Next Article in Journal
Latent Tuberculosis: A Promising New Compound to Treat Non-Replicating and Intramacrophagic Mycobacteria
Next Article in Special Issue
Application of Plant-Derived Nanoparticles (PDNP) in Food-Producing Animals as a Bio-Control Agent against Antimicrobial-Resistant Pathogens
Previous Article in Journal
A Single Session of Bifrontal tDCS Can Improve Facial Emotion Recognition in Major Depressive Disorder: An Exploratory Pilot Study
Previous Article in Special Issue
The Potential of Antibody Technology and Silver Nanoparticles for Enhancing Photodynamic Therapy for Melanoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Discovery of the Role of Outer Membrane Vesicles against Bacteria

by
Sofia Combo
1,2,
Sérgio Mendes
1,2,
Kaare Magne Nielsen
3,
Gabriela Jorge da Silva
1,2 and
Sara Domingues
1,2,*
1
Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
2
Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
3
Department of Life Sciences and Health, Faculty of Health Sciences at Oslo Metropolitan University, 0130 Oslo, Norway
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(10), 2399; https://doi.org/10.3390/biomedicines10102399
Submission received: 3 August 2022 / Revised: 2 September 2022 / Accepted: 22 September 2022 / Published: 26 September 2022
(This article belongs to the Special Issue Nanobiomaterials: From Fundamentals to Biomedical Applications)

Abstract

:
Gram-negative bacteria are intrinsically resistant to many commercialized antibiotics. The outer membrane (OM) of Gram-negative bacteria prevents the entry of such antibiotics. Outer membrane vesicles (OMV) are naturally released from the OM of Gram-negative bacteria for a range of purposes, including competition with other bacteria. OMV may carry, as part of the membrane or lumen, molecules with antibacterial activity. Such OMV can be exposed to and can fuse with the cell surface of different bacterial species. In this review we consider how OMV can be used as tools to deliver antimicrobial agents. This includes the characteristics of OMV production and how this process can be used to create the desired antibacterial activity of OMV.

1. Introduction

Membrane vesicles (MV) are formed and released by a broad range of cells, from bacteria to human cells, with different nomenclature attributed according to the budding cell [1,2,3]. These MV are mostly spheres from the membrane of the cells with a large size range from 20 nm to 10 µm, depending on the donor cell [4,5]. The formation of vesicles occurs by vesiculation from living cells as a result of a disruption of the membrane caused by internal mechanisms or induced by an external signal. Depending on the donor, the bacterial MV have different functions, and the same cell can also produce MV with different functions. The main function of MV is the transport of different molecules, such as lipids, proteins, and nucleic acids. The molecules that constitute the cargo of the MV as well as their target will differ depending on the stimuli [6,7,8,9]. The MV show diverse functions from secretion of toxins and virulence factors to the modulation of the target cell, acquisition of nutrients and even resistance to stress [8,10,11,12].
Depending on their constitution and biogenesis, MV secreted by Gram-negative bacteria can be classified as outer membrane vesicles (OMV), outer-inner membrane vesicles, explosive outer membrane vesicles or tube-shaped membranous structures [13]. The most common MV from Gram-negative bacteria are the OMV, also named membrane blebs or outer membrane blebs [6,14]. MV released by Gram-positive bacteria include the cytoplasmic membrane vesicles and the tube-shaped membranous structures [13].
Antibiotic resistance is due to the bacteria’s ability of adaptation to the action of antimicrobial molecules as well as to prevent them reaching the target site [15,16]. OMV have a natural role in antimicrobial resistance since they can act as a decoy or remove antibiotics that cross the cell wall, allowing bacteria to survive [6,16]. New approaches have been developed to understand whether OMV may be used as a tool to deliver antibiotics against bacteria. Due to the composition similarity of OMV and the outer membrane (OM) of the cell wall of Gram-negative bacteria, delivery of OMV content into Gram-negative bacterial cells is more efficient; nonetheless, an antimicrobial effect can also be seen against Gram-positive bacteria. Additional advantages of OMV as antibiotic delivery tools include their stability, the cargo protection against enzymatic degradation, the ability to incorporate both hydrophilic and hydrophobic molecules and the capacity to selectively target other bacterial cells [17]. Several challenges have been reported including the need to optimize the loading of different antibiotics, as well as the binding of OMV with the desired target cell and resulting toxicity.
Although different types of MV are secreted by bacteria, here we focus on OMV, the archetypal vesicles of Gram-negative bacteria. We discuss key aspects of bacterial OMV and their potential role as a delivery tool of molecules with antibacterial activity. Emphasis will be given to the biogenesis and the antimicrobial activity of OMV. Different studies have shown OMV activity against different bacterial species due to the carriage of antibiotic molecules, or due to some molecules that are naturally present in OMV such as lysins, which behave as an antibacterial molecule when exposed to recipient bacteria [12].

2. OMV Biogenesis in Gram-Negative Bacteria

Gram-negative bacteria have an envelope comprising an OM and an inner membrane (IM) with a periplasmic space in between, which contains a layer of peptidoglycan (PG). In the OM there are lipopolysaccharides (LPS) linked covalently by the lipidic moiety and proteins bound as β-barrels, while in the IM the proteins are bound as α-helical [18].
The destabilization of ligations in a bacterium’s cell wall can lead to the detachment of the OM from the cell membrane. Consequently, the natural stabilization of the molecular charges allows the formation of the OMV [18,19]. OMV are nanostructures with size range between 20 and 250 nm that are secreted from the bacteria’s OM, being composed of phospholipids, LPS and outer membrane proteins (OMP) [19]. During formation, molecules such as nucleic acids and proteins from the periplasm and cytoplasm of the cells can be localized to the lumen of the vesicle, making it a vehicle for antibiotic resistance and virulence dissemination [18,20]. Some bacteria can also incorporate external antibiotics into vesicles, allowing isolation of antibiotics inside OMV and cell membrane stabilization [6].
The first step to the formation of OMV is the disruption of the connection OM-PG-IM without damage or loss of membrane integrity [18]. To explain the formation of OMV, three models have been created (Figure 1), which are not mutually exclusive: deficiency of lipoprotein (LPP) or its links in OM; increase of PG or other lipids residues; repulsion of negatively charged LPS [19].
  • LPP links deficiency (Figure 1a): the presence of LPP in the unbound form has been found in OMV, indicating that the covalent links were broken, or their distribution was not homogenous, since the conversion of free-form LPP into bound form is reversible [21]. These characteristics seems to be induced by the non-proportional growth of the OM compared with the PG layer [22]. The relation of the lack of link between OmpA and PG has been proven to be essential to the production of OMV in Salmonella spp. [23].
  • Increase of misfolded PG (Figure 1b): Autolysins have a role in cleaving the covalent links of PG, resulting in cell wall remodeling. The lack of these enzymes increases the amount of peptides in periplasmatic space and other components leading to turgor pressure and therefore to OMV formation. Several studies explore the lack of autolysins to increase the concentration of proteins in the periplasmatic space and therefore converge to this model [24,25].
  • Repulsion of negatively charged LPS (Figure 1c): A study suggested that the repulsion of negatively charge B-band LPS in cells exposed to gentamicin, with great affinity to LPS, induce the release of OMV as a way of antibiotic resistance in which gentamicin was incorporated into OMV. That repulsion increased the production of vesicles in P. aeruginosa [6].
Several molecules are important components and regulators of OMV, including as a regulator of OMV protein composition, such as OmpA from A. baumannii [26]. Cells have several mechanisms of response to the disruptions of the connection OM-PG-IM that can be different between species. In E. coli, if mutations lead to the absence of LPP, Omp may substitute it [19]. σE, a transcriptional factor and a modulator of OMV formation, is activated to respond to the increase of misfolded OMP and consequently downregulates OmpA and LPP by MicA and Reg26, respectively [18]. Other molecules such as DegP, which behaves as a periplasmatic chaperone at high temperature and a protease at low temperature, seems to prevent the accumulation of proteinaceous waste in periplasmatic space [18]. However, for some perturbations such as LPS with highly charged O-antigen in OM, those molecules are enriched in P. aeruginosa OMV. OMV seem to be a bacterium’s own mechanism of defense for the stabilization of the OM charges and therefore cell survival [2].
In general, in the biogenesis of OMV, an increase of the space between the connection OM-PG-IM is necessary. However, its complex formation allows the presence of multiple molecules in OMV that are involved in some essential survival mechanisms of the cell.
To produce higher levels of OMV, growth conditions may be manipulated, including change or addition of parameters such as temperature, antibiotics, serum, active oxygen species mimic molecules, EDTA and lack of amino acids [6,27,28,29,30,31]. The increase of temperature may precipitate proteins increasing the space between OM and IM and leading to the disruption of the LPP links [32,33]. In addition, the use of other physical methods such as electroporation, extrusion and sonication are used to increase the production of OMV, but also to load non-natural molecules inside, once they cause high damage to the cell wall [34,35,36,37,38,39].

