also cover the less-studied and other EV subtypes like apoptotic bodies, large oncosomes, bacterial outer membrane vesicles, and migrasomes to highlight therapeutically-diverse opportunities of EV armoury.
Collapse
|
13
|
Mat Rani NNI, Alzubaidi ZM, Butt AM, Mohammad Faizal NDF, Sekar M, Azhari H, Mohd Amin MCI. Outer membrane vesicles as biomimetic vaccine carriers against infections and cancers. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1784. [PMID: 35194964 DOI: 10.1002/wnan.1784] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/18/2022] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
In the last decade, nanoparticle-based therapeutic modalities have emerged as promising treatment options for cancer and infectious diseases. To improve prognosis, chemotherapeutic and antimicrobial drugs must be delivered selectively to the target sites. Researchers have increasingly focused their efforts on improving drug delivery, with a particular emphasis on cancer and infectious diseases. When drugs are administered systemically, they become diluted and can diffuse to all tissues but only until the immune system intervenes and quickly removes them from circulation. To enhance and prolong the systemic circulation of drugs, nanocarriers have been explored and used; however, nanocarriers have a major drawback in that they can trigger immune responses. Numerous nanocarriers for optimal drug delivery have been developed using innovative and effective biointerface technologies. Autologous cell-derived drug carriers, such as outer membrane vesicles (OMVs), have demonstrated improved bioavailability and reduced toxicity. Thus, this study investigates the use of biomimetic OMVs as biomimetic vaccine carriers against infections and cancers to improve our understanding in the field of nanotechnology. In addition, discussion on the advantages, disadvantages, and future prospects of OMVs will also be explored. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Nur Najihah Izzati Mat Rani
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh, Perak, Malaysia
| | - Zahraa M Alzubaidi
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
| | - Adeel Masood Butt
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Nur Dini Fatini Mohammad Faizal
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh, Perak, Malaysia
| | - Hanisah Azhari
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, Malaysia
| |
Collapse
|
14
|
Yang Z, Hua L, Yang M, Li W, Ren Z, Zheng X, Chen H, Long Q, Bai H, Huang W, Ma Y. Polymerized porin as a novel delivery platform for coronavirus vaccine. J Nanobiotechnology 2022; 20:260. [PMID: 35672856 PMCID: PMC9171476 DOI: 10.1186/s12951-022-01469-8] [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: 11/30/2021] [Accepted: 05/20/2022] [Indexed: 11/10/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), seriously threatens human life and health. The correct folding and polymerization of the receptor-binding domain (RBD) protein of coronavirus in Escherichia coli may reduce the cost of SARS-CoV-2 vaccines. In this study, we constructed this nanopore by using the principle of ClyA porin polymerization triggered by the cell membrane. We used surfactants to "pick" the ClyA-RBD nanopore from the bacterial outer membrane. More importantly, the polymerized RBD displayed on the ClyA-RBD polymerized porin (RBD-PP) already displays some correct spatial conformational epitopes that can induce neutralizing antibodies. The nanostructures of RBD-PP can target lymph nodes and promote antigen uptake and processing by dendritic cells, thereby effectively eliciting the production of anti-SARS-CoV-2 neutralizing antibodies, systemic cellular immune responses, and memory T cells. We applied this PP-based vaccine platform to fabricate an RBD-based subunit vaccine against SARS-CoV-2, which will provide a foundation for the development of inexpensive coronavirus vaccines. The development of a novel vaccine delivery system is an important part of innovative drug research. This novel PP-based vaccine platform is likely to have additional applications, including other viral vaccines, bacterial vaccines, tumor vaccines, drug delivery, and disease diagnosis.
Collapse
Affiliation(s)
- Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Liangqun Hua
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
- Yunnan University, Kunming, 650091, China
| | - Mengli Yang
- National Kunming High-Level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Zhaoling Ren
- The Second Affiliated Hospital of Kunming Medical University, Kunming, 650033, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
- Yunnan University, Kunming, 650091, China
| | - Haoqian Chen
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
- Yunnan Minzu University, Kunming, 650504, China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Hongmei Bai
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China.
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650031, China.
| |
Collapse
|
15
|
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
|
16
|
Osterloh A. Vaccination against Bacterial Infections: Challenges, Progress, and New Approaches with a Focus on Intracellular Bacteria. Vaccines (Basel) 2022; 10:751. [PMID: 35632507 PMCID: PMC9144739 DOI: 10.3390/vaccines10050751] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Many bacterial infections are major health problems worldwide, and treatment of many of these infectious diseases is becoming increasingly difficult due to the development of antibiotic resistance, which is a major threat. Prophylactic vaccines against these bacterial pathogens are urgently needed. This is also true for bacterial infections that are still neglected, even though they affect a large part of the world's population, especially under poor hygienic conditions. One example is typhus, a life-threatening disease also known as "war plague" caused by Rickettsia prowazekii, which could potentially come back in a war situation such as the one in Ukraine. However, vaccination against bacterial infections is a challenge. In general, bacteria are much more complex organisms than viruses and as such are more difficult targets. Unlike comparatively simple viruses, bacteria possess a variety of antigens whose immunogenic potential is often unknown, and it is unclear which antigen can elicit a protective and long-lasting immune response. Several vaccines against extracellular bacteria have been developed in the past and are still used successfully today, e.g., vaccines against tetanus, pertussis, and diphtheria. However, while induction of antibody production is usually sufficient for protection against extracellular bacteria, vaccination against intracellular bacteria is much more difficult because effective defense against these pathogens requires T cell-mediated responses, particularly the activation of cytotoxic CD8+ T cells. These responses are usually not efficiently elicited by immunization with non-living whole cell antigens or subunit vaccines, so that other antigen delivery strategies are required. This review provides an overview of existing antibacterial vaccines and novel approaches to vaccination with a focus on immunization against intracellular bacteria.
