logo logo
Spontaneously released Neisseria meningitidis outer membrane vesicles as vaccine platform: production and purification. Gerritzen Matthias J H,Salverda Merijn L M,Martens Dirk E,Wijffels René H,Stork Michiel Vaccine Outer membrane vesicles (OMVs) are nanoparticles produced by Gram-negative bacteria that can be used as vaccines. The application of OMVs as vaccine component can be expanded by expressing heterologous antigens on OMVs, creating an OMV-based antigen presenting platform. This study aims to develop a production process for such OMV-based vaccines and studies a production method based on meningococcal OMVs that express heterologous antigens on their surface. As a proof of concept, the Borrelia burgdorferi antigens OspA and OspC were expressed on Neisseria meningitidis OMVs to create a concept anti-Lyme disease vaccine. Production of OMVs released in the culture supernatant was induced by high dissolved oxygen concentrations and purification was based on scalable unit operations. A crude recovery of 90 mg OMV protein could be obtained per liter culture. Expressing heterologous antigens on the OMVs did result in minor reduction of bacterial growth, while OMV production remained constant. The antigen expression did not alter the OMV characteristics. This study shows that production of well characterized OMVs containing heterologous antigens is possible with high yields by combining high oxygen concentrations with an optimized purification process. It is concluded that heterologous OMVs show potential as a vaccine platform. 10.1016/j.vaccine.2019.01.076
The Francisella tularensis Polysaccharides: What Is the Real Capsule? Microbiology and molecular biology reviews : MMBR is a tier 1 select agent responsible for tularemia in humans and a wide variety of animal species. Extensive research into understanding the virulence factors of the bacterium has been ongoing to develop an efficacious vaccine. At least two such virulence factors are described as capsules of : the O-antigen capsule and the capsule-like complex (CLC). These two separate entities aid in avoiding host immune defenses but have not been clearly differentiated. These components are distinct and differ in composition and genetic basis. The O-antigen capsule consists of a polysaccharide nearly identical to the lipopolysaccharide (LPS) O antigen, whereas the CLC is a heterogeneous complex of glycoproteins, proteins, and possibly outer membrane vesicles and tubes (OMV/Ts). In this review, the current understanding of these two capsules is summarized, and the historical references to "capsules" of are clarified. A significant amount of research has been invested into the composition of each capsule and the genes involved in synthesis of the polysaccharide portion of each capsule. Areas of future research include further exploration into the molecular regulation and pathways responsible for expression of each capsule and further elucidating the role that each capsule plays in virulence. 10.1128/MMBR.00065-19
Bioengineered bacteria-derived outer membrane vesicles as a versatile antigen display platform for tumor vaccination via Plug-and-Display technology. Nature communications An effective tumor vaccine vector that can rapidly display neoantigens is urgently needed. Outer membrane vesicles (OMVs) can strongly activate the innate immune system and are qualified as immunoadjuvants. Here, we describe a versatile OMV-based vaccine platform to elicit a specific anti-tumor immune response via specifically presenting antigens onto OMV surface. We first display tumor antigens on the OMVs surface by fusing with ClyA protein, and then simplify the antigen display process by employing a Plug-and-Display system comprising the tag/catcher protein pairs. OMVs decorated with different protein catchers can simultaneously display multiple, distinct tumor antigens to elicit a synergistic antitumour immune response. In addition, the bioengineered OMVs loaded with different tumor antigens can abrogate lung melanoma metastasis and inhibit subcutaneous colorectal cancer growth. The ability of the bioengineered OMV-based platform to rapidly and simultaneously display antigens may facilitate the development of these agents for personalized tumour vaccines. 10.1038/s41467-021-22308-8