3. OMV Functions

OMV have several functions that are related with their diverse molecular composition (Table 1), which can be different for each bacterium but also differs with different environmental inducers. Due to its complex composition, one OMV can have more than one function. OMV are used by Gram-negative bacteria to secrete toxins and other virulence factors that can induce cytotoxicity activity, but also other molecules that are important for modulation and invasion of the host that are capable to induce responses from host organisms. In addition, the OMV can also carry molecules responsible for improving the bacterial survival such as those associated with signaling, biofilm production or gene transfer. Beyond the capability to survive, OMV also have an ability to attach to other bacteria, and deliver enzymes known to have antibacterial effect [40].

4. Optimization of OMV Production

Several vesiculation-stimulating agents, such as antibiotics and other stimulating factors, such as the presence of serum or limitation of amino acids, can be used to destabilize the cell wall of Gram-negative bacteria and hence increase formation of vesicles [6,27,28,29,30].
Exposure to some antibiotics increases the production of OMV, such as in P. aeruginosa, where OMV secretion is enhanced by ciprofloxacin [33,56], or in Stenotrophomonas maltophilia, where ciprofloxacin and imipenem increased secretion of OMV but with different compositions and through different mechanisms of formation [57].
Increased temperatures cause the misfolding of proteins and consequently lead to an accumulation of proteins in the periplasmatic space. The accumulation will lead to the formation of OMV through the created pressure [32,33]. However, for some Gram-negative bacteria, OMV formation is regulated by temperature, where low temperature allows vesiculation and a high temperature decreases production or has no effect [58,59]. The effects of temperature changes with the bacterial species may be related with the presence of enzymes with the ability to change their function at higher temperatures, such as heat shock protein DegP (HtrA) in E. coli that can change from chaperones to protease [60] or MucD in P. aeruginosa that can acquire protease functions [61].
The presence of serum can have some antibacterial effects on bacteria, related with their own components, namely antibodies [31]. However, the hyperproduction of OMV seems to be a resistance mechanism in Neisseria gonorrhoeae and Haemophilus influenzae [27,28]. Reactive oxygen species mimic molecules, such as hydrogen peroxidase, that have an impact on OMV overproduction in P. aeruginosa [59]. The chelating agent ethylenediaminetetra-acetic acid (EDTA) has been proven to be effective in the stimulation of the production of OMV from Neisseria meningitidis by capturing the calcium ions from the medium that allows the stabilization of the membrane [62,63]. There are also reports that show that limitation of amino acids induces and regulates the production of OMV in E. coli and in Francisella spp. [29,30].

5. Antimicrobial Activity of OMV

The OMV antimicrobial activity is appealing for treatment purposes, especially against Gram-negative bacteria since it allows bridging with its cell wall. During OMV biogenesis, some molecules with antibacterial activity will be naturally included in the vesicle lumen (Table 1). At the same time, several environmental inductors can enhance or contribute to the inclusion of those molecules into the OMV. In both cases, OMV can act as an antimicrobial agent.

5.1. OMV with Natural Antimicrobial Activity Cargo

The functions and roles of autolysin, also known as murein hydrolase [64,65], have being explored in Bacillus spp. [66]. Autolysins are usually PG-hydrolyzing endogenous enzymes that naturally lyse the peptide bridges in the PG layer [67], although there are some exceptions where it can have glycosidic activity [68]. There are several types of autolysins related with different mechanisms, such as protein and toxin secretion [69], flagellar formation [70,71], cell separation [72] and antibacterial activity associated to bacterial competition [12,73]. Its relation with vesicles and antimicrobial activity was observed for the first time in P. aeruginosa [6]. The presence of autolysins in OMV seem to be related with its normal location at the PG layer, being included in the cargo of the OMV during the bleb of the OM [6,64]. However, a recent study in Lysobacter spp. found that the distribution of the L5 enzyme, a bacteriolytic peptidase, may not be random, because it is only present in bacteria when this is exposed to a 30% sucrose medium. This study demonstrated that this autolysin is unevenly placed through the periplasmatic space, specifically where the vesiculation occurred and therefore L5 seems to be a factor in OMV biogenesis [74].
There are a few studies that show the antibacterial activity of vesicles with autolysins, such as peptidoglycan hydrolases, from P. aeruginosa PAO1 against different species of Gram-negative bacteria, such as E. coli DH5α and P. aeruginosa PAO1, and Gram-positive bacteria such as Brachybacterium conglomeratum CCM2134, Bacillus spp., Lactococcus lactis ATCC 7962 and Staphylococcus aureus D2C [6,12,75,76]. The antibacterial activity of peptidoglycan hydrolases from OMV from several Gram-negative bacteria have some affinity to some chemotypes of peptidoglycan from target bacteria, such as the A1Υ chemotype, making it more susceptible to cell lysis [66].
Hemolysin is another type of enzyme that is present in OMV from Gram-negative bacteria, such as P. aeruginosa and E. coli [6,54]. One hemolysin from P. aeruginosa has been shown to be responsible for co-regulation of protein secretion but also injection of toxin proteins into other Gram-negative bacteria, including E. coli [77], by type VI secretion system (T6SS) [78]. In addition, T6SS has been shown to be incorporated in OMV from P. aeruginosa [8]. The T6SS system allows the bacteria to compete through the delivery of toxins, that are capable of killing other bacteria [78].
Antimicrobial quinolines, 4-hydroxy-2-heptylquinoline (HHQ) and 4-hydroxy-2-nonylquinoline (HNQ), are synthetized by P. aeruginosa and incorporated into OMV, which acted as an antimicrobial molecule against Staphylococcus epidermidis successfully [55].
Alkaline phosphatase is an enzyme that has been found active in P. aeruginosa and in Myxococcus xanthus OMV, where the active packaging of this enzyme is suggested [6,44,65]. Alkaline phosphatase from E. coli was shown to have antimicrobial activity against Gram-negative bacteria, namely P. aeruginosa [79], however the search for a match between OMV and its antibacterial activity has not yet been proven.
Other groups of enzymes, such as phospholipases and proteases, are present in OMV and may have antibacterial effect, however the need for more studies about their presence and action is necessary [6].

5.2. OMV with Loaded Antimicrobial Cargo

Several studies have reported the use of OMV as a delivery system to transport antibiotics into Gram-negative bacteria. There are two main ways to incorporate antibiotics into OMV: the passive loading, where the addition of the antibiotics during bacterial growth is enough to produce antibiotic-carrying OMV (aOMV), and the active loading approach, where the antibiotics are forced to enter or coat the OMV or OM of bacteria, so it can be part of the produced OMV.

5.2.1. Passive Loading

Passive loading methods use diffusion by osmotic gradient but only for hydrophobic positively charged small molecules. These molecules may pass through the lipophilic membrane because of their opposite charges and their similar affinity to water [80]. In these methods, only the components of the medium or environmental characteristics are changed to destabilise the membrane and allow the entrance of antibiotics and other molecules [80,81].
Up to now, only antibiotics that have been demonstrated to pass through the cell wall of Gram-negative bacteria have been inserted into the OMV. Addition of gentamicin to P. aeruginosa cells showed the production of OMV-carrying gentamicin [6], which had antibacterial effects against both Gram-negative and Gram-positive bacteria species [12]. These gentamicin-carrying OMV showed antibacterial activity against P. aeruginosa 8803, which has a permeability-type resistance to gentamicin [12], highlighting the potential of OMV antimicrobial delivery to overcome resistance. More recently, different antibiotics, such as ceftriaxone, amikacin, azithromycin, ampicillin and levofloxacin, were loaded into A. baumannii OMV, which showed antibacterial effects against enterotoxigenic E. coli, Klebsiella pneumoniae and P. aeruginosa without toxic activity in mice [82].
Further studies are necessary to determine the capability of other medium components or environmental changes to affect the cell wall in ways that allow the intentional entrance of antimicrobials.