Collapse
Affiliation(s)
- Anke Osterloh
- Department of Infection Immunology, Research Center Borstel, Parkallee 22, 23845 Borstel, Germany
| |
Collapse
|
17
|
The tremendous biomedical potential of bacterial extracellular vesicles. Trends Biotechnol 2022; 40:1173-1194. [DOI: 10.1016/j.tibtech.2022.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 12/20/2022]
|
18
|
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
|
19
|
Domínguez Rubio AP, D’Antoni CL, Piuri M, Pérez OE. Probiotics, Their Extracellular Vesicles and Infectious Diseases. Front Microbiol 2022; 13:864720. [PMID: 35432276 PMCID: PMC9006447 DOI: 10.3389/fmicb.2022.864720] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Probiotics have been shown to be effective against infectious diseases in clinical trials, with either intestinal or extraintestinal health benefits. Even though probiotic effects are strain-specific, some "widespread effects" include: pathogen inhibition, enhancement of barrier integrity and regulation of immune responses. The mechanisms involved in the health benefits of probiotics are not completely understood, but these effects can be mediated, at least in part, by probiotic-derived extracellular vesicles (EVs). However, to date, there are no clinical trials examining probiotic-derived EVs health benefits against infectious diseases. There is still a long way to go to bridge the gap between basic research and clinical practice. This review attempts to summarize the current knowledge about EVs released by probiotic bacteria to understand their possible role in the prevention and/or treatment of infectious diseases. A better understanding of the mechanisms whereby EVs package their cargo and the process involved in communication with host cells (inter-kingdom communication), would allow further advances in this field. In addition, we comment on the potential use and missing knowledge of EVs as therapeutic agents (postbiotics) against infectious diseases. Future research on probiotic-derived EVs is needed to open new avenues for the encapsulation of bioactives inside EVs from GRAS (Generally Regarded as Safe) bacteria. This could be a scientific novelty with applications in functional foods and pharmaceutical industries.
Collapse
Affiliation(s)
- A. Paula Domínguez Rubio
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Cecilia L. D’Antoni
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Mariana Piuri
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Oscar E. Pérez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
20
|
Huang W, Meng L, Chen Y, Dong Z, Peng Q. Bacterial outer membrane vesicles as potential biological nanomaterials for antibacterial therapy. Acta Biomater 2022; 140:102-115. [PMID: 34896632 DOI: 10.1016/j.actbio.2021.12.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/05/2021] [Accepted: 12/03/2021] [Indexed: 02/05/2023]
Abstract
Antibiotic therapy is one of the most important approaches against bacterial infections. However, the improper use of antibiotics and the emergence of drug resistance have compromised the efficacy of traditional antibiotic therapy. In this regard, it is of great importance and significance to develop more potent antimicrobial therapies, including the development of functionalized antibiotics delivery systems and antibiotics-independent antimicrobial agents. Outer membrane vesicles (OMVs), secreted by Gram-negative bacteria and with similar structure to cell-derived exosomes, are natural functional nanomaterials and known to play important roles in many bacterial life events, such as communication, biofilm formation and pathogenesis. Recently, more and more reports have demonstrated the use of OMVs as either active antibacterial agents or antibiotics delivery carriers, implying the great potentials of OMVs in antibacterial therapy. Herein, we aim to provide a comprehensive understanding of OMV and its antibacterial applications, including its biogenesis, biofunctions, isolation, purification and its potentials in killing bacteria, delivering antibiotics and developing vaccine or immunoadjuvants. In addition, the concerns in clinical use of OMVs and the possible solutions are discussed. STATEMENT OF SIGNIFICANCE: The emergence of antibiotic-resistant bacteria has led to the failure of traditional antibiotic therapy, and thus become a big threat to human beings. In this regard, developing more potent antibacterial approaches is of great importance and significance. Recently, bacterial outer membrane vesicles (OMVs), which are natural functional nanomaterials secreted by Gram-negative bacteria, have been used as active agents, drug carriers and vaccine adjuvant for antibacterial therapy. This review provides a comprehensive understanding of OMVs and summarizes the recent progress of OMVs in antibacterial applications. The concerns of OMVs in clinical use and the possible solutions are also discussed. As such, this review may guide the future works in antibacterial OMVs and appeal to both scientists and clinicians.
Collapse
Affiliation(s)
- Wenlong Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Lingxi Meng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuan Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zaiquan Dong
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
21
|
Iyer S, Yadav R, Agarwal S, Tripathi S, Agarwal R. Bioengineering Strategies for Developing Vaccines against Respiratory Viral Diseases. Clin Microbiol Rev 2022; 35:e0012321. [PMID: 34788128 PMCID: PMC8597982 DOI: 10.1128/cmr.00123-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Respiratory viral pathogens like influenza and coronaviruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have caused outbreaks leading to millions of deaths. Vaccinations are, to date, the best and most economical way to control such outbreaks and have been highly successful for several pathogens. Currently used vaccines for respiratory viral pathogens are primarily live attenuated or inactivated and can risk reversion to virulence or confer inadequate immunity. The recent trend of using potent biomolecules like DNA, RNA, and protein antigenic components to synthesize vaccines for diseases has shown promising results. Still, it remains challenging to translate due to their high susceptibility to degradation during storage and after delivery. Advances in bioengineering technology for vaccine design have made it possible to control the physicochemical properties of the vaccines for rapid synthesis, heightened antigen presentation, safer formulations, and more robust immunogenicity. Bioengineering techniques and materials have been used to synthesize several potent vaccines, approved or in trials, against coronavirus disease 2019 (COVID-19) and are being explored for influenza, SARS, and Middle East respiratory syndrome (MERS) vaccines as well. Here, we review bioengineering strategies such as the use of polymeric particles, liposomes, and virus-like particles in vaccine development against influenza and coronaviruses and the feasibility of adopting these technologies for clinical use.