Designer outer membrane vesicles as immunomodulatory systems - Reprogramming bacteria for vaccine delivery. Gnopo Yehou M D,Watkins Hannah C,Stevenson Taylor C,DeLisa Matthew P,Putnam David Advanced drug delivery reviews Vaccines often require adjuvants to be effective. Traditional adjuvants, like alum, activate the immune response but in an uncontrolled way. Newer adjuvants help to direct the immune response in a more coordinated fashion. Here, we review the opportunity to use the outer membrane vesicles (OMVs) of bacteria as a way to modulate the immune response toward making more effective vaccines. This review outlines the different types of OMVs that have been investigated for vaccine delivery and how they are produced. Because OMVs are derived from bacteria, they have compositions that may not be compatible with parenteral delivery in humans; therefore, we also review the strategies brought to bear to detoxify OMVs while maintaining an adjuvant profile. OMV-based vaccines can be derived from the pathogens themselves, or can be used as surrogate constructs to mimic a pathogen through the heterologous expression of specific antigens in a desired host source strain, and approaches to doing so are reviewed. Additionally, the emerging area of engineered pathogen-specific carbohydrate sequences, or glycosylated OMVs is reviewed and contrasted with protein antigen delivery. Existing OMV-based vaccines as well as their routes of administration round out the text. Overall, this is an exciting time in the OMV field as it matures and leads to more effective and targeted ways to induce desired pathogen-specific immune responses. 10.1016/j.addr.2017.05.003
Bioengineering Bacterial Vesicle-Coated Polymeric Nanomedicine for Enhanced Cancer Immunotherapy and Metastasis Prevention. Chen Qi,Bai Hongzhen,Wu Wangteng,Huang Guojun,Li Yang,Wu Min,Tang Guping,Ping Yuan Nano letters We herein propose a bioengineering approach where bacterial outer membrane vesicles (OMVs) were coated on drug-loaded polymeric micelles to generate an innovative nanomedicine for effective cancer immunotherapy and metastasis prevention. Whereas OMVs could activate the host immune response for cancer immunotherapy, the loaded drug within polymeric micelles would exert both chemotherapeutic and immunomodulatory roles to sensitize cancer cells to cytotoxic T lymphocytes (CTLs) and to kill cancer cells directly. We demonstrated that the systemic injection of such a bioinspired immunotherapeutic agent would not only provide effective protective immunity against melanoma occurrence but also significantly inhibited tumor growth in vivo and extended the survival rate of melanoma mice. Importantly, the nanomedicine could also effectively inhibit tumor metastasis to the lung. The bioinspired immunomodulatory nanomedicine we have developed repurposes the bacterial-based formulation for cancer immunotherapy, which also defines a useful bioengineering strategy to the improve current cancer immunotherapeutic agents and delivery systems. 10.1021/acs.nanolett.9b02182
A naturally derived outer-membrane vesicle vaccine protects against lethal pulmonary Burkholderia pseudomallei infection. Nieves Wildaliz,Asakrah Saja,Qazi Omar,Brown Katherine A,Kurtz Jonathan,Aucoin David P,McLachlan James B,Roy Chad J,Morici Lisa A Vaccine Burkholderia pseudomallei, and other members of the Burkholderia, are among the most antibiotic-resistant bacterial species encountered in human infection. Mortality rates associated with severe B. pseudomallei infection approach 50% despite therapeutic treatment. A protective vaccine against B. pseudomallei would dramatically reduce morbidity and mortality in endemic areas and provide a safeguard for the U.S. and other countries against biological attack with this organism. In this study, we investigated the immunogenicity and protective efficacy of B. pseudomallei-derived outer membrane vesicles (OMVs). Vesicles are produced by Gram-negative and Gram-positive bacteria and contain many of the bacterial products recognized by the host immune system during infection. We demonstrate that subcutaneous (SC) immunization with OMVs provides significant protection against an otherwise lethal B. pseudomallei aerosol challenge in BALB/c mice. Mice immunized with B. pseudomallei OMVs displayed OMV-specific serum antibody and T-cell memory responses. Furthermore, OMV-mediated immunity appears species-specific as cross-reactive antibody and T cells were not generated in mice immunized with Escherichia coli-derived OMVs. These results provide the first compelling evidence that OMVs represent a non-living vaccine formulation that is able to produce protective humoral and cellular immunity against an aerosolized intracellular bacterium. This vaccine platform constitutes a safe and inexpensive immunization strategy against B. pseudomallei that can be exploited for other intracellular respiratory pathogens, including other Burkholderia and bacteria capable of establishing persistent infection. 10.1016/j.vaccine.2011.08.058