5.2.2. Active Loading

The active loading methods consist of forcing the entrance of the molecules into the vesicles, normally by physically damaging the cells or the vesicles. There are three main methods for active loading: electroporation, sonication, and extrusion, and they are already commonly used to produce vesicles from animal cells such as exosomes.
Electroporation is an electro-physical method that uses electron impulses to rearrange the OM of the cell with the consequent creation of pores [34,83]. This method is typically used to make cell transfections of DNA, RNA and proteins, however its ability to translocate large molecules leads to an instable cell wall and therefore more cell death by lysis and less efficacy [34]. This method has been successfully used to insert small interference RNA into OMV from E. coli [35]. Another study showed the possibility to insert any nanoparticles under 10 nm, such as nanoparticles of gold, into OMV from P. aeruginosa through this method, a first step to deliver nanoparticles with antibacterial activity through OMV [84].
Sonication uses ultrasound to compress and decompress cells in order to compromise membrane stability, followed by a second sonication to assemble the membrane fragments and allow the incorporation of external molecules [36]. Mild sonication has been used successfully to induce paclitaxel loading into exosomes from macrophages to treat cancer cells and also to load small RNA into extracellular vesicles from different cell lines [37,85]. This method has already been used in Haemophilus parasuis to induce OMV production, however the protein content changed when compared to the natural OMV [86]. As far as we know, the use of sonication in bacterial studies is mostly used to induce cell lysis, and for detection of biofilms; studies related to OMV loading with sonication have not yet been performed [86,87,88,89].
Extrusion involves the mixture of cells and antibiotics added to a syringe extruder and then forced to pass through a porous membrane, under controlled temperature. The hydrostatic fluid pressure will disrupt the membrane, by increasing the axial tension, and allow the drug entrance at the same time that vesicles are formed; however, variation of size and zeta potential can occur [9,38]. Despite the yield in loading and forming vesicles with this method is high, the vesicles may not be homogenous and its impact in protein membrane structures is not clear, though higher pressures may lead to protein damage and cell death [38,39]. Extrusion has been proven to be efficient to coat OMV from E. coli with gold nanoparticles as well as to load 5-fluorouracil into OMV from E. coli [90,91]. It has also been found in E. coli that this method disrupts the CusCBA, an efflux pump associated with toxic metals that belongs to the same family related to antibiotic resistance [92]. As far as we know, this methodology has not been used for active loading of antibiotics, however it seems a promising technique to produce OMV with antibacterial activity.
Other methods of active loading include a drastic change of temperature, and utilization of saponin. Severe change of temperature in exosomes has proven to be effective in inducing the incorporation of proteins through the compression and decompression of water molecules in cells that lead to the formation of pores and allow the entrance of molecules into vesicles [36,93]. Incubation with saponin, a surfactant molecule that reacts with cholesterol in cell membranes, has been effective in increasing the loading of molecules into extracellular vesicles without altering the size or even the zeta potential [9]. Although there is no cholesterol in Gram-negative bacteria, an analogue called hopanoid has been discovered [94]. Further studies are necessary to understand the possibility of this method in the production of OMV in Gram-negative bacteria.

6. Merging of Vesicles with Target Cells and Delivery of OMV Cargo

The merging of vesicles with Gram-negative target cells occurs in three steps. The first is the membrane contact, followed by the mixing of lipids and fusion of the external layer of the membrane and finally the reorganization of the inner lipidic layer through pore formation and finally mixing of content [95]. The first step requires a high amount of energy that can be given by fusogenic agents. These agents can disrupt the lipidic membrane through the influence of transition from lamellar bilayer-phase lipids into inverted hexagonal-phase lipids; lipid quality is crucial for the membrane rearrangement and content mixing [96,97]. Some of these agents are positive ions, small organic molecules or physical damage to the membrane, such as temperature, in which the difference between them is the energy harnessing process [95].
Attachment of some vesicles can be explained by the charges of Mg2+ and Ca2+ at the bacterial membrane. These ions can form salt-bridges between the vesicles and the membrane of exposed cells, that are rich in those ions [12]. Increasing the composition and concentration of ions in the medium reduces the repulsive electrostatic force, allowing particle aggregation. A recent study demonstrated that acidic pH enhances the merging of OMV-OMV from E. coli [98]. More recently, a fusogenic enzyme, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), has been discovered alongside OMV from M. xanthus and it seems to modulate the fusion with E. coli cells (Figure 2) [65].
Vesicles from several Gram-negative bacterial species can promote the lysis of other bacteria, despite the specific interactions related with different PG chemotypes mentioned above [73]. For instance, P. aeruginosa vesicles do not merge with the cell wall of S. aureus, but rather tend to attach and release their content, such as gentamicin and autolysins, into the extracellular matrix, allowing enzyme activity and therefore antibacterial activity [12]. Although the antibacterial effect is similar to the antibiotic free effect, the antibiotic is delivered near the target, which may increase the success of Gram-positive infection treatments. S-layered species, such as Aneurinibacillus thermoaerophilus and Bacillus spp., prevent the passage of vesicles through the cell wall but not the attachment of the vesicles from P. aeruginosa [69].
In all these cases, the equilibrium of the membrane charges seems to be mandatory. It is necessary that new studies determine if there is a difference between the merging of the different vesicles with other species of bacteria and what cell signaling is associated with the merging after the attachment to the cell wall [65,99,100].

7. Other Nano-Sized Techniques Used for Delivery of Antibiotics

Nowadays, new techniques are developed as a different approach to antibiotic delivery, specifically the coating of nanoparticles with OMV membrane and nanotransformation of antibiotics [101,102]. These techniques have been shown to reduce the toxicity associated with OMV [103,104]. A recent study demonstrated that OMV and extracellular vesicles, from E. coli and S. aureus, respectively, with an antibiotic coat have in vitro and in vivo antibacterial efficacy against S. aureus. However, this study only considered the entrance of the vesicles into the infected macrophages and not into the bacteria. Nonetheless, it was proven that it is possible to use the OMV membrane as a coat for antibiotics such as vancomycin and rifampicin with a diameter of approximately 90 nm [104]. A recent study explored the same principle by using extrusion to coat rifampicin-loaded mesoporous silica nanoparticles with OMV from E. coli. Unlike the previous study, it was proven the entrance of these vesicles into E. coli bacteria but not into S. aureus [103].
A novel mechanism of repurposing antibiotics is the sonochemistry that is capable of nanotransforming vancomycin, an antibiotic used against Gram-positive bacteria, to pass through and disrupt the OMs of Gram-negative bacteria. This technique has proven to be effective against E. coli and P. aeruginosa bacteria [102].
Due to the similarity to the OM composition, natural OMV represent an advantage relative to synthetic nanoparticles, which lack the intercellular interaction’s essential ability to promote trafficking and delivery of antibiotics into bacterial cells [3]. However, the combination of the new approaches is still in its beginning and further developments and applied outcomes are expected.

8. Conclusions

OMV from Gram-negative bacteria have innate antibacterial activity due to the incorporation of several enzymes such as lysins. The incorporation of non-natural molecules into OMV with additional antibacterial effect has been shown. This suggests that a delivery system could be developed to overcome the OM barrier of the Gram-negative bacteria and that bacterial OMV can be used to deliver antibiotics to targeted populations of Gram-negative and -positive pathogenic bacteria. The OMV antimicrobial effect will depend on their cargo and the bacterial species targeted, as well as the resistance mechanism present in the target bacterial cells. The repurposing of antibiotics that are ineffective due to the permeability barrier of the cell wall of bacteria may also be possible with the use of OMV.
The industrial production of OMV can be enhanced by changing the growth conditions or through different techniques that force cell damage; an alternative to OMV production with active lumen content is coating of the OMV with nanoparticles. However, several technical challenges that hamper the use of OMV remain, such as the types of MV isolated, the purification yield, and the optimal technique to produce vesicles with desired lumen content, especially hydrophilic molecules which cannot enter by passive loading. The reduction of the LPS toxicity is another point that needs to be optimized in order to reduce the immunogenic potential of OMV; for instance, the engineering of strains with altered LPS [105] might be a solution to produce OMV with reduced cytotoxicity. There is also still the need to better understand the target populations, including the specifics of the interaction between OMV and the target as well as subsequent host cell reactions.
Overall, despite the challenges that must still be overcome, OMV represent a cost-effective and safe drug delivery tool, representing a promising alternative for the treatment of bacterial infections caused by antibiotic-resistant bacteria and for the repurposing of antibiotics that are not usually effective against Gram-negative bacteria.