Collapse
Affiliation(s)
- Shalini Iyer
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Rajesh Yadav
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Smriti Agarwal
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Shashank Tripathi
- Department of Microbiology and Cell Biology, Center for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
| | - Rachit Agarwal
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
22
|
Debnath SK, Srivastava R. Potential Application of Bionanoparticles to Treat Severe Acute Respiratory Syndrome Coronavirus-2 Infection. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2021.813847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is a contagious virus that spreads exponentially across the world, resulting in serious viral pneumonia. Several companies and researchers have put their tremendous effort into developing novel vaccines or drugs for the complete eradication of COVID-19 caused by SARS-CoV-2. Bionanotechnology plays a vital role in designing functionalized biocompatible nanoparticulate systems with higher antiviral capabilities. Thus, several nanocarriers have been explored in designing and delivering drugs and vaccines. This problem can be overcome with the intervention of biomaterials or bionanoparticles. The present review describes the comparative analysis of SARS infection and its associated etiological agents. This review also highlighted some nanoparticles that have been explored in the treatment of COVID-19. However, these carriers elicit several problems once they come in contact with biological systems. Often, the body’s immune system treats these nanocarriers as foreign particles and antigens. In contrast, some bionanoparticles are highlighted here with their potential application in SARS-CoV-2. However, bionanoparticles have demonstrated some drawbacks discussed here with the possible outcomes. The scope of bioinspired nanoparticles is also discussed in detail to explore the new era of research. It is highly essential for the effective delivery of these nanoparticles to the target site. For effective management of SARS-CoV-2, different delivery patterns are also discussed here.
Collapse
|
23
|
Al-Karmalawy AA, Soltane R, Abo Elmaaty A, Tantawy MA, Antar SA, Yahya G, Chrouda A, Pashameah RA, Mustafa M, Abu Mraheil M, Mostafa A. Coronavirus Disease (COVID-19) Control between Drug Repurposing and Vaccination: A Comprehensive Overview. Vaccines (Basel) 2021; 9:1317. [PMID: 34835248 PMCID: PMC8622998 DOI: 10.3390/vaccines9111317] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023] Open
Abstract
Respiratory viruses represent a major public health concern, as they are highly mutated, resulting in new strains emerging with high pathogenicity. Currently, the world is suffering from the newly evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This virus is the cause of coronavirus disease 2019 (COVID-19), a mild-to-severe respiratory tract infection with frequent ability to give rise to fatal pneumonia in humans. The overwhelming outbreak of SARS-CoV-2 continues to unfold all over the world, urging scientists to put an end to this global pandemic through biological and pharmaceutical interventions. Currently, there is no specific treatment option that is capable of COVID-19 pandemic eradication, so several repurposed drugs and newly conditionally approved vaccines are in use and heavily applied to control the COVID-19 pandemic. The emergence of new variants of the virus that partially or totally escape from the immune response elicited by the approved vaccines requires continuous monitoring of the emerging variants to update the content of the developed vaccines or modify them totally to match the new variants. Herein, we discuss the potential therapeutic and prophylactic interventions including repurposed drugs and the newly developed/approved vaccines, highlighting the impact of virus evolution on the immune evasion of the virus from currently licensed vaccines for COVID-19.
Collapse
Affiliation(s)
- Ahmed A. Al-Karmalawy
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Raya Soltane
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (R.S.); (R.A.P.)
- Department of Biology, Faculty of Sciences, Tunis El Manar University, Tunis 1068, Tunisia
| | - Ayman Abo Elmaaty
- Department of Medicinal Chemistry, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt;
| | - Mohamed A. Tantawy
- Hormones Department, Medical Research and Clinical Studies Research Institute, National Research Centre, Dokki 12622, Egypt;
- Stem Cells Laboratory, Center of Excellence for Advanced Sciences, National Research Centre, Dokki 12622, Egypt
| | - Samar A. Antar
- Department of Pharmacology, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt;
| | - Galal Yahya
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Amani Chrouda
- Department of Chemistry, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11932, Saudi Arabia;
- Laboratory of Interfaces and Advanced Materials, Faculty of Sciences, Monastir University, Monastir 5000, Tunisia
- Institute of Analytical Sciences, UMR CNRS-UCBL-ENS 5280, 5 Rue la Doua, CEDEX, 69100 Villeurbanne, France
| | - Rami Adel Pashameah
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (R.S.); (R.A.P.)