Lytic potential of Lysobacter capsici VKM B-2533: bacteriolytic enzymes and outer membrane vesicles. Afoshin A S,Kudryakova I V,Borovikova A O,Suzina N E,Toropygin I Yu,Shishkova N A,Vasilyeva N V Scientific reports Recent recurrent outbreaks of bacterial resistance to antibiotics have shown the critical need to identify new lytic agents to combat them. The species Lysobacter capsici VKM B-2533 possesses a potent antimicrobial action against a number of bacteria, fungi and yeasts. Its activity can be due to the impact of bacteriolytic enzymes, antibiotics and peptides. This work isolated four homogeneous bacteriolytic enzymes and a mixture of two proteins, which also had a bacteriolytic activity. The isolates included proteins identical to L. enzymogenes α- and β-lytic proteases and lysine-specific protease. The proteases of 26 kDa and 29 kDa and a protein identified as N-acetylglycosaminidase had not been isolated in Lysobacter earlier. The isolated β-lytic protease digested live methicillin-resistant staphylococcal cells with high efficiency (minimal inhibitory concentration, 2.85 μg/mL). This property makes the enzyme deserving special attention. A recombinant β-lytic protease was produced. The antimicrobial potential of the bacterium was contributed to by outer membrane vesicles (OMVs). L. capsici cells were found to form a group of OMVs responsible for antifungal activity. The data are indicative of a significant antimicrobial potential of this bacterium that requires thorough research. 10.1038/s41598-020-67122-2
Vaccine Based on Outer Membrane Vesicles Using Hydrogels as Vaccine Delivery System. Pastor Yadira,Ting Isaiah,Berzosa Melibea,Irache Juan M,Gamazo Carlos Methods in molecular biology (Clifton, N.J.) A simple procedure for obtaining outer membrane vesicles from Salmonella enterica and the use of hydrogels as vaccine delivery system is described. A heat treatment in saline solution of whole bacteria rendered the release of outer membrane vesicles containing relevant antigenic components. The immunogenicity of these antigens when administered by the intranasal route may be improved after embedment into hydrogels to increase residence half-time and thus activate the mucosal immune system. 10.1007/978-1-0716-0791-6_14
The Effect of Growth Stage and Isolation Method on Properties of ClearColi™ Outer Membrane Vesicles (OMVs). Current microbiology Outer membrane vesicles (OMVs) are nanosized spherical blebs derived from the outer membrane of gram-negative bacteria. Outer membrane vesicles (OMVs) play important roles in various physiological functions of bacteria. They can be applied as native vaccines or vaccine adjuvants. The objective of this study was to determine the appropriate growth phase and isolation method for OMV separation from ClearColi™, an endotoxin-free strain of E. coli. It was demonstrated that the yield of OMVs is increased while the bacteria are growing. Herein, although total protein concentration of OMVs isolated from the stationary phase is more than other phases; the pre-stationary phase was selected for OMV isolation due to release of smaller size of OMVs as compared to other phases. In the current study, to obtain OMVs with high yield, proper size, and homogeneity, different concentration methods including protein precipitation by ammonium sulfate (AS) and ultrafiltration (UF) were combined to ultracentrifugation (UC) or precipitation-based exosome isolation kit. Among the examined isolation methods, AS (70%) + UC resulted in the highest yield of OMVs. The TEM results demonstrated bilayer round-shaped OMVs isolated by this method. Although AS (70%) + kit resulted in more heterogeneous in size and larger OMVs as compared to AS (70%) + UC, it is applicable when high yield of OMVs is required and UC is not available. Totally, isolation of ClearColi™ OMVs from pre-stationary phase using AS (70%) + UC with enhanced yield can be applied in vaccine research studies. 10.1007/s00284-021-02414-y