Author Contributions

Conceptualization, S.C., G.J.d.S. and S.D.; writing—original draft preparation, S.C. and S.M.; writing—review and editing, K.M.N., G.J.d.S. and S.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The Faculty of Pharmacy of the University of Coimbra and Center for Neurosciences and Cell Biology through COMPETE 2020—Operational Programme for Competitiveness and Internationalisation and Portuguese national funds via FCT—Fundação para a Ciência e a Tecnologia under projects UIDB/04539/2020, UIDP/04539/2020 and LA/P/0058/2020. Sérgio Mendes acknowledges Fundação para a Ciência e a Tecnologia (FCT) for his PhD grant SFRH/BD/06289/2021.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Garaeva, L.; Kamyshinsky, R.; Kil, Y.; Varfolomeeva, E.; Verlov, N.; Komarova, E.; Garmay, Y.; Landa, S.; Burdakov, V.; Myasnikov, A.; et al. Delivery of Functional Exogenous Proteins by Plant-Derived Vesicles to Human Cells in Vitro. Sci. Rep. 2021, 11, 6489. [Google Scholar] [CrossRef]
  2. Fiocca, R.; Necchi, V.; Sommi, P.; Ricci, V.; Telford, J.; Cover, T.L.; Solcia, E. Release of Helicobacter Pylori Vacuolating Cytotoxin by Both a Specific Secretion Pathway and Budding of Outer Membrane Vesicles. Uptake of Released Toxin and Vesicles by Gastric Epithelium. J. Pathol. 1999, 188, 220–226. [Google Scholar] [CrossRef]
  3. Marchant, P.; Carreño, A.; Vivanco, E.; Silva, A.; Nevermann, J.; Otero, C.; Araya, E.; Gil, F.; Calderón, I.L.; Fuentes, J.A. “One for All”: Functional Transfer of OMV-Mediated Polymyxin B Resistance From Salmonella Enterica Sv. Typhi ΔtolR and ΔdegS to Susceptible Bacteria. Front. Microbiol. 2021, 12, 1–15. [Google Scholar] [CrossRef]
  4. Shima, T.; Muraoka, T.; Hamada, T.; Morita, M.; Takagi, M.; Fukuoka, H.; Inoue, Y.; Sagawa, T.; Ishijima, A.; Omata, Y.; et al. Micrometer-Size Vesicle Formation Triggered by UV Light. Langmuir 2014, 30, 7289–7295. [Google Scholar] [CrossRef]
  5. Redeker, C.; Briscoe, W.H. Interactions between Mutant Bacterial Lipopolysaccharide (LPS-Ra) Surface Layers: Surface Vesicles, Membrane Fusion, and Effect of Ca2+ and Temperature. Langmuir 2019, 35, 15739–15750. [Google Scholar] [CrossRef]
  6. Kadurugamuwa, J.L.; Beveridge, T.J. Virulence Factors Are Released from Pseudomonas Aeruginosa in Association with Membrane Vesicles during Normal Growth and Exposure to Gentamicin: A Novel Mechanism of Enzyme Secretion. J. Bacteriol. 1995, 177, 3998–4008. [Google Scholar] [CrossRef]
  7. Chatterjee, S.; Mondal, A.; Mitra, S.; Basu, S. Acinetobacter Baumannii Transfers the BlaNDM-1 Gene via Outer Membrane Vesicles. J. Antimicrob. Chemother. 2017, 72, 2201–2207. [Google Scholar] [CrossRef]
  8. Lin, J.; Zhang, W.; Cheng, J.; Yang, X.; Zhu, K.; Wang, Y.; Wei, G.; Qian, P.-Y.; Luo, Z.-Q.; Shen, X. A Pseudomonas T6SS Effector Recruits PQS-Containing Outer Membrane Vesicles for Iron Acquisition. Nat. Commun. 2017, 8, 14888. [Google Scholar] [CrossRef]
  9. Fuhrmann, G.; Serio, A.; Mazo, M.; Nair, R.; Stevens, M.M. Active Loading into Extracellular Vesicles Significantly Improves the Cellular Uptake and Photodynamic Effect of Porphyrins. J. Control. Release 2015, 205, 35–44. [Google Scholar] [CrossRef]
  10. Michel, L.V.; Gallardo, L.; Konovalova, A.; Bauer, M.; Jackson, N.; Zavorin, M.; McNamara, C.; Pierce, J.; Cheng, S.; Snyder, E.; et al. Ampicillin Triggers the Release of Pal in Toxic Vesicles from Escherichia Coli. Int. J. Antimicrob. Agents 2020, 56, 106163. [Google Scholar] [CrossRef]
  11. Marion, C.R.; Lee, J.; Sharma, L.; Park, K.; Lee, C.; Liu, W.; Liu, P.; Feng, J.; Gho, Y.S.; Dela Cruz, C.S. Toll-Like Receptors 2 and 4 Modulate Pulmonary Inflammation and Host Factors Mediated by Outer Membrane Vesicles Derived from Acinetobacter Baumannii. Infect. Immun. 2019, 87, e00243-19. [Google Scholar] [CrossRef] [PubMed]
  12. Kadurugamuwa, J.L.; Beveridge, T.J. Bacteriolytic Effect of Membrane Vesicles from Pseudomonas Aeruginosa on Other Bacteria Including Pathogens: Conceptually New Antibiotics. J. Bacteriol. 1996, 178, 2767–2774. [Google Scholar] [CrossRef] [PubMed]
  13. Toyofuku, M.; Nomura, N.; Eberl, L. Types and Origins of Bacterial Membrane Vesicles. Nat. Rev. Microbiol. 2019, 17, 13–24. [Google Scholar] [CrossRef] [PubMed]
  14. Domingues, S.; Nielsen, K.M. Membrane Vesicles and Horizontal Gene Transfer in Prokaryotes. Curr. Opin. Microbiol. 2017, 38, 16–21. [Google Scholar] [CrossRef] [PubMed]
  15. Domingues, S.; Rosário, N.; Ben Cheikh, H.; Da Silva, G.J. ISAba1 and Tn6168 Acquisition by Natural Transformation Leads to Third-Generation Cephalosporins Resistance in Acinetobacter Baumannii. Infect. Genet. Evol. 2018, 63, 13–16. [Google Scholar] [CrossRef] [PubMed]
  16. Park, J.; Kim, M.; Shin, B.; Kang, M.; Yang, J.; Lee, T.K.; Park, W. A Novel Decoy Strategy for Polymyxin Resistance in Acinetobacter Baumannii. Elife 2021, 10, 1–29. [Google Scholar] [CrossRef] [PubMed]
  17. Collins, S.M.; Brown, A.C. Bacterial Outer Membrane Vesicles as Antibiotic Delivery Vehicles. Front. Immunol. 2021, 12, 733064. [Google Scholar] [CrossRef]
  18. Schwechheimer, C.; Sullivan, C.J.; Kuehn, M.J. Envelope Control of Outer Membrane Vesicle Production in Gram-Negative Bacteria. Biochemistry 2013, 52, 3031–3040. [Google Scholar] [CrossRef]
  19. Avila-Calderón, E.D.; Ruiz-Palma, M.D.S.; Aguilera-Arreola, M.G.; Velázquez-Guadarrama, N.; Ruiz, E.A.; Gomez-Lunar, Z.; Witonsky, S.; Contreras-Rodríguez, A. Outer Membrane Vesicles of Gram-Negative Bacteria: An Outlook on Biogenesis. Front. Microbiol. 2021, 12, 557902. [Google Scholar] [CrossRef]
  20. Lee, E.-Y.; Bang, J.Y.; Park, G.W.; Choi, D.-S.; Kang, J.S.; Kim, H.-J.; Park, K.-S.; Lee, J.-O.; Kim, Y.-K.; Kwon, K.-H.; et al. Global Proteomic Profiling of Native Outer Membrane Vesicles Derived from Escherichia Coli. Proteomics 2007, 7, 3143–3153. [Google Scholar] [CrossRef]
  21. Hoekstra, D.; van der Laan, J.W.; de Leij, L.; Witholt, B. Release of Outer Membrane Fragments from Normally Growing Escherichia Coli. Biochim. Biophys. Acta 1976, 455, 889–899. [Google Scholar] [CrossRef]
  22. Wensink, J.; Witholt, B. Outer-Membrane Vesicles Released by Normally Growing Escherichia Coli Contain Very Little Lipoprotein. Eur. J. Biochem. 1981, 116, 331–335. [Google Scholar] [CrossRef] [PubMed]