| | - Muhamad Mustafa
- Department of Medicinal Chemistry, Deraya University, Minia 61111, Egypt;
| | - Mobarak Abu Mraheil
- German Center for Infection Research (DZIF), Institute of Medical Microbiology, Justus-Liebig University, 35392 Giessen, Germany;
| | - Ahmed Mostafa
- German Center for Infection Research (DZIF), Institute of Medical Microbiology, Justus-Liebig University, 35392 Giessen, Germany;
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Dokki 12622, Egypt
| |
Collapse
|
24
|
Claridge B, Lozano J, Poh QH, Greening DW. Development of Extracellular Vesicle Therapeutics: Challenges, Considerations, and Opportunities. Front Cell Dev Biol 2021; 9:734720. [PMID: 34616741 PMCID: PMC8488228 DOI: 10.3389/fcell.2021.734720] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) hold great promise as therapeutic modalities due to their endogenous characteristics, however, further bioengineering refinement is required to address clinical and commercial limitations. Clinical applications of EV-based therapeutics are being trialed in immunomodulation, tissue regeneration and recovery, and as delivery vectors for combination therapies. Native/biological EVs possess diverse endogenous properties that offer stability and facilitate crossing of biological barriers for delivery of molecular cargo to cells, acting as a form of intercellular communication to regulate function and phenotype. Moreover, EVs are important components of paracrine signaling in stem/progenitor cell-based therapies, are employed as standalone therapies, and can be used as a drug delivery system. Despite remarkable utility of native/biological EVs, they can be improved using bio/engineering approaches to further therapeutic potential. EVs can be engineered to harbor specific pharmaceutical content, enhance their stability, and modify surface epitopes for improved tropism and targeting to cells and tissues in vivo. Limitations currently challenging the full realization of their therapeutic utility include scalability and standardization of generation, molecular characterization for design and regulation, therapeutic potency assessment, and targeted delivery. The fields' utilization of advanced technologies (imaging, quantitative analyses, multi-omics, labeling/live-cell reporters), and utility of biocompatible natural sources for producing EVs (plants, bacteria, milk) will play an important role in overcoming these limitations. Advancements in EV engineering methodologies and design will facilitate the development of EV-based therapeutics, revolutionizing the current pharmaceutical landscape.
Collapse
Affiliation(s)
- Bethany Claridge
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jonathan Lozano
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Qi Hui Poh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - David W. Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Zhao Y, Li X, Zhang W, Yu L, Wang Y, Deng Z, Liu M, Mo S, Wang R, Zhao J, Liu S, Hao Y, Wang X, Ji T, Zhang L, Wang C. Trends in the biological functions and medical applications of extracellular vesicles and analogues. Acta Pharm Sin B 2021; 11:2114-2135. [PMID: 34522580 PMCID: PMC8424226 DOI: 10.1016/j.apsb.2021.03.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/19/2021] [Accepted: 03/01/2021] [Indexed: 12/18/2022] Open
Abstract
Natural extracellular vesicles (EVs) play important roles in many life processes such as in the intermolecular transfer of substances and genetic information exchanges. Investigating the origins and working mechanisms of natural EVs may provide an understanding of life activities, especially regarding the occurrence and development of diseases. Additionally, due to their vesicular structure, EVs (in small molecules, nucleic acids, proteins, etc.) could act as efficient drug-delivery carriers. Herein, we describe the sources and biological functions of various EVs, summarize the roles of EVs in disease diagnosis and treatment, and review the application of EVs as drug-delivery carriers. We also assess the challenges and perspectives of EVs in biomedical applications.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Xiaolu Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Wenbo Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Lanlan Yu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yang Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Zhun Deng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Mingwei Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Shanshan Mo
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Ruonan Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Jinming Zhao
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
| | - Shuli Liu
- Department of Clinical Laboratory, Peking University Civil Aviation School of Clinical Medicine, Beijing 100123, China
| | - Yun Hao
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
| | - Xiangdong Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
| | - Tianjiao Ji
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Chenxuan Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
26
|
Yang Z, Hua L, Yang M, Liu SQ, Shen J, Li W, Long Q, Bai H, Yang X, Ren Z, Zheng X, Sun W, Ye C, Li D, Zheng P, He J, Chen Y, Huang W, Peng X, Ma Y. RBD-Modified Bacterial Vesicles Elicited Potential Protective Immunity against SARS-CoV-2. NANO LETTERS 2021; 21:5920-5930. [PMID: 34279108 PMCID: PMC8315139 DOI: 10.1021/acs.nanolett.1c00680] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/02/2021] [Indexed: 05/13/2023]
Abstract
The disease caused by SARS-CoV-2 infection threatens human health. In this study, we used high-pressure homogenization technology not only to efficiently drive the bacterial membrane to produce artificial vesicles but also to force the fusion protein ClyA-receptor binding domain (RBD) to pass through gaps in the bacterial membrane to increase the contact between ClyA-RBD and the membrane. Therefore, the load of ClyA-RBD on the membrane is substantially increased. Using this technology, we constructed a "ring-like" bacterial biomimetic vesicle (BBV) loaded with polymerized RBD (RBD-BBV). RBD-BBVs injected subcutaneously can accumulate in lymph nodes, promote antigen uptake and processing, and elicit SARS-CoV-2-specific humoral and cellular immune responses in mice. In conclusion, we evaluated the potential of this novel bacterial vesicle as a vaccine delivery system and provided a new idea for the development of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Zhongqian Yang
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Liangqun Hua
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
- Yunnan
University, Kunming, China
| | - Mengli Yang
- National
Kunming High-level Biosafety Primate Research Center, Institute of
Medical Biology, Chinese Academy of Medical