Isolation of Outer Membrane Vesicles from Helicobacter pylori. Windle Henry J Methods in molecular biology (Clifton, N.J.) Outer membrane vesicles (OMV) shed by pathogenic bacteria have multifunctional roles in disease initiation and progression. Further, their efficacy as novel vaccines has underscored their importance as potential therapeutics. Consequently, to advance allied research related to their immunogenicity and pathogenicity it is important to separate these vesicular structures from parental cells and demonstrate them to be free from cellular debris and other non-vesicle-related constituents such as protein aggregates. To do so represents a key step in initiating OMV-related studies and the techniques and strategies adopted by the H. pylori community to achieve this will be the focus of this chapter.The key methods used typically to obtain a heterogeneous mixture of OMV (size range: ~20-300 nm in diameter) include growth of bacteria in broth culture followed by differential centrifugation, filtration, and concentration to separate OMV from the intact organisms. Additional measures may be adopted to further size-fractionate the population of OMV including gel filtration or density gradient ultra-centrifugation in order to facilitate differentiation between the activities of small versus large OMV, as recent studies have demonstrated differential modes of entry into host cells as well as size-dependent differences in the OMV proteome (Turner et al., Front Immunol 9:1466, 2018). The OMV from H. pylori harbor many of the virulence factors associated with gastric disease including the CagA oncoprotein, the cytotoxin VacA, and the HtrA protease (Olofsson et al., mBio 5:e00979-14, 2014; Mullaney et al., Proteomics Clin Appl 3:785-96, 2009) and their close association with areas of cell-cell contact and efficient endocytosis supports a role for these complexes in gastric disease (Turkina et al., FEMS Microbiol Lett 362:fnv076, 2015). 10.1007/978-1-0716-1302-3_13
Bacterial extracellular vesicles: Understanding biology promotes applications as nanopharmaceuticals. Jahromi Leila Pourtalebi,Fuhrmann Gregor Advanced drug delivery reviews Extracellular vesicle (EV)-mediated communication between proximal and distant cells is a highly conserved characteristic in all of the life domains, including bacteria. These vesicles that contain a variety of biomolecules, such as proteins, lipids, nucleic acids, and small-molecule metabolites play a key role in the biology of bacteria. They are one of the key underlying mechanisms behind harmful or beneficial effects of many pathogenic, symbiont, and probiotic bacteria. These nanoscale EVs mediate extensive crosstalk with mammalian cells and deliver their cargos to the host. They are stable in physiological condition, can encapsulate diverse biomolecules and nanoparticles, and their surface could be engineered with available technologies. Based on favorable characteristics of bacterial vesicles, they can be harnessed for designing a diverse range of therapeutics and diagnostics for treatment of disorders including tumors and resistant infections. However, technical limitations for their production, purification, and characterization must be addressed in future studies. 10.1016/j.addr.2021.03.012
Fight bacteria with bacteria: Bacterial membrane vesicles as vaccines and delivery nanocarriers against bacterial infections. Gan Yingying,Li Chengnan,Peng Xinran,Wu Shuang,Li Yuzhen,Tan Jeremy P K,Yang Yi Yan,Yuan Peiyan,Ding Xin Nanomedicine : nanotechnology, biology, and medicine Bacterial membrane vesicles (MVs) are particles secreted by bacteria with diameter of 20-400 nm. The pathogen-associated molecular patterns (PAMPs) present on the surface of MVs are capable of activating human immune system, leading to non-specific immune response and specific immune response. Due to the immunostimulatory properties and proteoliposome nanostructures, MVs have been increasingly explored as vaccines or delivery systems for the prevention and treatment of bacterial infections. Herein, the recent progresses of MVs for antibacterial applications are reviewed to provide an overview of MVs vaccines and MVs-related delivery systems. In addition, the safety issues of bacterial MVs are discussed to demonstrate their potential for clinical translation. In the end of this review, the challenges of bacterial MVs as vaccines and delivery systems for clinical applications are highlighted with the purpose of predicting future research directions in this field. 10.1016/j.nano.2021.102398