  23. Deatherage, B.L.; Lara, J.C.; Bergsbaken, T.; Rassoulian Barrett, S.L.; Lara, S.; Cookson, B.T. Biogenesis of Bacterial Membrane Vesicles. Mol. Microbiol. 2009, 72, 1395–1407. [Google Scholar] [CrossRef] [PubMed]
  24. Hayashi, J.; Hamada, N.; Kuramitsu, H.K. The Autolysin of Porphyromonas Gingivalis Is Involved in Outer Membrane Vesicle Release. FEMS Microbiol. Lett. 2002, 216, 217–222. [Google Scholar] [CrossRef]
  25. Zhou, L.; Srisatjaluk, R.; Justus, D.E.; Doyle, R.J. On the Origin of Membrane Vesicles in Gram-Negative Bacteria. FEMS Microbiol. Lett. 1998, 163, 223–228. [Google Scholar] [CrossRef]
  26. Moon, D.C.; Choi, C.H.; Lee, J.H.; Choi, C.-W.; Kim, H.-Y.; Park, J.S.; Kim, S.I.; Lee, J.C. Acinetobacter Baumannii Outer Membrane Protein A Modulates the Biogenesis of Outer Membrane Vesicles. J. Microbiol. 2012, 50, 155–160. [Google Scholar] [CrossRef]
  27. Roier, S.; Zingl, F.G.; Cakar, F.; Durakovic, S.; Kohl, P.; Eichmann, T.O.; Klug, L.; Gadermaier, B.; Weinzerl, K.; Prassl, R.; et al. A Novel Mechanism for the Biogenesis of Outer Membrane Vesicles in Gram-Negative Bacteria. Nat. Commun. 2016, 7, 10515:1–10515:13. [Google Scholar] [CrossRef]
  28. Pettit, R.K.; Judd, R.C. The Interaction of Naturally Elaborated Blebs from Serum-Susceptible and Serum-Resistant Strains of Neisseria Gonorrhoeae with Normal Human Serum. Mol. Microbiol. 1992, 6, 729–734. [Google Scholar] [CrossRef]
  29. Sampath, V.; McCaig, W.D.; Thanassi, D.G. Amino Acid Deprivation and Central Carbon Metabolism Regulate the Production of Outer Membrane Vesicles and Tubes by Francisella. Mol. Microbiol. 2018, 107, 523–541. [Google Scholar] [CrossRef]
  30. Loeb, M.R.; Kilner, J. Effect of Growth Medium on the Relative Polypeptide Composition of Cellular Outer Membrane and Released Outer Membrane Material in Escherichia Coli. J. Bacteriol. 1979, 137, 1031–1034. [Google Scholar] [CrossRef] [Green Version]
  31. Nakamura, S.; Shchepetov, M.; Dalia, A.B.; Clark, S.E.; Murphy, T.F.; Sethi, S.; Gilsdorf, J.R.; Smith, A.L.; Weiser, J.N. Molecular Basis of Increased Serum Resistance among Pulmonary Isolates of Non-Typeable Haemophilus Influenzae. PLoS Pathog. 2011, 7, e1001247. [Google Scholar] [CrossRef]
  32. de Jonge, E.F.; Balhuizen, M.D.; van Boxtel, R.; Wu, J.; Haagsman, H.P.; Tommassen, J. Heat Shock Enhances Outer-Membrane Vesicle Release in Bordetella spp. Curr. Res. Microb. Sci. 2021, 2, 100009. [Google Scholar] [CrossRef] [PubMed]
  33. McBroom, A.J.; Kuehn, M.J. Release of Outer Membrane Vesicles by Gram-Negative Bacteria Is a Novel Envelope Stress Response. Mol. Microbiol. 2007, 63, 545–558. [Google Scholar] [CrossRef]
  34. Weaver, J.C. Electroporation Theory. Concepts and Mechanisms. Methods Mol. Biol. 1995, 47, 1–26. [Google Scholar] [CrossRef] [PubMed]
  35. Gujrati, V.; Kim, S.; Kim, S.-H.; Min, J.J.; Choy, H.E.; Kim, S.C.; Jon, S. Bioengineered Bacterial Outer Membrane Vesicles as Cell-Specific Drug-Delivery Vehicles for Cancer Therapy. ACS Nano 2014, 8, 1525–1537. [Google Scholar] [CrossRef] [PubMed]
  36. Pick, U. Liposomes with a Large Trapping Capacity Prepared by Freezing and Thawing of Sonicated Phospholipid Mixtures. Arch. Biochem. Biophys. 1981, 212, 186–194. [Google Scholar] [CrossRef]
  37. Lamichhane, T.N.; Jeyaram, A.; Patel, D.B.; Parajuli, B.; Livingston, N.K.; Arumugasaamy, N.; Schardt, J.S.; Jay, S.M. Oncogene Knockdown via Active Loading of Small RNAs into Extracellular Vesicles by Sonication. Cell. Mol. Bioeng. 2016, 9, 315–324. [Google Scholar] [CrossRef]
  38. Zhang, Y.-F.; Shi, J.-B.; Li, C. Small Extracellular Vesicle Loading Systems in Cancer Therapy: Current Status and the Way Forward. Cytotherapy 2019, 21, 1122–1136. [Google Scholar] [CrossRef] [PubMed]
  39. Gayán, E.; Govers, S.K.; Aertsen, A. Impact of High Hydrostatic Pressure on Bacterial Proteostasis. Biophys. Chem. 2017, 231, 3–9. [Google Scholar] [CrossRef] [PubMed]
  40. Zwarycz, A.S.; Livingstone, P.G.; Whitworth, D.E. Within-Species Variation in OMV Cargo Proteins: The Myxococcus Xanthus OMV Pan-Proteome. Mol. Omi. 2020, 16, 387–397. [Google Scholar] [CrossRef] [PubMed]
  41. Choi, C.H.; Lee, E.Y.; Lee, Y.C.; Park, T.I.; Kim, H.J.; Hyun, S.H.; Kim, S.A.; Lee, S.-K.; Lee, J.C. Outer Membrane Protein 38 of Acinetobacter Baumannii Localizes to the Mitochondria and Induces Apoptosis of Epithelial Cells. Cell. Microbiol. 2005, 7, 1127–1138. [Google Scholar] [CrossRef] [PubMed]
  42. Choi, C.H.; Hyun, S.H.; Lee, J.Y.; Lee, J.S.; Lee, Y.S.; Kim, S.A.; Chae, J.-P.; Yoo, S.M.; Lee, J.C. Acinetobacter Baumannii Outer Membrane Protein A Targets the Nucleus and Induces Cytotoxicity. Cell. Microbiol. 2008, 10, 309–319. [Google Scholar] [CrossRef] [PubMed]
  43. Jin, J.S.; Kwon, S.-O.; Moon, D.C.; Gurung, M.; Lee, J.H.; Kim, S.I.; Lee, J.C. Acinetobacter Baumannii Secretes Cytotoxic Outer Membrane Protein A via Outer Membrane Vesicles. PLoS ONE 2011, 6, e17027. [Google Scholar] [CrossRef] [PubMed]
  44. Bomberger, J.M.; Maceachran, D.P.; Coutermarsh, B.A.; Ye, S.; O’Toole, G.A.; Stanton, B.A. Long-Distance Delivery of Bacterial Virulence Factors by Pseudomonas Aeruginosa Outer Membrane Vesicles. PLoS Pathog. 2009, 5, e1000382. [Google Scholar] [CrossRef]
  45. Kolling, G.L.; Matthews, K.R. Export of Virulence Genes and Shiga Toxin by Membrane Vesicles of Escherichia Coli O157:H7. Appl. Environ. Microbiol. 1999, 65, 1843–1848. [Google Scholar] [CrossRef] [PubMed]
  46. Pérez, A.; Merino, M.; Rumbo-Feal, S.; Álvarez-Fraga, L.; Vallejo, J.A.; Beceiro, A.; Ohneck, E.J.; Mateos, J.; Fernández-Puente, P.; Actis, L.A.; et al. The FhaB/FhaC Two-Partner Secretion System Is Involved in Adhesion of Acinetobacter Baumannii AbH12O-A2 Strain. Virulence 2017, 8, 959–974. [Google Scholar] [CrossRef]
  47. Darvish Alipour Astaneh, S.; Rasooli, I.; Mousavi Gargari, S.L. The Role of Filamentous Hemagglutinin Adhesin in Adherence and Biofilm Formation in Acinetobacter Baumannii ATCC19606(T). Microb. Pathog. 2014, 74, 42–49. [Google Scholar] [CrossRef] [PubMed]
  48. Koeppen, K.; Hampton, T.H.; Jarek, M.; Scharfe, M.; Gerber, S.A.; Mielcarz, D.W.; Demers, E.G.; Dolben, E.L.; Hammond, J.H.; Hogan, D.A.; et al. A Novel Mechanism of Host-Pathogen Interaction through SRNA in Bacterial Outer Membrane Vesicles. PLoS Pathog. 2016, 12, e1005672. [Google Scholar] [CrossRef]