Sciences and Peking Union Medical College, Kunming, China
| | - Shu-Qun Liu
- Yunnan
University, Kunming, China
- State
Key Laboratory for Conservation and Utilization of Bio-Resources in
Yunnan & School of Life Sciences, Yunnan
University, Kunming, China
| | - Jianxin Shen
- Yunnan
University, Kunming, China
- State
Key Laboratory for Conservation and Utilization of Bio-Resources in
Yunnan & School of Life Sciences, Yunnan
University, Kunming, China
| | - Weiran Li
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Qiong Long
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Hongmei Bai
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Xu Yang
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Zhaoling Ren
- The
Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Kunming
Medical University, Kunming, China
| | - Xiao Zheng
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
- Yunnan
University, Kunming, China
| | - Wenjia Sun
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Chao Ye
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Duo Li
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
- Department
of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Center for Disease Control and Prevention, Kunming, China
| | - Peng Zheng
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Jinrong He
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
- Kunming
Medical University, Kunming, China
| | - Yongjun Chen
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Weiwei Huang
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| | - Xiaozhong Peng
- National
Kunming High-level Biosafety Primate Research Center, Institute of
Medical Biology, Chinese Academy of Medical
Sciences and Peking Union Medical College, Kunming, China
- State
Key Laboratory of Medical Molecular Biology, Department of Molecular
Biology and Biochemistry, Institute of Basic Medical Sciences, Medical
Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking
Union Medical College, Beijing, China
| | - Yanbing Ma
- Laboratory
of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union
Medical College, Kunming, China
| |
Collapse
|
27
|
Roy R, You RI, Chang CH, Yang CY, Lin NT. Carboxy-Terminal Processing Protease Controls Production of Outer Membrane Vesicles and Biofilm in Acinetobacter baumannii. Microorganisms 2021; 9:microorganisms9061336. [PMID: 34203028 PMCID: PMC8234194 DOI: 10.3390/microorganisms9061336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Carboxy-terminal processing protease (Ctp) is a serine protease that controls multiple cellular processes through posttranslational modification of proteins. Acinetobacter baumannii ATCC 17978 ctp mutant, namely MR14, is known to cause cell wall defects and autolysis. The objective of this study was to investigate the role of ctp mutation-driven autolysis in regulating biofilms in A. baumannii and to evaluate the vesiculation caused by cell wall defects. We found that in A. baumannii, Ctp is localized in the cytoplasmic membrane, and loss of Ctp function enhances the biofilm-forming ability of A. baumannii. Quantification of the matrix components revealed that extracellular DNA (eDNA) and proteins were the chief constituents of MR14 biofilm, and the transmission electron microscopy further indicated the presence of numerous dead cells compared with ATCC 17978. The large number of MR14 dead cells is potentially the result of compromised outer membrane integrity, as demonstrated by its high sensitivity to sodium dodecyl sulfate (SDS) and ethylenediaminetetraacetic acid (EDTA). MR14 also exhibited the hypervesiculation phenotype, producing outer-membrane vesicles (OMVs) of large mean size. The MR14 OMVs were more cytotoxic toward A549 cells than ATCC 17978 OMVs. Our overall results indicate that A. baumanniictp negatively controls pathogenic traits through autolysis and OMV biogenesis.
Collapse
Affiliation(s)
- Rakesh Roy
- Institute of Medical Sciences, Tzu Chi University, No. 701, Sec. 3, Zhongyang Rd., Hualien 97004, Taiwan;
| | - Ren-In You
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, No. 701, Sec. 3, Zhongyang Rd., Hualien 97004, Taiwan;
| | - Chan-Hua Chang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 40227, Taiwan;
| | - Chiou-Ying Yang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 40227, Taiwan;
- Correspondence: (C.-Y.Y.); (N.-T.L.); Tel.: +886-3-856 5301 (ext. 2080) (N.-T.L.); Fax: +886-3-8566724 (N.-T.L.)
| | - Nien-Tsung Lin
- Institute of Medical Sciences, Tzu Chi University, No. 701, Sec. 3, Zhongyang Rd., Hualien 97004, Taiwan;
- Department of Microbiology, School of Medicine, Tzu Chi University, No. 701, Sec. 3, Zhongyang Rd., Hualien 97004, Taiwan
- Correspondence: (C.-Y.Y.); (N.-T.L.); Tel.: +886-3-856 5301 (ext. 2080) (N.-T.L.); Fax: +886-3-8566724 (N.-T.L.)
| |
Collapse
|
28
|
Zingl FG, Leitner DR, Thapa HB, Schild S. Outer membrane vesicles as versatile tools for therapeutic approaches. MICROLIFE 2021; 2:uqab006. [PMID: 37223254 PMCID: PMC10117751 DOI: 10.1093/femsml/uqab006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/05/2021] [Indexed: 05/25/2023]
Abstract
Budding of the bacterial surface results in the formation and secretion of outer membrane vesicles, which is a conserved phenomenon observed in Gram-negative bacteria. Recent studies highlight that these sphere-shaped facsimiles of the donor bacterium's surface with enclosed periplasmic content may serve multiple purposes for their host bacterium. These include inter- and intraspecies cell-cell communication, effector delivery to target cells and bacterial adaptation strategies. This review provides a concise overview of potential medical applications to exploit outer membrane vesicles for therapeutic approaches. Due to the fact that outer membrane vesicles resemble the surface of their donor cells, they represent interesting nonliving candidates for vaccine development. Furthermore, bacterial donor species can be genetically engineered to display various proteins and glycans of interest on the outer membrane vesicle surface or in their lumen. Outer membrane vesicles also possess valuable bioreactor features as they have the natural capacity to protect, stabilize and enhance the activity of luminal enzymes. Along these features, outer membrane vesicles not only might be suitable for biotechnological applications but may also enable cell-specific delivery of designed therapeutics as they are efficiently internalized by nonprofessional phagocytes. Finally, outer membrane vesicles are potent modulators of our immune system with pro- and anti-inflammatory properties. A deeper understanding of immunoregulatory effects provoked by different outer membrane vesicles is the basis for their possible future applications ranging from inflammation and immune response modulation to anticancer therapy.