Rapid On-Demand Extracellular Vesicle Augmentation with Versatile Oligonucleotide Tethers. Yerneni Saigopalakrishna S,Lathwal Sushil,Shrestha Pradeep,Shirwan Haval,Matyjaszewski Krzysztof,Weiss Lee,Yolcu Esma S,Campbell Phil G,Das Subha R ACS nano Exosomes show potential as ideal vehicles for drug delivery because of their natural role in transferring biological cargo between cells. However, current methods to engineer exosomes without negatively impacting their function remain challenging. Manipulating exosome-secreting cells is complex and time-consuming, while direct functionalization of exosome surface proteins suffers from low specificity and low efficiency. We demonstrate a rapid, versatile, and scalable method with oligonucleotide tethers to enable diverse surface functionalization on both human and murine exosomes. These exosome surface modifiers, which range from reactive functional groups and small molecules to aptamers and large proteins, can readily and efficiently enhance native exosome properties. We show that cellular uptake of exosomes can be specifically altered with a tethered AS1411 aptamer, and targeting specificity can be altered with a tethered protein. We functionalize exosomes with an immunomodulatory protein, FasL, and demonstrate their biological activity both and . FasL-functionalized exosomes, when bioprinted on a collagen matrix, allows spatial induction of apoptosis in tumor cells and, when injected in mice, suppresses proliferation of alloreactive T cells. This oligonucleotide tethering strategy is independent of the exosome source and further circumvents the need to genetically modify exosome-secreting cells. 10.1021/acsnano.9b04651
Bacterial components as naturally inspired nano-carriers for drug/gene delivery and immunization: Set the bugs to work? Biotechnology advances Drug delivery is a rapidly growing area of research motivated by the nanotechnology revolution, the ideal of personalized medicine, and the desire to reduce the side effects of toxic anti-cancer drugs. Amongst a bewildering array of different nanostructures and nanocarriers, those examples that are fundamentally bio-inspired and derived from natural sources are particularly preferred. Delivery of vaccines is also an active area of research in this field. Bacterial cells and their components that have been used for drug delivery, include the crystalline cell-surface layer known as "S-layer", bacterial ghosts, bacterial outer membrane vesicles, and bacterial products or derivatives (e.g. spores, polymers, and magnetic nanoparticles). Considering the origin of these components from potentially pathogenic microorganisms, it is not surprising that they have been applied for vaccines and immunization. The present review critically summarizes their applications focusing on their advantages for delivery of drugs, genes, and vaccines. 10.1016/j.biotechadv.2018.02.016
Extracellular vesicles from symbiotic vaginal lactobacilli inhibit HIV-1 infection of human tissues. Ñahui Palomino Rogers A,Vanpouille Christophe,Laghi Luca,Parolin Carola,Melikov Kamran,Backlund Peter,Vitali Beatrice,Margolis Leonid Nature communications The vaginal microbiota, dominated by Lactobacillus spp., plays a key role in preventing HIV-1 transmission. Here, we investigate whether the anti-HIV effect of lactobacilli is mediated by extracellular vesicles (EVs) released by these bacteria. Human cervico-vaginal and tonsillar tissues ex vivo, and cell lines were infected with HIV-1 and treated with EVs released by lactobacilli isolated from vaginas of healthy women. EVs released by L. crispatus BC3 and L. gasseri BC12 protect tissues ex vivo and isolated cells from HIV-1 infection. This protection is associated with a decrease of viral attachment to target cells and viral entry due to diminished exposure of Env that mediates virus-cell interactions. Inhibition of HIV-1 infection is associated with the presence in EVs of several proteins and metabolites. Our findings demonstrate that the protective effect of Lactobacillus against HIV-1 is, in part, mediated by EVs released by these symbiotic bacteria. If confirmed in vivo, this finding may lead to new strategies to prevent male-to-female sexual HIV-1 transmission. 10.1038/s41467-019-13468-9
Release of Staphylococcus aureus extracellular vesicles and their application as a vaccine platform. Wang Xiaogang,Thompson Christopher D,Weidenmaier Christopher,Lee Jean C Nature communications Secretion of extracellular vesicles (EVs), a process common to eukaryotes, archae, and bacteria, represents a secretory pathway that allows cell-free intercellular communication. Microbial EVs package diverse proteins and influence the host-pathogen interaction, but the mechanisms underlying EV production in Gram-positive bacteria are poorly understood. Here we show that EVs purified from community-associated methicillin-resistant Staphylococcus aureus package cytosolic, surface, and secreted proteins, including cytolysins. Staphylococcal alpha-type phenol-soluble modulins promote EV biogenesis by disrupting the cytoplasmic membrane; whereas, peptidoglycan cross-linking and autolysin activity modulate EV production by altering the permeability of the cell wall. We demonstrate that EVs purified from a S. aureus mutant that is genetically engineered to express detoxified cytolysins are immunogenic in mice, elicit cytolysin-neutralizing antibodies, and protect the animals in a lethal sepsis model. Our study reveals mechanisms underlying S. aureus EV production and highlights the usefulness of EVs as a S. aureus vaccine platform. 10.1038/s41467-018-03847-z
Bacterial outer membrane vesicles suppress tumor by interferon-γ-mediated antitumor response. Kim Oh Youn,Park Hyun Taek,Dinh Nhung Thi Hong,Choi Seng Jin,Lee Jaewook,Kim Ji Hyun,Lee Seung-Woo,Gho Yong Song Nature communications Gram-negative bacteria actively secrete outer membrane vesicles, spherical nano-meter-sized proteolipids enriched with outer membrane proteins, to the surroundings. Outer membrane vesicles have gained wide interests as non-living complex vaccines or delivery vehicles. However, no study has used outer membrane vesicles in treating cancer thus far. Here we investigate the potential of bacterial outer membrane vesicles as therapeutic agents to treat cancer via immunotherapy. Our results show remarkable capability of bacterial outer membrane vesicles to effectively induce long-term antitumor immune responses that can fully eradicate established tumors without notable adverse effects. Moreover, systematically administered bacterial outer membrane vesicles specifically target and accumulate in the tumor tissue, and subsequently induce the production of antitumor cytokines CXCL10 and interferon-γ. This antitumor effect is interferon-γ dependent, as interferon-γ-deficient mice could not induce such outer membrane vesicle-mediated immune response. Together, our results herein demonstrate the potential of bacterial outer membrane vesicles as effective immunotherapeutic agent that can treat various cancers without apparent adverse effects.Bacterial outer membrane vesicles (OMVs) contain immunogens but no study has yet examined their potential in treating cancer. Here, the authors demonstrate that OMVs can suppress established tumours and prevent tumour metastasis by an interferon-γ mediated antitumor response. 10.1038/s41467-017-00729-8
Bioengineering bacterial outer membrane vesicles as vaccine platform. Gerritzen Matthias J H,Martens Dirk E,Wijffels René H,van der Pol Leo,Stork Michiel Biotechnology advances Outer membrane vesicles (OMVs) are naturally non-replicating, highly immunogenic spherical nanoparticles derived from Gram-negative bacteria. OMVs from pathogenic bacteria have been successfully used as vaccines against bacterial meningitis and sepsis among others and the composition of the vesicles can easily be engineered. OMVs can be used as a vaccine platform by engineering heterologous antigens to the vesicles. The major advantages of adding heterologous proteins to the OMV are that the antigens retain their native conformation, the ability of targeting specific immune responses, and a single production process suffices for many vaccines. Several promising vaccine platform concepts have been engineered based on decorating OMVs with heterologous antigens. This review discusses these vaccine concepts and reviews design considerations as the antigen location, the adjuvant function, physiochemical properties, and the immune response. 10.1016/j.biotechadv.2017.05.003