  49. Roszkowiak, J.; Jajor, P.; Guła, G.; Gubernator, J.; Żak, A.; Drulis-Kawa, Z.; Augustyniak, D. Interspecies Outer Membrane Vesicles (OMVs) Modulate the Sensitivity of Pathogenic Bacteria and Pathogenic Yeasts to Cationic Peptides and Serum Complement. Int. J. Mol. Sci. 2019, 20, 5577. [Google Scholar] [CrossRef]
  50. Dell’Annunziata, F.; Dell’Aversana, C.; Doti, N.; Donadio, G.; Dal Piaz, F.; Izzo, V.; De Filippis, A.; Galdiero, M.; Altucci, L.; Boccia, G.; et al. Outer Membrane Vesicles Derived from Klebsiella Pneumoniae Are a Driving Force for Horizontal Gene Transfer. Int. J. Mol. Sci. 2021, 22, 8732. [Google Scholar] [CrossRef]
  51. Bielaszewska, M.; Daniel, O.; Karch, H.; Mellmann, A. Dissemination of the BlaCTX-M-15 Gene among Enterobacteriaceae via Outer Membrane Vesicles. J. Antimicrob. Chemother. 2020, 75, 2442–2451. [Google Scholar] [CrossRef]
  52. Gasperini, G.; Arato, V.; Pizza, M.; Aricò, B.; Leuzzi, R. Physiopathological Roles of Spontaneously Released Outer Membrane Vesicles of Bordetella Pertussis. Future Microbiol. 2017, 12, 1247–1259. [Google Scholar] [CrossRef]
  53. Feitosa-Junior, O.R.; Stefanello, E.; Zaini, P.A.; Nascimento, R.; Pierry, P.M.; Dandekar, A.M.; Lindow, S.E.; da Silva, A.M. Proteomic and Metabolomic Analyses of Xylella Fastidiosa OMV-Enriched Fractions Reveal Association with Virulence Factors and Signaling Molecules of the DSF Family. Phytopathology 2019, 109, 1344–1353. [Google Scholar] [CrossRef]
  54. Chen, S.; Yang, D.; Wen, Y.; Jiang, Z.; Zhang, L.; Jiang, J.; Chen, Y.; Hu, T.; Wang, Q.; Zhang, Y.; et al. Dysregulated Hemolysin Liberates Bacterial Outer Membrane Vesicles for Cytosolic Lipopolysaccharide Sensing. PLoS Pathog. 2018, 14, e1007240. [Google Scholar] [CrossRef] [PubMed]
  55. Mashburn, L.M.; Whiteley, M. Membrane Vesicles Traffic Signals and Facilitate Group Activities in a Prokaryote. Nature 2005, 437, 422–425. [Google Scholar] [CrossRef]
  56. Piddock, L.J.V.; Wise, R. Induction of the SOS Response in Escherichia Coli by 4-Quinolone Antimicrobial Agents. FEMS Microbiol. Lett. 1987, 41, 289–294. [Google Scholar] [CrossRef]
  57. Devos, S.; Van Putte, W.; Vitse, J.; Van Driessche, G.; Stremersch, S.; Van Den Broek, W.; Raemdonck, K.; Braeckmans, K.; Stahlberg, H.; Kudryashev, M.; et al. Membrane Vesicle Secretion and Prophage Induction in Multidrug-Resistant Stenotrophomonas Maltophilia in Response to Ciprofloxacin Stress. Environ. Microbiol. 2017, 19, 3930–3937. [Google Scholar] [CrossRef]
  58. McMahon, K.J.; Castelli, M.E.; García Vescovi, E.; Feldman, M.F. Biogenesis of Outer Membrane Vesicles in Serratia Marcescens Is Thermoregulated and Can Be Induced by Activation of the Rcs Phosphorelay System. J. Bacteriol. 2012, 194, 3241–3249. [Google Scholar] [CrossRef]
  59. Macdonald, I.A.; Kuehn, M.J. Stress-Induced Outer Membrane Vesicle Production by Pseudomonas Aeruginosa. J. Bacteriol. 2013, 195, 2971–2981. [Google Scholar] [CrossRef]
  60. Spiess, C.; Beil, A.; Ehrmann, M. A Temperature-Dependent Switch from Chaperone to Protease in a Widely Conserved Heat Shock Protein. Cell 1999, 97, 339–347. [Google Scholar] [CrossRef] [Green Version]
  61. Wood, L.F.; Ohman, D.E. Independent Regulation of MucD, an HtrA-like Protease in Pseudomonas Aeruginosa, and the Role of Its Proteolytic Motif in Alginate Gene Regulation. J. Bacteriol. 2006, 188, 3134–3137. [Google Scholar] [CrossRef] [PubMed]
  62. van de Waterbeemd, B.; Zomer, G.; Kaaijk, P.; Ruiterkamp, N.; Wijffels, R.H.; van den Dobbelsteen, G.P.J.M.; van der Pol, L.A. Improved Production Process for Native Outer Membrane Vesicle Vaccine against Neisseria Meningitidis. PLoS ONE 2013, 8, e65157. [Google Scholar] [CrossRef]
  63. Estrela, C.; Costa E Silva, R.; Urban, R.C.; Gonçalves, P.J.; Silva, J.A.; Estrela, C.R.A.; Pecora, J.D.; Peters, O.A. Demetallization of Enterococcus Faecalis Biofilm: A Preliminary Study. J. Appl. Oral Sci. 2018, 26, e20170374. [Google Scholar] [CrossRef] [PubMed]
  64. Li, Z.; Clarke, A.J.; Beveridge, T.J. A Major Autolysin of Pseudomonas Aeruginosa: Subcellular Distribution, Potential Role in Cell Growth and Division and Secretion in Surface Membrane Vesicles. J. Bacteriol. 1996, 178, 2479–2488. [Google Scholar] [CrossRef] [PubMed]
  65. Evans, A.G.L.; Davey, H.M.; Cookson, A.; Currinn, H.; Cooke-Fox, G.; Stanczyk, P.J.; Whitworth, D.E. Predatory Activity of Myxococcus Xanthus Outer-Membrane Vesicles and Properties of Their Hydrolase Cargo. Microbiology 2012, 158, 2742–2752. [Google Scholar] [CrossRef] [PubMed]
  66. HOSODA, J.; NOMURA, M. Nature of the Primary Action of the Autolysin of Bacillus Subtilis. J. Bacteriol. 1956, 72, 573–581. [Google Scholar] [CrossRef]
  67. Ghuysen, J.M. Use of Bacteriolytic Enzymes in Determination of Wall Structure and Their Role in Cell Metabolism. Bacteriol. Rev. 1968, 32, 425–464. [Google Scholar] [CrossRef] [PubMed]
  68. Oshida, T.; Sugai, M.; Komatsuzawa, H.; Hong, Y.M.; Suginaka, H.; Tomasz, A. A Staphylococcus Aureus Autolysin That Has an N-Acetylmuramoyl-L-Alanine Amidase Domain and an Endo-Beta-N-Acetylglucosaminidase Domain: Cloning, Sequence Analysis, and Characterization. Proc. Natl. Acad. Sci. USA 1995, 92, 285–289. [Google Scholar] [CrossRef] [PubMed]
  69. Hodak, H.; Galán, J.E. A Salmonella Typhi Homologue of Bacteriophage Muramidases Controls Typhoid Toxin Secretion. EMBO Rep. 2013, 14, 95–102. [Google Scholar] [CrossRef]
  70. Scheurwater, E.; Reid, C.W.; Clarke, A.J. Lytic Transglycosylases: Bacterial Space-Making Autolysins. Int. J. Biochem. Cell Biol. 2008, 40, 586–591. [Google Scholar] [CrossRef] [Green Version]
  71. Ishida, S.T.; Ishida, T. Zinc-Induced Immune Anti-Bacterial Vaccine Activity in Bacterial Cell Walls against Gram-Positive and Gram-Negative Bacteria. Proc. ARC J. Immunol. Vaccines 2019, 4, 10–19. [Google Scholar]
  72. Yamada, S.; Sugai, M.; Komatsuzawa, H.; Nakashima, S.; Oshida, T.; Matsumoto, A.; Suginaka, H. An Autolysin Ring Associated with Cell Separation of Staphylococcus Aureus. J. Bacteriol. 1996, 178, 1565–1571. [Google Scholar] [CrossRef] [PubMed]
  73. Li, Z.; Clarke, A.J.; Beveridge, T.J. Gram-Negative Bacteria Produce Membrane Vesicles Which Are Capable of Killing Other Bacteria. J. Bacteriol. 1998, 180, 5478–5483. [Google Scholar] [CrossRef] [PubMed]