Collapse
Affiliation(s)
- Franz G Zingl
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Deborah R Leitner
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Himadri B Thapa
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Stefan Schild
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
- BioTechMed-Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| |
Collapse
|
29
|
Root-Bernstein R. Innate Receptor Activation Patterns Involving TLR and NLR Synergisms in COVID-19, ALI/ARDS and Sepsis Cytokine Storms: A Review and Model Making Novel Predictions and Therapeutic Suggestions. Int J Mol Sci 2021; 22:ijms22042108. [PMID: 33672738 PMCID: PMC7924650 DOI: 10.3390/ijms22042108] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 01/08/2023] Open
Abstract
Severe COVID-19 is characterized by a “cytokine storm”, the mechanism of which is not yet understood. I propose that cytokine storms result from synergistic interactions among Toll-like receptors (TLR) and nucleotide-binding oligomerization domain-like receptors (NLR) due to combined infections of SARS-CoV-2 with other microbes, mainly bacterial and fungal. This proposition is based on eight linked types of evidence and their logical connections. (1) Severe cases of COVID-19 differ from healthy controls and mild COVID-19 patients in exhibiting increased TLR4, TLR7, TLR9 and NLRP3 activity. (2) SARS-CoV-2 and related coronaviruses activate TLR3, TLR7, RIG1 and NLRP3. (3) SARS-CoV-2 cannot, therefore, account for the innate receptor activation pattern (IRAP) found in severe COVID-19 patients. (4) Severe COVID-19 also differs from its mild form in being characterized by bacterial and fungal infections. (5) Respiratory bacterial and fungal infections activate TLR2, TLR4, TLR9 and NLRP3. (6) A combination of SARS-CoV-2 with bacterial/fungal coinfections accounts for the IRAP found in severe COVID-19 and why it differs from mild cases. (7) Notably, TLR7 (viral) and TLR4 (bacterial/fungal) synergize, TLR9 and TLR4 (both bacterial/fungal) synergize and TLR2 and TLR4 (both bacterial/fungal) synergize with NLRP3 (viral and bacterial). (8) Thus, a SARS-CoV-2-bacterium/fungus coinfection produces synergistic innate activation, resulting in the hyperinflammation characteristic of a cytokine storm. Unique clinical, experimental and therapeutic predictions (such as why melatonin is effective in treating COVID-19) are discussed, and broader implications are outlined for understanding why other syndromes such as acute lung injury, acute respiratory distress syndrome and sepsis display varied cytokine storm symptoms.
Collapse
|
30
|
Sabanovic B, Piva F, Cecati M, Giulietti M. Promising Extracellular Vesicle-Based Vaccines against Viruses, Including SARS-CoV-2. BIOLOGY 2021; 10:94. [PMID: 33513850 PMCID: PMC7912280 DOI: 10.3390/biology10020094] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/07/2021] [Accepted: 01/24/2021] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) are secreted from almost all human cells and mediate intercellular communication by transferring heterogeneous molecules (i.e., DNA, RNAs, proteins, and lipids). In this way, EVs participate in various biological processes, including immune responses. Viruses can hijack EV biogenesis systems for their dissemination, while EVs from infected cells can transfer viral proteins to uninfected cells and to immune cells in order to mask the infection or to trigger a response. Several studies have highlighted the role of native or engineered EVs in the induction of B cell and CD8(+) T cell reactions against viral proteins, strongly suggesting these antigen-presenting EVs as a novel strategy for vaccine design, including the emerging COVID-19. EV-based vaccines overcome some limitations of conventional vaccines and introduce novel unique characteristics useful in vaccine design, including higher bio-safety and efficiency as antigen-presenting systems and as adjuvants. Here, we review the state-of-the-art for antiviral EV-based vaccines, including the ongoing projects of some biotech companies in the development of EV-based vaccines for SARS-CoV-2. Finally, we discuss the limits for further development of this promising class of therapeutic agents.
Collapse
Affiliation(s)
| | | | | | - Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (B.S.); (F.P.); (M.C.)
| |
Collapse
|
31
|
The Use of Nanobiotechnology in Immunology and Vaccination. Vaccines (Basel) 2021; 9:vaccines9020074. [PMID: 33494441 PMCID: PMC7910821 DOI: 10.3390/vaccines9020074] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 01/07/2023] Open
Abstract
Nanotechnology uses the unique properties of nanostructures with a size of 1 to 200 nanometers. Different nanoparticles have shown great promise for the production of new vaccines and drugs. Nanostructures can be used to deliver immunological compounds more effectively than microstructures to target sites. Different nanostructures can be applied to form a new generation of vaccines, adjuvants, and immune system drugs. The goal of nanotechnology is to better respond to a wide range of infectious and non-infectious diseases.
Collapse
|
32
|
Gao J, Wang S, Dong X, Wang Z. RGD-expressed bacterial membrane-derived nanovesicles enhance cancer therapy via multiple tumorous targeting. Theranostics 2021; 11:3301-3316. [PMID: 33537088 PMCID: PMC7847689 DOI: 10.7150/thno.51988] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background: A tumor microenvironment is a complicated multicellular system comprised of tumor cells, immune cells and blood vessels. Blood vessels are the barriers for drug tissue penetration. Effectively treating a cancer requires drug delivery systems to overcome biological barriers present in tumor microenvironments (TMEs). Methods: We designed a drug delivery system made of bacterial (Escherichia coli) double layer membrane-derived nanovesicles (DMVs) with the expression of RGD peptides and endogenous targeting ligands of bacteria. The physical and biological characteristics of DMVs were assessed by cryogenic transmission electron microscopy, western blotting, flow cytometry and confocal microscopy. Doxorubicin (DOX) was loaded in DMVs via a pH gradient driven drug loading method. Therapeutical effects of DOX-loaded DMVs were studied in a melanoma xenograft mouse model. Results:In vitro and in vivo experiments showed that DMVs can target neutrophils and monocytes that mediated the transport of DMVs across blood vessel barriers and they can also directly target tumor vasculature and tumor cells, resulting in enhanced delivery of therapeutics to TMEs. Furthermore, we developed a remote drug loading approach to efficiently encapsulate DOX inside DMVs, and the drug loading was 12% (w/w). In the B16-F10 melanoma mouse model, we showed that DOX-RGD-DMVs significantly inhibited the tumor growth compared to several controls. Conclusion: Our studies reveal that DMVs are a powerful tool to simultaneously target multiple cells in TMEs, thus increasing drug delivery for improved cancer therapies.
Collapse
|
33
|
Loo YS, Bose RJ, McCarthy JR, Mat Azmi ID, Madheswaran T. Biomimetic bacterial and viral-based nanovesicles for drug delivery, theranostics, and vaccine applications. Drug Discov Today 2020; 26:902-915. [PMID: 33383213 DOI: 10.1016/j.drudis.2020.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/16/2020] [Accepted: 12/21/2020] [Indexed: 01/04/2023]
Abstract
Smart nanocarriers obtained from bacteria and viruses offer excellent biomimetic properties which has led to significant research into the creation of advanced biomimetic materials. Their versatile biomimicry has application as biosensors, biomedical scaffolds, immobilization, diagnostics, and targeted or personalized treatments. The inherent natural traits of biomimetic and bioinspired bacteria- and virus-derived nanovesicles show potential for their use in clinical vaccines and novel therapeutic drug delivery systems. The past few decades have seen significant progress in the bioengineering of bacteria and viruses to manipulate and enhance their therapeutic benefits. From a pharmaceutical perspective, biomimetics enable the safe integration of naturally occurring bacteria and virus particles to achieve high, stable rates of cellular transfection/infection and prolonged circulation times. In addition, biomimetic technologies can overcome safety concerns associated with live-attenuated and inactivated whole bacteria or viruses. In this review, we provide an update on the utilization of bacterial and viral particles as drug delivery systems, theranostic carriers, and vaccine/immunomodulation modalities.
Collapse
Affiliation(s)
- Yan Shan Loo
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia
| | - Rajendran Jc Bose
- Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY 13501, USA
| | - Jason R McCarthy
- Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY 13501, USA
| | - Intan Diana Mat Azmi
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia.
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, International Medical University, No. 126 Jalan Jalil Perkasa 19, Bukit Jalil, 57000 Kuala Lumpur, Malaysia.
| |
Collapse
|
34
|
Oroojalian F, Haghbin A, Baradaran B, Hemmat N, Shahbazi MA, Baghi HB, Mokhtarzadeh A, Hamblin MR. Novel insights into the treatment of SARS-CoV-2 infection: An overview of current clinical trials. Int J Biol Macromol 2020; 165:18-43. [PMID: 32991900 PMCID: PMC7521454 DOI: 10.1016/j.ijbiomac.2020.09.204] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
Abstract
The emergence of the global pandemic caused by the novel SARS-CoV-2 virus has motivated scientists to find a definitive treatment or a vaccine against it in the shortest possible time. Current efforts towards this goal remain fruitless without a full understanding of the behavior of the virus and its adaptor proteins. This review provides an overview of the biological properties, functional mechanisms, and molecular components of SARS-CoV-2, along with investigational therapeutic and preventive approaches for this virus. Since the proteolytic cleavage of the S protein is critical for virus penetration into cells, a set of drugs, such as chloroquine, hydroxychloroquine, camostat mesylate have been tested in clinical trials to suppress this event. In addition to angiotensin-converting enzyme 2, the role of CD147 in the viral entrance has also been proposed. Mepolizumab has shown to be effective in blocking the virus's cellular entrance. Antiviral drugs, such as remdesivir, ritonavir, oseltamivir, darunavir, lopinavir, zanamivir, peramivir, and oseltamivir, have also been tested as treatments for COVID-19. Regarding preventive vaccines, the whole virus, vectors, nucleic acids, and structural subunits have been suggested for vaccine development. Mesenchymal stem cells and natural killer cells could also be used against SARS-CoV-2. All the above-mentioned strategies, as well as the role of nanomedicine for the diagnosis and treatment of SARS-CoV-2 infection, have been discussed in this review.
Collapse
Affiliation(s)
- Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Ali Haghbin
- Department of Pediatrics, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| |
Collapse
|
35
|
Micoli F, MacLennan CA. Outer membrane vesicle vaccines. Semin Immunol 2020; 50:101433. [PMID: 33309166 DOI: 10.1016/j.smim.2020.101433] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 10/22/2022]
Abstract
Outer Membrane Vesicles (OMV) have received increased attention in recent years as a vaccine platform against bacterial pathogens. OMV from Neisseria meningitidis serogroup B have been extensively explored. Following the success of the MeNZB OMV vaccine in controlling an outbreak of N. meningitidis B in New Zealand, additional research and development resulted in the licensure of the OMV-containing four-component 4CMenB vaccine, Bexsero. This provided broader protection against multiple meningococcal B strains. Advances in the field of genetic engineering have permitted further improvements in the platform resulting in increased yields, reduced endotoxicity and decoration with homologous and heterologous antigens to enhance immuno genicity and provide broader protection. The OMV vaccine platform has been extended to many other pathogens. In this review, we discuss progress in the development of the OMV vaccine delivery platform, highlighting successful applications, together with potential challenges and gaps.
Collapse
Affiliation(s)
| | - Calman A MacLennan
- Bill & Melinda Gates Foundation, 62 Buckingham Gate, London, United Kingdom; Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
36
|
Cardoso VMDO, Moreira BJ, Comparetti EJ, Sampaio I, Ferreira LMB, Lins PMP, Zucolotto V. Is Nanotechnology Helping in the Fight Against COVID-19? FRONTIERS IN NANOTECHNOLOGY 2020. [DOI: 10.3389/fnano.2020.588915] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
37
|
Vaccines based on virus-like nano-particles for use against Middle East Respiratory Syndrome (MERS) coronavirus. Vaccine 2020; 38:5742-5746. [PMID: 32684497 PMCID: PMC7837099 DOI: 10.1016/j.vaccine.2020.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
Recent advances in virus-like nanoparticles against Middle East respiratory syndrome-related coronavirus (MERS-CoV) can initiate vaccine production faster for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), while ensuring the safety, easy administration, and long-term effects. Patients with this viral pathogen suffer from excess mortality. MERS-CoV can spread through bioaerosol transmission from animal or human sources. The appearance of an outbreak in South Korea sparked off a strong urge to design strategies for developing an effective vaccine since the emergence of MERS-CoV in 2012. Well unfortunately, this is an important fact in virus risk management. The studies showed that virus-like nanoparticles (VLPs) could be effective in its goal of stopping the symptoms of MERS-CoV infection. Besides, due to the genetic similarities in the DNA sequencing of SARS-CoV-2 with MERS-CoV and the first identified severe acute respiratory syndrome (SARS-CoV) in China since 2002/2003, strategic approaches could be used to manage SARS-CoV 2. Gathering the vital piece of information obtained so far could lead to a breakthrough in the development of an effective vaccine against SARS-CoV-2, which is prioritized and focussed by the World Health Organization (WHO). This review focuses on the virus-like nanoparticle that got successful results in animal models of MERS-CoV.
Collapse
|
38
|
Mostafa A, Kandeil A, Shehata M, El Shesheny R, Samy AM, Kayali G, Ali MA. Middle East Respiratory Syndrome Coronavirus (MERS-CoV): State of the Science. Microorganisms 2020; 8:E991. [PMID: 32630780 PMCID: PMC7409282 DOI: 10.3390/microorganisms8070991] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Coronaviruses belong to a large family of viruses that can cause disease outbreaks ranging from the common cold to acute respiratory syndrome. Since 2003, three zoonotic members of this family evolved to cross species barriers infecting humans and resulting in relatively high case fatality rates (CFR). Compared to Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV, CFR = 10%) and pandemic Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2, CFR = 6%), the Middle East Respiratory Syndrome Coronavirus (MERS-CoV) has scored the highest CFR (approximately 35%). In this review, we systematically summarize the current state of scientific knowledge about MERS-CoV, including virology and origin, epidemiology, zoonotic mode of transmission, and potential therapeutic or prophylactic intervention modalities.
Collapse
Affiliation(s)
- Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
| | - Ahmed Kandeil
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
| | - Mahmoud Shehata
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
| | - Rabeh El Shesheny
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Abdallah M. Samy
- Entomology Department, Faculty of Science, Ain Shams University, Abbassia, Cairo 11566, Egypt;
| | - Ghazi Kayali
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas, Houston, TX 77030, USA
- Human Link, Baabda 1109, Lebanon
| | - Mohamed A. Ali
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
| |
Collapse
|
39
|
A Recombinant Influenza A/H1N1 Carrying A Short Immunogenic Peptide of MERS-CoV as Bivalent Vaccine in BALB/c Mice. Pathogens 2019; 8:pathogens8040281. [PMID: 31810359 PMCID: PMC6963271 DOI: 10.3390/pathogens8040281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/21/2019] [Accepted: 11/30/2019] [Indexed: 01/26/2023] Open
Abstract
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) became a global human health threat since its first documentation in humans in 2012. An efficient vaccine for the prophylaxis of humans in hotspots of the infection (e.g., Saudi Arabia) is necessary but no commercial vaccines are yet approved. In this study, a chimeric DNA construct was designed to encode an influenza A/H1N1 NA protein which is flanking immunogenic amino acids (aa) 736–761 of MERS-CoV spike protein. Using the generated chimeric construct, a novel recombinant vaccine strain against pandemic influenza A virus (H1N1pdm09) and MERS-CoV was generated (chimeric bivalent 5 + 3). The chimeric bivalent 5 + 3 vaccine strain comprises a recombinant PR8-based vaccine, expressing the PB1, HA, and chimeric NA of pandemic 2009 H1N1. Interestingly, an increase in replication efficiency of the generated vaccine strain was observed when compared to the PR8-based 5 + 3 H1N1pdm09 vaccine strain that lacks the MERS-CoV spike peptide insert. In BALB/c mice, the inactivated chimeric bivalent vaccine induced potent and specific neutralizing antibodies against MERS-CoV and H1N1pdm09. This novel approach succeeded in developing a recombinant influenza virus with potential use as a bivalent vaccine against H1N1pdm09 and MERS-CoV. This approach provides a basis for the future development of chimeric influenza-based vaccines against MERS-CoV and other viruses.
Collapse
|