  74. Kudryakova, I.V.; Suzina, N.E.; Vasilyeva, N.V. Biogenesis of Lysobacter Sp. XL1 Vesicles. FEMS Microbiol. Lett. 2015, 362, fnv137. [Google Scholar] [CrossRef] [PubMed]
  75. Kadurugamuwa, J.L.; Beveridge, T.J. Membrane Vesicles Derived from Pseudomonas Aeruginosa and Shigella Flexneri Can Be Integrated into the Surfaces of Other Gram-Negative Bacteria. Microbiology 1999, 145, 2051–2060. [Google Scholar] [CrossRef] [PubMed]
  76. Kadurugamuwa, J.L.; Mayer, A.; Messner, P.; Sára, M.; Sleytr, U.B.; Beveridge, T.J. S-Layered Aneurinibacillus and Bacillus Spp. Are Susceptible to the Lytic Action of Pseudomonas Aeruginosa Membrane Vesicles. J. Bacteriol. 1998, 180, 2306–2311. [Google Scholar] [CrossRef]
  77. Hood, R.D.; Singh, P.; Hsu, F.; Güvener, T.; Carl, M.A.; Trinidad, R.R.S.; Silverman, J.M.; Ohlson, B.B.; Hicks, K.G.; Plemel, R.L.; et al. A Type VI Secretion System of Pseudomonas Aeruginosa Targets a Toxin to Bacteria. Cell Host Microbe 2010, 7, 25–37. [Google Scholar] [CrossRef]
  78. Russell, A.B.; Hood, R.D.; Bui, N.K.; LeRoux, M.; Vollmer, W.; Mougous, J.D. Type VI Secretion Delivers Bacteriolytic Effectors to Target Cells. Nature 2011, 475, 343–347. [Google Scholar] [CrossRef]
  79. Hashem, K.A.; Authman, S.H.; Hameed, L. In Vivo Antibacterial Activity of Alkaline Phosphatase Isolates from Escherichia Coli Isolated from Diarrhea Patients against Pseudomonas Aeruginosa. Pharma Innov. J. 2016, 5, 32–34. [Google Scholar]
  80. Nikaido, H.; Vaara, M. Molecular Basis of Bacterial Outer Membrane Permeability. Microbiol. Rev. 1985, 49, 1–32. [Google Scholar] [CrossRef]
  81. Blokhina, S.V.; Sharapova, A.V.; Ol’khovich, M.V.; Volkova, T.V.; Perlovich, G.L. Solubility, Lipophilicity and Membrane Permeability of Some Fluoroquinolone Antimicrobials. Eur. J. Pharm. Sci. 2016, 93, 29–37. [Google Scholar] [CrossRef] [PubMed]
  82. Huang, W.; Zhang, Q.; Li, W.; Yuan, M.; Zhou, J.; Hua, L.; Chen, Y.; Ye, C.; Ma, Y. Development of Novel Nanoantibiotics Using an Outer Membrane Vesicle-Based Drug Efflux Mechanism. J. Control. Release 2020, 317, 1–22. [Google Scholar] [CrossRef] [PubMed]
  83. Stankevic, V.; Simonis, P.; Zurauskiene, N.; Stirke, A.; Dervinis, A.; Bleizgys, V.; Kersulis, S.; Balevicius, S. Compact Square-Wave Pulse Electroporator with Controlled Electroporation Efficiency and Cell Viability. Symmetry 2020, 12, 412. [Google Scholar] [CrossRef]
  84. Ayed, Z.; Cuvillier, L.; Dobhal, G.; Goreham, R.V. Electroporation of Outer Membrane Vesicles Derived from Pseudomonas Aeruginosa with Gold Nanoparticles. SN Appl. Sci. 2019, 1, 1600. [Google Scholar] [CrossRef]
  85. Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of Exosome-Encapsulated Paclitaxel to Overcome MDR in Cancer Cells. Nanomedicine 2016, 12, 655–664. [Google Scholar] [CrossRef] [PubMed]
  86. McCaig, W.D.; Loving, C.L.; Hughes, H.R.; Brockmeier, S.L. Characterization and Vaccine Potential of Outer Membrane Vesicles Produced by Haemophilus Parasuis. PLoS ONE 2016, 11, e0149132. [Google Scholar] [CrossRef] [PubMed]
  87. Li, K.; Yuan, X.-X.; Sun, H.-M.; Zhao, L.-S.; Tang, R.; Chen, Z.-H.; Qin, Q.-L.; Chen, X.-L.; Zhang, Y.-Z.; Su, H.-N. Atomic Force Microscopy of Side Wall and Septa Peptidoglycan From Bacillus Subtilis Reveals an Architectural Remodeling During Growth. Front. Microbiol. 2018, 9, 620. [Google Scholar] [CrossRef]
  88. Foladori, P.; Laura, B.; Gianni, A.; Giuliano, Z. Effects of Sonication on Bacteria Viability in Wastewater Treatment Plants Evaluated by Flow Cytometry--Fecal Indicators, Wastewater and Activated Sludge. Water Res. 2007, 41, 235–243. [Google Scholar] [CrossRef]
  89. Gomes, A.; van Oosten, M.; Bijker, K.L.B.; Boiten, K.E.; Salomon, E.N.; Rosema, S.; Rossen, J.W.A.; Natour, E.; Douglas, Y.L.; Kampinga, G.A.; et al. Sonication of Heart Valves Detects More Bacteria in Infective Endocarditis. Sci. Rep. 2018, 8, 12967. [Google Scholar] [CrossRef] [PubMed]
  90. Gao, W.; Fang, R.H.; Thamphiwatana, S.; Luk, B.T.; Li, J.; Angsantikul, P.; Zhang, Q.; Hu, C.-M.J.; Zhang, L. Modulating Antibacterial Immunity via Bacterial Membrane-Coated Nanoparticles. Nano Lett. 2015, 15, 1403–1409. [Google Scholar] [CrossRef] [PubMed]
  91. Shi, J.; Ma, Z.; Pan, H.; Liu, Y.; Chu, Y.; Wang, J.; Chen, L. Biofilm-Encapsulated Nano Drug Delivery System for the Treatment of Colon Cancer. J. Microencapsul. 2020, 37, 481–491. [Google Scholar] [CrossRef] [PubMed]
  92. Genova, L.A.; Roberts, M.F.; Wong, Y.-C.; Harper, C.E.; Santiago, A.G.; Fu, B.; Srivastava, A.; Jung, W.; Wang, L.M.; Krzemiński, Ł.; et al. Mechanical Stress Compromises Multicomponent Efflux Complexes in Bacteria. Proc. Natl. Acad. Sci. USA 2019, 116, 25462–25467. [Google Scholar] [CrossRef] [PubMed]
  93. Sato, Y.T.; Umezaki, K.; Sawada, S.; Mukai, S.; Sasaki, Y.; Harada, N.; Shiku, H.; Akiyoshi, K. Engineering Hybrid Exosomes by Membrane Fusion with Liposomes. Sci. Rep. 2016, 6, 21933. [Google Scholar] [CrossRef] [PubMed]
  94. Sáenz, J.P.; Grosser, D.; Bradley, A.S.; Lagny, T.J.; Lavrynenko, O.; Broda, M.; Simons, K. Hopanoids as Functional Analogues of Cholesterol in Bacterial Membranes. Proc. Natl. Acad. Sci. USA 2015, 112, 11971–11976. [Google Scholar] [CrossRef] [PubMed]
  95. Mondal Roy, S.; Sarkar, M. Membrane Fusion Induced by Small Molecules and Ions. J. Lipids 2011, 2011, 528784. [Google Scholar] [CrossRef]
  96. Martens, S.; McMahon, H.T. Mechanisms of Membrane Fusion: Disparate Players and Common Principles. Nat. Rev. Mol. Cell Biol. 2008, 9, 543–556. [Google Scholar] [CrossRef]
  97. Hamai, C.; Yang, T.; Kataoka, S.; Cremer, P.S.; Musser, S.M. Effect of Average Phospholipid Curvature on Supported Bilayer Formation on Glass by Vesicle Fusion. Biophys. J. 2006, 90, 1241–1248. [Google Scholar] [CrossRef] [PubMed]
  98. Gnopo, Y.M.D.; Misra, A.; Hsu, H.-L.; DeLisa, M.P.; Daniel, S.; Putnam, D. Induced Fusion and Aggregation of Bacterial Outer Membrane Vesicles: Experimental and Theoretical Analysis. J. Colloid Interface Sci. 2020, 578, 522–532. [Google Scholar] [CrossRef] [PubMed]
  99. Schulz, E.; Goes, A.; Garcia, R.; Panter, F.; Koch, M.; Müller, R.; Fuhrmann, K.; Fuhrmann, G. Biocompatible Bacteria-Derived Vesicles Show Inherent Antimicrobial Activity. J. Control. Release 2018, 290, 46–55. [Google Scholar] [CrossRef] [PubMed]
  100. Remis, J.P.; Wei, D.; Gorur, A.; Zemla, M.; Haraga, J.; Allen, S.; Witkowska, H.E.; Costerton, J.W.; Berleman, J.E.; Auer, M. Bacterial Social Networks: Structure and Composition of Myxococcus Xanthus Outer Membrane Vesicle Chains. Environ. Microbiol. 2014, 16, 598–610. [Google Scholar] [CrossRef]
  101. Naskar, A.; Cho, H.; Lee, S.; Kim, K. Biomimetic Nanoparticles Coated with Bacterial Outer Membrane Vesicles as a New-Generation Platform for Biomedical Applications. Pharmaceutics 2021, 13, 1887. [Google Scholar] [CrossRef] [PubMed]
  102. Fernandes, M.M.; Ivanova, K.; Hoyo, J.; Pérez-Rafael, S.; Francesko, A.; Tzanov, T. Nanotransformation of Vancomycin Overcomes the Intrinsic Resistance of Gram-Negative Bacteria. ACS Appl. Mater. Interfaces 2017, 9, 15022–15030. [Google Scholar] [CrossRef]
  103. Wu, S.; Huang, Y.; Yan, J.; Li, Y.; Wang, J.; Yang, Y.Y.; Yuan, P.; Ding, X. Bacterial Outer Membrane-Coated Mesoporous Silica Nanoparticles for Targeted Delivery of Antibiotic Rifampicin against Gram-Negative Bacterial Infection In Vivo. Adv. Funct. Mater. 2021, 31, 2103442. [Google Scholar] [CrossRef]
  104. Gao, F.; Xu, L.; Yang, B.; Fan, F.; Yang, L. Kill the Real with the Fake: Eliminate Intracellular Staphylococcus Aureus Using Nanoparticle Coated with Its Extracellular Vesicle Membrane as Active-Targeting Drug Carrier. ACS Infect. Dis. 2019, 5, 218–227. [Google Scholar] [CrossRef] [PubMed]
  105. Sharif, E.; Eftekhari, Z.; Mohit, E. The Effect of Growth Stage and Isolation Method on Properties of ClearColiTM Outer Membrane Vesicles (OMVs). Curr. Microbiol. 2021, 78, 1602–1614. [Google Scholar] [CrossRef]
Figure 1. Models of OMV biogenesis (a) Vesiculation caused by the absence of lipoprotein links. (b) Vesiculation caused by accumulation of misfolded peptidoglycan. (c) Vesiculation caused by negatively charged lipopolysaccharide.
Figure 1. Models of OMV biogenesis (a) Vesiculation caused by the absence of lipoprotein links. (b) Vesiculation caused by accumulation of misfolded peptidoglycan. (c) Vesiculation caused by negatively charged lipopolysaccharide.
Biomedicines 10 02399 g001
Figure 2. Merging of OMV with bacterial membrane with the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) released by M. xanthus as a signal molecule.
Figure 2. Merging of OMV with bacterial membrane with the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) released by M. xanthus as a signal molecule.
Biomedicines 10 02399 g002
Table 1. Functions of OMV according to their composition.
Table 1. Functions of OMV according to their composition.
FunctionSpeciesActive Factor/Molecular StructureHowHost/TargetEffectReference
Secretion of Toxins and Other Virulence FactorsA. baumanniiOuter membrane protein A (OMPA)Regulating the induction of cell death in hostMitochondria and nucleus from host cells Cytotoxicity activity[41,42,43]
P. aeruginosaHemolytic phospholipase C (Cif virulence factor)Released directly into cytoplasmAirway epithelial cellsCytotoxicity activity[44]
Helicobacter pyloriVacuolating toxin (VacA)OMV enter the gastric mucosa and binds to MKN28 cellsMKN28 cellsCytotoxicity activity[2]
E. coliShiga toxin 1 and 2[45]
E. coliPeptidoglycan-associated LPPRelease of peptidoglycan-associated LPP from OMV, enhanced by ampicillin[10]
Adhesion and BiofilmsA. baumanniiAbFhaB/FhaC systemFhaC protein transports AbFhaB exoprotein to the bacterial surfaceA. baumannii[46,47]
Invasion of HostP. aeruginosaSmall RNA (sRNA)Attenuation of IL–8 secretion and neutrophil infiltrationHost immune systemReduction of host innate immune response[48]
ModulationA. baumanniiLPSMediation of toll like receptors (TLRs) like TLR4 and TLR2 in macrophages that release chemokines and cytokines to recruit neutrophilsMice macrophagesInduction of pulmonary inflammatory reaction[11]
Mechanism of ResistanceMoraxella catarrhalisOMV membraneTrapping azoles even in the presence of other antibiotic that increase its actionBacteria and fungus like Candida albicansDefence against combined antibiotics [49]
A. baumannii
Salmonella Typhi
OMV membraneAct as a decoy for antibiotics, Polymixin BProtection of bacterial cells[3,16]
Gene TransferE. colieae, stx1 and stx2, and uidA genesNon-competent E. coli Resistance to β-galactams[45]
K. pneumoniaePlasmid with resistant gene to β-lactamsBurkholderia cepacia, E. coli, P. aeruginosa and Salmonella entericaResistance to β-lactams[50]
Salmonella TyphiResistance gene to Polymixin BResistance to Polymixin B, OMV as a decoy in cocultures[3]
E. coliblaCTX-M-15 gene on pESBL plasmidEnterobacteriaceae[51]
A. baumanniiblaNDM-1 gene on plasmidA. baumannii and E. coli[7]
Acquisition of NutrientsP. aeruginosaT6SS substrate TseF (Type VI secretion system effector for Fe uptake) Incorporation of T6SS substrate TseF into OMV by reacting with iron binding PQS moleculeIron acquisition[8]
Bordetella pertussisIron receptors and iron binding proteinsIron acquisition[52]
SignallingXylella fastidiosaDiffuse signalling factor 2 (DSF2)Regulation of expression of virulence and pathogenic determinantsXylella fastidiosaVirulence and pathogenicity[53]
Bacterial Mortality/CompetitionP. aeruginosaPeptidoglycan hydrolases (autolysins)P. aeruginosa resistant to gentamicinAntibacterial effect [12]
P. aeruginosa and E. coliHemolysinsCo-regulation of protein secretionAntibacterial effect[6,54]
P. aeruginosaQuinolinesStaphylococcus epidermidisAntibacterial effect[55]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Combo, S.; Mendes, S.; Nielsen, K.M.; da Silva, G.J.; Domingues, S. The Discovery of the Role of Outer Membrane Vesicles against Bacteria. Biomedicines 2022, 10, 2399. https://doi.org/10.3390/biomedicines10102399

AMA Style

Combo S, Mendes S, Nielsen KM, da Silva GJ, Domingues S. The Discovery of the Role of Outer Membrane Vesicles against Bacteria. Biomedicines. 2022; 10(10):2399. https://doi.org/10.3390/biomedicines10102399

Chicago/Turabian Style

Combo, Sofia, Sérgio Mendes, Kaare Magne Nielsen, Gabriela Jorge da Silva, and Sara Domingues. 2022. "The Discovery of the Role of Outer Membrane Vesicles against Bacteria" Biomedicines 10, no. 10: 2399. https://doi.org/10.3390/biomedicines10102399

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop