Abstract
Bacteria constitutively produce extracellular vesicles (EVs), which play a pivotal role in linking bacterial physiology to therapeutics. Despite significant advances over the past decade, critical challenges persist in isolating EVs due to the absence of standardized purification methods. While the International Society for Extracellular Vesicles provides well-defined minimal guidelines for human EVs, bacterial EV research still faces inconsistencies across laboratories, limiting reproducibility and broader utility. This review provides an in-depth overview of technical aspects of bacterial EV purification and gives future perspectives for achieving high-purity and reproducibility when isolating EVs from diverse bacterial species. It further addresses the necessity of distinguishing bacterial EVs from host-derived vesicles and potential contaminants, particularly in clinical and microbiota contexts. Establishing gold-standard protocols and defining bona fide bacterial EVs in parallel with the rigor seen in human EV studies will advance our understanding of their roles in host-microbe interactions, microbiota dysbiosis, and therapeutic potential.
Keywords: Extracellular vesicle, bacteria, outer membrane vesicle, microbiota, purification
Introduction
In living organisms, almost all eukaryotic cells produce extracellular vesicles (EVs) for intercellular communication [1]. Bacteria also release EVs during their normal growth, characterized by nanoscale, membrane-encapsulated structures [2-4]. Although bacterial and eukaryotic EVs share some physicochemical properties (e.g., size, density, charge), bacterial EVs exhibit unique heterogeneity and distinct structural features defined by size and composition closely related to their biogenesis pathways [3]. Unlike eukaryotic EVs enriched in sphingomyelin and canonical markers such as CD63 [5, 6], bacterial EVs display pathogen-associated molecular pattern (PAMP)-rich membranes and biogenesis-dependent cargo including proteins and nucleic acids [3, 7]. These differences in membrane composition, cargo, and biogenesis necessitate bacteria-specific purification and quality control (QC) parameters.
Historically, bacterial EV research has focused predominantly on Gram-negative bacteria known to produce outer membrane vesicles (OMVs), whereas their Gram-positive counterparts were long thought unable to generate EVs as part of their normal physiology [2]. However, the discovery and characterization of Gram-positive bacterial EVs, especially from Staphylococcus aureus, bridged critical gaps in our understanding of EV evolution and demonstrated the universality of EV production across bacterial species [8]. Studies on EV production have since been extended to other species, including mycobacteria [9], mycoplasma [10], and yeast [11], highlighting the natural shedding of EVs across microbes. Moreover, the widespread use of anaerobic chambers in laboratory settings has provided low-oxygen conditions that mimic the host gut environment, enabling the isolation of EVs from anaerobic bacteria, including commensal species such as Akkermansia muciniphila [12, 13] and various Bacteroidetes strains [14-16]. These advancements have broadened our understanding of bacterial EVs, particularly their emerging roles in human microbiota, host-microbe interactions, and their potential applications as biomarkers in diagnostics and therapeutics.
Despite advances in the field of bacterial EVs, comprehensive guidelines for their purification methods are still lacking. Bacterial EVs can vary in type, function, and molecular cargo depending on the species, strain, growth conditions, and biogenesis [17, 18]. Therefore, differentiating EV subtypes such as OMVs, cytoplasmic membrane vesicles (CMVs), vesicles with distinct biogenetic origins remains a technical challenge. Inconsistencies in purification protocols across laboratories can result in substantial differences of EV yield, purity, and function [19]. While Gram-negative and Gram-positive EVs originate from distinct biogenesis mechanisms, similar purification methods are often applied to both [20, 21]. Early studies on Gram-positive bacterial EVs have adapted protocols originally designed for Gram-negative OMVs, raising concerns about their suitability due to their differences in size, content, density, and other physicochemical properties [8, 22, 23]. These differences might also lead to selective loss of Gram-positive EV subpopulations when general protocols for Gram-negative bacteria or eukaryotes are applied [17]. To address these issues, optimizing growth phase, buffer composition, and centrifugal parameters should be considered to enhance Gram-positive EV yield and purity. Particularly, density gradient ultracentrifugation should be applied to reduce contaminants such as cell wall fragments and enrich high-purity EVs [8, 24]. Moreover, bacterial EV properties may vary within the same strain depending on growth conditions and stressors [25-27]. Therefore, establishing standardized yet strain-specific guidelines remains a high priority to optimize EV recovery, improve quality, and ensure reproducibility.
Unlike bacterial EVs, eukaryotic EV isolation is far more standardized with established minimal guidelines [1, 17, 28]. Notably, the International Society for Extracellular Vesicles (ISEV) has continuously refined its guidelines with the latest update, Minimal Information for Studies of Extracellular Vesicles (MISEV) 2023 [1], which provides comprehensive recommendations for EV research across various biological sources. These guidelines emphasize rigor, reproducibility, and transparency, addressing key challenges including nomenclature, separation methods, characterization techniques, and functional analyses [20]. ‘EV’ is an umbrella term encompassing all subtypes of eukaryotic vesicles including exosomes, ectosomes (microvesicle, microparticle), and apoptotic bodies, each of which displays distinct characteristics such as size, shape, density, and molecular markers based on biogenesis pathways [29-31]. Classically, EVs were isolated by differential centrifugation. However, this method cannot discriminate diverse EV subtypes. Current research studies on eukaryotic EVs have applied complementary techniques such as density gradient ultracentrifugation, size-exclusion chromatography, affinity chromatography, asymmetric flow field-flow fractionation, and others to discriminate the heterogenic nature of EVs, enabling extensive characterization of specific subtypes [1]. While no single marker or method can universally define all eukaryotic EVs, immunoaffinity-based isolation using specific markers can enrich particular subtypes [32-34]. In contrast, bacterial EV studies have yet to establish a detailed classification system for EV subtypes. For instance, Gram-positive EVs are broadly categorized as CMVs without further subclassification [7]. In addition, Gram-negative bacterial EVs are classified into OMVs, outer-inner membrane vesicles (OIMVs), and explosive outer membrane vesicles (EOMVs). However, their detailed molecular and biophysical profiles remain poorly defined [35-37]. Akin to those in eukaryotic EV research, it would be necessary to focus on comprehensive characterization of EV types by their biochemical composition, surface markers, genetic cargo, and physicochemical properties.
This review primarily focuses on purification methods for bacterial EVs, emphasizing in vitro culture-based isolation techniques. While it highlights challenges of isolating bacterial EVs from biological samples, where host-derived EVs are predominant, the primary goal is to establish a straightforward procedure for achieving high-purity and reproducibility of EV isolations from bacterial cultures. Effective methodologies will be pivotal in bacterial EV research for robust comparative analyses across strains. Ultimately, these approaches hold promise for unlocking the potential of EVs as reliable biomarkers and safe therapeutic tools in clinical settings.
Optimization of Bacterial Culture Conditions
Bacterial growth conditions can affect the production, content, dead vesicles, yield, purity, and degree of potential contaminants, thus affecting the quality of EVs [19, 38]. Various factors, including nutrient availability, salt concentration, pH, and oxygen levels, can significantly impact the generation and functionality of EVs (Fig. 1)[25, 32, 38]. Unlike eukaryotic cells, bacteria usually show rapid proliferation with concurrent cell division and lysis that can complicate the isolation of intact EVs from live bacteria [39, 40]. Thus, a major challenge in EV purification is distinguishing genuine EVs from contaminants such as cell debris and death-associated particles. It is quite difficult to discriminate natural EVs from other particles from dead bacteria. Thus, minimizing bacterial death with careful maintenance of viability and optimization of growth conditions tailored to each species is critical. Importantly, bacterial growth dynamics vary depending on species and environmental conditions. For this reason, it is highly recommended to establish a growth curve for each strain to fine-tune culture parameters and obtain an optimal growth phase. For experimental reproducibility, batch culture techniques are widely used for consistent bacterial growth, with aliquoted bacterial stocks stored at -80°C for providing long-term preservation and consistency across experiments to prevent genetic mutation [41]. For large-scale bacterial cultures, performing serial dilutions for sub-culture is effective in achieving an optimal optical density (OD) [19]. Inoculating large-scale cultures directly from frozen stocks can cause inconsistent bacterial growth from batch-to-batch. Thus, initiating bacterial culture with a small-volume starter culture (3-5 ml) enables stable bacterial growth. This dilution-based adaptation strategy helps synchronize bacterial populations and stabilize bacterial growth within a short time frame [42].
Fig. 1. Environmental and cellular factors influencing bacterial EV production.
Bacterial EV release is dynamically regulated by a variety of environmental factors (left), including temperature, pH, oxygen availability, nutrient conditions, antibiotics, osmotic pressure, and host interaction. Likewise, cellular states (right), such as viability, growth phase, cell density, phage infection status, metabolic activity, motility behavior, and membrane integrity, collectively influence the quantity and composition of secreted EVs. Understanding the interplay between these factors is essential for standardizing EV isolation and interpreting results across diverse microbial systems.
Bacterial Growth Phase
The bacterial growth phase critically shapes EV properties. During the logarithmic (log) phase, bacteria undergo rapid growth with heightened metabolic activity, resulting in extensive EV production [43]. However, the stationary phase causes stress responses and quorum sensing of bacteria, which can modify EV contents with accumulation of EVs produced at earlier phases and generation of vesicles from bacterial lysis [44, 45]. To reduce these artifacts, EVs are often harvested during the mid-to-late log phase, in which bacterial viability remains high with minimal substantial cell death (Table 1). For instance, Escherichia coli EVs are commonly collected before reaching an OD600 of 1.0 corresponding to the late-log phase to reduce contamination from dying cells [46]. Recent studies have demonstrated distinct characteristics of growth phase-specific EVs. For example, Helicobacter pylori EVs isolated from the late-exponential phase contain higher levels of protein, DNA, and RNA than EVs isolated from early-exponential and stationary growth phases [47, 48]. Similarly, Gram-positive Staphylococcus aureus EVs exhibit distinctive cargo profiles across growth phases, with variations in metabolite and protein compositions [49]. Bacillus subtilis EVs also display differential protein expression patterns between sporulation and vegetative growth phases, reflecting bacterial developmental transitions [50]. Along with changes in EV compositions, the bacterial growth phase can influence EV functionality. For example, Streptococcus pneumoniae EVs interact differently with macrophages depending on their harvest point [51]. Additionally, EVs from Lacticaseibacillus rhamnosus harvested during the late stationary-phase (48 h) exhibit higher immunomodulatory capacity than those collected at earlier time points (6 h and 12 h) [38]. This enhancement is attributed to elevated levels of lipoteichoic acid (LTA), a key TLR2 activator essential for immune modulation [38]. Although L. rhamnosus EVs derived from the stationary-phase exhibit enhanced therapeutic potential, some of them might have originated from cell death of bacteria rather than active secretion. These studies provide insight into bacterial EV functionality, which is largely affected by parental bacterial growth status and density. Thus, careful evaluation of their growth is needed. Likewise, determining the optimal timing for EV harvesting is crucial to acquire natural purity-favoring log-phase EVs for higher yields and potential functional advantages. Further studies are required to distinguish naturally secreted EVs from death-associated vesicles in late stationary-phase cultures to understand bacterial EV-mediated intercellular networks.
Table 1.
Reported methods for EV purification from representative bacterial species.
|
Culture Media Composition
Next, the composition of growth media serves as another critical factor influencing bacterial EV release and content. Complex media such as lysogeny broth (LB), brain heart infusion (BHI), and tryptic soy broth (TSB) contain rich nutrients and support robust bacterial growth, making them suitable for large-scale EV production [52-54]. It has been reported that 2-fold concentrated LB can induce 70% increase of EV production in Vibrio vulnificus compared to standard LB [25]. However, complex media contain undefined components from yeast or tissue extracts that might interfere with downstream applications. Although autoclaving can disrupt vesicle-like artifacts and denature proteins, residual impurities may still persist [55]. For this reason, defined media are used in EV isolation because they could allow precise control over nutrient availability and bacterial metabolic states while minimizing unwanted impurities in undefined media. Moreover, adding specific supplements to defined media can significantly alter the quantity and molecular profiles of EVs. For example, supplementing 1% glycine to E. coli Nissle cultures led to a 69-fold increase in EV protein content and a 51-fold increase in lipid content while simultaneously reducing lipopolysaccharide (LPS) endotoxin activity [56]. This precisely defined culture medium ensures greater consistency in EV production and composition, providing advantages for certain targeted applications that demand strict control over purity and molecular content, such as for vaccine development [57, 58]. Nonetheless, the limited nutrient availability in defined media often results in stress and slower growth of bacteria, which can affect EV secretion kinetics [19]. Therefore, the choice of bacterial culture medium is important for robust bacterial growth and EV release, especially for large-scale EV production.
Environmental Factors
Environmental factors are key determinants of bacterial EV release (Fig. 1) [59]. Temperature is a major effector of bacterial growth. Many bacterial species show optimal growth and EV release at around 37°C. However, their production varies according to the temperature across species. For instance, E. coli increases EV production at elevated temperatures, whereas Pseudomonas aeruginosa shows little changes in EV production between 25°C and 37-39°C [59, 60]. Similarly, V. vulnificus usually grows optimally at 30°C, but a higher temperature at 37°C induces a marked increase of EVs [25]. pH is another crucial factor affecting bacterial metabolism and EV properties [61]. Although many bacterial species favor a neutral pH (7.0-7.4) for optimal growth and EV production, alteration in pH can affect EV formation. For example, the facultative intracellular pathogen Salmonella Typhimurium increases EV production inside acidic vacuoles of host cells by changing expression of outer membrane proteins [62]. Notably, oxygen availability affects bacterial EV production. Some facultative anaerobes benefit from proper oxygenation and agitation (e.g., shaking) for promoting consistent growth and EV release [63]. In contrast, obligate anaerobes such as gut microbiota require strictly anaerobic conditions, often necessitating anaerobic chambers to sustain their viability and EV release [64, 65]. Although opportunistic strains including P. aeruginosa and S. aureus exhibit flexibility in oxygen tolerance, they still require careful oxygen adjustments to optimize EV release [66]. Salt concentration in culture medium also affects bacterial viability and EV production [67, 68]. Proper osmotic conditions during bacterial growth can support consistent EV formation, whereas extreme hypertonic or hypotonic conditions can induce osmotic stress known to disrupt EV membranes [68]. Finally, specific stressors such as antibiotics can modulate the yield and composition of EVs [69]. For instance, treatments with polymyxin B or D-cycloserine can enhance EV release potentially as part of a bacterial defense mechanism [59].
Overall, optimizing environmental conditions for bacterial growth while avoiding excessive stress to parental cells is essential for obtaining EVs with high purity and yield. Establishing species-specific culture conditions allows precise control over EV production and provides valuable insights into how environmental factors mimic natural infection sites or host-associated niches, such as pH fluctuations in inflamed tissues, hypoxic conditions in the gut, and mucin-rich environments in gastrointestinal tracts. Understanding these adaptations of bacteria with EV release can facilitate meaningful comparisons between in vivo bacterial EVs and those produced in vitro under laboratory conditions. To enhance reproducibility and standardization in EV research, detailed documentation of bacterial growth parameters, including culture conditions and OD thresholds, should be thoroughly reported in experimental protocols [17].
Removal of Bacteria and Harvesting Supernatant
Once bacterial growth is optimized, the next critical step is obtaining a bacteria-free supernatant to prevent contamination from live bacteria, cell debris, and nucleic acid-rich substances (Fig. 2), which can lead to EV aggregation and loss (EVs here refer collectively to CMVs, EOMVs, OIMVs, and OMVs) [70, 71]. Therefore, complete removal of bacteria is fundamental to EV isolation, as residual cells and their byproducts can largely affect EV purity and functionality [17]. To reduce contamination, bacteria should be removed immediately after harvesting them in conditioned medium at an appropriate OD or growth phase. Delayed processing and prolonged exposure to room temperature can allow bacteria to continue growing. Sudden temperature shifts to ice can also induce stress or lysis of bacteria [72]. This is particularly concerning for anaerobic bacteria as oxygen exposure can trigger lysis to release sticky materials such as nucleic acids, which can hinder bacterial pelleting while promoting EV aggregation [73-75]. Moreover, freezing bacterial culture medium before bacterial removal is strongly discouraged because freeze-thaw cycles are known to rupture bacteria and induce further contamination of bacterial debris with ruptured cytoplasmic contents.
Fig. 2. Overview of bacterial EV isolation workflow.
Different types of EVs are produced by Gram-positive (e.g., CMV) and Gram-negative bacteria (e.g., OMV, OIMV, EOMV). EV isolation begins with pre-isolation processing including bacterial culture, pre-clearance of culture supernatant, and concentration. For EV purification, size- or density-based separation methods such as size-exclusion chromatography and ultracentrifugation are used. Density gradient ultracentrifugation enables high-purity EV fractionation based on EV density.
Differential Centrifugation of Bacteria Cultures
Differential centrifugation (pre-clearing phase) is widely used to remove bacteria and their debris. The success of this technique largely depends on the precise selection of centrifugal forces in a step-wise manner to ensure efficient bacteria removal while minimizing EV loss and damage. This stage requires a carefully chosen centrifugal force to minimize shear stress, which could otherwise lead to bacterial lysis and the release of unwanted cellular components, causing a low EV purity [76]. Since many bacterial cells typically range from 1-3 μm in diameter, low-speed centrifugation (e.g., 3,000-6,000 ×g) is applied to effectively remove intact bacterial cells [46, 77]. For representative laboratory bacteria E. coli, a relative centrifugal force (RCF) of 6,000 ×g for 20 min at 4°C is commonly used to sediment intact bacterial cells (Table 1). This step is recommended to be repeated at least twice to ensure complete removal [78]. However, some bacterial species present additional challenges. For example, for A. muciniphila, which grows in mucin-rich media, it is difficult to completely remove them due to their adhesive properties to mucin, which can cause inefficient pelleting [12]. In addition, bacteria with low viability (e.g., some obligate anaerobes, slow-growing bacteria) are prone to lysis during centrifuge, potentially leading to contamination with cellular debris and nucleic acids in the supernatant [79]. For these bacteria, repeating the low-speed centrifugation step can improve bacterial removal efficiency with while preventing their lysis before proceeding to the next purification step. To check bacterial clearance, plating the supernatant onto an agar plate can be used to confirm the absence of viable bacteria by colony counting.
Once bacterial cells are removed, the supernatant is subjected to higher-speed centrifugation to eliminate smaller cell debris and large aggregates [80]. Typically, centrifugation at 10,000 ×g for 20 min is sufficient to pellet non-EV particles from the supernatant (Table 1) [81, 82]. This step can also be repeated, if necessary, to improve the removal of residual contaminants. Careful transfer of the supernatant is important, as disturbing the pellet may reintroduce debris into the purified fraction. It is also recommended to avoid collecting the entire supernatant down to the last drop, as this can carry over unwanted particles into subsequent steps.
Differential sequential centrifugation steps take advantage of differences in size and mass among bacterial cells, cellular debris, and EVs, enabling an effective pre-purification process [83, 84]. Differential centrifugation provides an indispensable, cost-effective, and scalable initial step in EV isolation [76]. However, differential centrifugation alone does not achieve absolute size-dependent separation, as sedimentation is also affected by particle density and cargo composition [85].
Filtration of Supernatant
Microfiltration (polishing phase) is commonly used to completely remove live bacteria for obtaining sterile bacterial EVs [86]. Filtration is a crucial step for EV purification as it can remove host debris, large particles, and potential contaminants. Filter pore size plays a key role in determining the efficiency of bacterial and debris removal. Typically, 0.22 μm or 0.45 μm pore-sized filters are used to eliminate bacteria while allowing EVs to pass through because most EVs have sizes ranging from 20 nm to 150 nm (Table 1). Filters with pore sizes of 0.22 μm and 0.45 μm can retain particles larger than 220 nm and 450 nm, respectively (±20%) [7]. In some cases, a sequential filtration approach using both 0.45 μm and 0.22 μm filters can be applied for more stringent bacterial removal [87]. The initial larger pore filter helps eliminate lipid-rich or sticky components that could otherwise clog the finer 0.22 μm filter, thereby reducing consumption of expensive disposable filters. However, filtration is not infallible despite its utility. Certain bacterial species have been shown to pass through 0.22 μm-pore sized filters and grow thereafter [88]. Therefore, parallel validation by microbial plating after filtration is necessary to confirm the absence of viable bacteria [89]. Filter pore sizes ranging from 0.45 μm to 0.1 μm can be selected to improve sample quality without compromising EV recovery depending on the bacterial strain and culture conditions.
Another technical consideration is the aggregation of EVs during filtration. This aggregation could be induced by the lipid-rich nature of EV membrane and its interaction with residual proteins or nucleic acids [90]. Although filtration can assist in removing aggregates, excessive filtering pressure and repetitive membrane usage may result in EV loss or membrane fouling. To mitigate such risks, temperature control is crucial to maintain supernatants at 4°C or on ice immediately after bacterial removal. Freezing the supernatant is not recommended as freeze-thaw cycles can alter media compositions and disrupt EV integrity [91]. If immediate processing is not feasible, storing the supernatant at 4°C for overnight is generally preferable rather than freezing. Additionally, to improve EV recovery, gentle swirling or agitation during filtration can help prevent vesicle adhesion to the filter membrane. The choice of filter material can also influence sample retention. Hydrophilized polyvinylidene difluoride (PVDF) membranes are recommended due to their low protein-binding properties, which can reduce non-specific binding of EVs on the membrane [92]. Finally, the use of stabilizing or cryo-protective agents such as trehalose, polyethylene glycol, and naturally derived polysaccharides has been found to be able to reduce EV aggregation in supernatant [93, 94]. In particular, trehalose has been shown to be able to enhance colloidal stability and improve particle yield by minimizing vesicle-surface interactions in human EV studies [93]. Examining whether such additives can similarly benefit bacterial EVs as observed in human EVs would be useful.
In summary, proper centrifugation and filtration strategies are required according to specific bacterial species and culture conditions to improve EV yield, quality, and reproducibility. Selecting appropriate centrifugal forces, choosing suitable filter pore size, minimizing membrane fouling, and considering strain-specific variables will allow researchers to confidently isolate bacterial EVs for functional studies and multi-omics analyses.
Concentration of Supernatant
Once a proper amount of pure supernatant (i.e., free of bacteria and cell debris) has been harvested, concentrating the supernatant is often required to obtain a sufficient EV yield. Reducing the sample volume can improve the efficiency of subsequent purification steps, with concentrating levels ranging from at least 2-fold to several hundred–fold, depending on experimental needs. Several methods are available for this purpose, each with distinct advantages and limitations.
Ultrafiltration via Tangential Flow Filtration (TFF)
Ultrafiltration is a pressure-driven membrane filtration method widely used to concentrate EVs from culture supernatant [95]. Polyethersulfone or regenerated cellulose is widely used as ultrafiltration membranes. They offer high permeability and low fouling tendencies, enabling for the selectivity of diverse sized particles with pores in a range of molecular weight cut-offs (MWCO) from 10 kDa to 100 kDa and allowing for precise separation of EVs from smaller contaminants [96]. Traditional ultrafiltration systems rely on normal dead-end flow filtration, where the fluid moves perpendicular to the membrane surface. However, this approach has limitations due to membrane clogging, as filtered particulates will accumulate on the membrane to form a ‘filter cake’ that can reduce flow efficiency and cause fouling over time [97]. By contrast, TFF has become a frequently used method for EV concentration because it can efficiently concentrate large sample volumes and prevent membrane fouling (Fig. 2) [98]. Unlike conventional filtration where particles accumulate on the membrane, TFF directs liquid tangentially along the membrane surface, generating shear forces that can block clogging and allow continuous filtration [97]. This method is particularly suitable for concentrating bacterial culture supernatants. It supports reproducible, scalable large-scale EV production as in eukaryotic EVs [99]. Earlier systems such as the QuixStand benchtop equipment have been employed for supernatant concentration and diafiltration [78]. The QuixStand system was practical for laboratory-scale applications due to its compact size and adaptability. However, this unit required manual pressure adjustments to prevent membrane damage, making it less useful for high-throughput workflows. Due to these limitations, this system has been discontinued and more advanced TFF platforms have since been developed [80]. Among these, the AKTA flux (Cytiva) is a versatile crossflow filtration system that uses hollow fiber cartridges and programmable settings suitable for diverse experimental scales [100]. Currently, different from hollow fiber systems, modern TFF workflows often adopt cassette (flat-sheet) membranes as they offer high packing density and pump compatibility, enabling greater throughput, more efficient large-scale bioprocessing, and higher yield of EV isolation [101]. In addition, alternative setups such as Pellicon 2 cassette filter membrane (Merck) combined with MasterFlex pump system (Cole-Parmer) can provide customizable configurations for EV concentration in flexible laboratory environments [102].
Centrifugal Filter Units (CFU)
For small sample volumes, CFU systems (e.g., Centriprep, Amicon, Vivaspin) offer a convenient and scalable alternative for concentrating EVs. Moreover, due to a dead volume in TFF systems, the final concentrated volume often remains around 100 ml, necessitating further concentration. To address this, CFU systems can be employed to achieve desired concentrations. CFUs operate by applying centrifugal force to drive liquid through a membrane with a defined MWCO, typically ranging from 10 kDa to 100 kDa, thereby retaining EVs while allowing smaller molecules to pass [103]. Sample concentration is usually performed using high-speed centrifugation (e.g., 3,000 ×g for 10-30 min), with repeated cycles as needed depending on sample volume and filter specification. While CFUs offer simplicity and convenience, they might cause vesicle loss due to membrane clogging, particularly when samples are viscous or protein-rich [104, 105]. To minimize this, pre-filtration and cold processing (4°C) are recommended to reduce the consumption of expensive CFU filters. Despite these limitations, CFUs remain a valuable tool for small scale or pilot EV preparations, especially when they are used judiciously with proper membrane selection and volume control strategies [106].
Precipitation-Based Methods
In addition to membrane-based methods, precipitation has been explored as an alternative method for concentrating EVs from culture supernatants [107-109]. Instead of physical concentration techniques, precipitation involves adding a high concentration of salts or polymers, which can disrupt hydrogen bonds and surface charges, leading to protein aggregation and facilitating EV isolation by centrifugation [95]. Ammonium sulfate is frequently used due to its high solubility and ionic strength, enabling bacterial EV precipitation through gradual addition with stirring at 4°C [95, 110]. It is considered a cost-effective and scalable process that does not require specialized equipment. However, a precipitation method has several drawbacks. Its use is limited with rich growth media, as various components can co-precipitate with EVs, restricting its applicability to minimal media. Furthermore, co-precipitation of bacterial components such as non-vesicular extracellular proteins can reduce sample purity and complicate downstream applications [111]. This method might also introduce variability in EV composition, as certain vesicle subtypes or aggregates may preferentially precipitate, potentially affecting reproducibility [112]. Additionally, residual precipitation agents must be thoroughly removed through extensive dialysis against a suitable buffer to prevent interference with subsequent analyses. Given these limitations, precipitation is generally not recommended as a standard method for bacterial EV purification. Similar to vesicles artificially generated through mechanical shearing, detergent treatment, or chemical induction, precipitated EVs might also exhibit structural and functional differences from naturally secreted EVs [113]. These artificially generated vesicles often with an aim to produce them in large quantities can lead to misinterpretation of EV characteristics and compromise the reproducibility of experimental applications. Therefore, precipitation methods are not suitable when high-purity or functionally intact EVs are required for downstream analyses.
In conclusion, ultrafiltration methods, particularly those based on TFF, are recommended. After ultrafiltration, a final concentrated supernatant can be sterilized with 0.22-μm pore membranes or high-speed centrifugation at 4°C to remove aggregates or precipitates that might have occurred during concentration [114]. Finally, after concentration, an additional filtration step (0.22-0.45 μm pore size) or high-speed centrifugation (e.g., 10,000 ×g for 10-30 min) is recommended to remove aggregates prior to further purifications.
Isolation and Separation of EVs
After concentrating a bacterial supernatant, further purification steps are essential to isolate pure nano-sized EVs. These steps focus on eliminating contaminants such as protein aggregates, cell debris, and bacterial appendages (e.g., flagella, fimbriae, pili) while preserving the structural integrity and biological functionality of EVs. Popular techniques include ultracentrifugation, size-exclusion chromatography (SEC), ultrafiltration, and sucrose cushion, while it is difficult to apply immunoaffinity-based purification for bacterial EVs due to strain-level diversities [97]. Each method has distinct advantages and limitations in terms of purity, scalability, and compatibility.
Ultracentrifugation
Ultracentrifugation is a conventional technique used to pellet EVs by applying high centrifugal forces, typically 100,000 ×g to 200,000 ×g for 1-3 h (Table 1). This method is relatively simple and fast. Although it is effective, it has several critical limitations that must be considered. Utilizing suboptimal RCF speed or insufficient spin time may result in incomplete EV recovery, leaving EVs in the concentrated supernatant and resulting in a low yield [84]. Conversely, excessive centrifugal force or prolonged duration can lead to heterogeneity in final preparations, such as a mixture of EVs, protein complexes, and other contaminants [84]. Moreover, high centrifugal forces can compact EVs into a dense pellet, leading to aggregation and structural damage, thereby reducing sample integrity and potentially altering biological functionality [115]. In human EV studies, ultracentrifugation has been associated with impaired functionality of EVs, potentially due to damaging forces exerted on vesicles during centrifugation at a high speed [76, 116]. Additionally, contamination risk is increased due to co-pelleting of cellular debris or bacterial appendages, which share similar sedimentation properties with EVs. Furthermore, ultracentrifugation may lead to structural deformation or rupture of EVs. Shear stress during high-speed spinning can destabilize vesicle membranes, compromising their integrity. Moreover, EV pellets often exhibit viscous or adhesive properties due to their lipid and nucleic acid contents, making them difficult to harvest and thereby increasing the risk of sample loss. Nevertheless, ultracentrifugation remains the most commonly used method due to its cost-effectiveness, simplicity, and accessibility, although careful consideration is required for optimal yield and purity.
Size-Exclusion Chromatography
SEC is a gentle and effective method for isolating EVs based on size differences [117, 118]. In this technique, EVs pass through a column filled with porous beads, where larger particles will be eluted first, while smaller contaminants (e.g., proteins, metabolites, solutes) are retained longer within pores [76]. Traditionally, SEC requires large fast performance liquid chromatography systems, limiting its practicality for routine EV isolation in typical laboratory settings [100]. However, commercially available pre-packed SEC kits such as qEV (Izon) have significantly simplified EV purification by utilizing passive gravity-driven flow, which can minimize vesicle aggregation and preserve structural integrity [97, 119]. These ready-made columns enable rapid purification with minimal (< 5%) cellular protein contamination in a human EV study [120]. To optimize the purity and yield, the pore size of SEC beads should be carefully considered as it determines the resolution for separating EVs from non-EV components with different sizes. Selecting an appropriate pore size is critical to ensure efficient exclusion of larger contaminants while allowing precise fractionation of target EV population [121]. Prior to loading a concentrated bacterial supernatant, the SEC column should be washed with PBS to remove trace amounts of storage materials (e.g., glycerin) to activate the separation matrix [117]. EVs can subsequently be eluted using 0.1 μm-filtered PBS [122]. SEC fractionation follows standardized elution profiles. EVs are typically eluted in specific early fractions, thereby ensuring reproducibility and consistency across experiments [122]. This predictable fractionation pattern allows researchers to collect EV-rich fractions while discarding later fractions containing smaller contaminants such as free proteins and metabolites [100]. As a result, SEC has been widely adopted as a standalone method or in combination with ultracentrifugation or density-gradient centrifugation to enhance EV purity (Fig. 2). It is particularly well-suited for being integrated into analytical pipelines involving proteomics and genomics [114, 123]. Its limitations include high cost, maintenance requirements including membrane washing and proper storage, and limited scalability for high-throughput or clinical practice.
Alternative Methods
Recently, the use of superabsorbent polymer beads has been explored as an alternative method for concentrating human EVs from cell culture medium [117, 124]. These beads composed of polyacrylamide-to-itaconic acid can selectively absorb liquid (water, media) while retaining EVs in a significantly concentrated state [124]. Although this method has not been applied to isolate bacterial EVs, it might be a promising technique for isolating host-derived fluids such as human urine or for large-volume processing. It offers improved isolation performance over conventional filtration [125]. By using concentrated EVs in supernatants, this method does not require further centrifugation steps. In addition, its EV losses are low. However, smaller byproducts such as metabolites and soluble proteins might still be retained, depending on their interactions with the polymer surface.
In some cases, a “cushion” of the concentrated supernatant is placed atop a dense medium at the bottom of the ultracentrifuge tube to form a step gradient [126]. This approach is useful for enhancing part of the separation by making it easier to re-suspend EVs and prevent damage to vesicles that might not withstand pelleting. To mitigate these risks, rather than stacking EVs by ultracentrifugation, a sucrose or iodixanol density cushion can be employed to absorb impact forces. As the word “cushion” implies, this approach can prevent vesicles from undergoing excessive mechanical stress by allowing them to form a distinct band at the interface of the sucrose or iodixanol layer [126]. This method not only minimizes contamination of soluble proteins and protects EVs from shear-induced damage, but also facilitates easier EV retrieval compared to direct ultracentrifugation. However, residual sucrose or iodixanol might remain in the sample. Thus, additional washing steps are needed for accurate characterization or quantification of EVs.
Many studies choose to terminate EV purification at this stage, particularly when large quantities are required, such as for in vivo mouse experiments. If QC methods, including transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA), confirm that the purity is sufficient, further purification can be omitted. Nonetheless, researchers working on immune response studies must be cautious as contamination with fimbriae or pili could affect immunological outcomes [1]. While EVs purified up to this step can achieve reasonable purity for many bacterial strains [64, 67], additional purification is strongly recommended for high-resolution analyses such as –omics studies to ensure the highest sample purity and consistency [8, 46]. This is particularly important when preparing EVs under conditions compatible with good manufacturing practice (GMP) for translational applications [127, 128].
Density Gradient Centrifugation
Density gradient centrifugation is one of the most effective methods for further purifying bacterial EVs after initial isolation steps (Fig. 2) [129]. By leveraging buoyant density differences, this method can effectively separate EVs from soluble proteins, free genetic materials, and other contaminants, providing high-purity EV compatible with follow-up analyses [130].
In earlier studies, sucrose gradients were often used for density-based EV separation due to their cost-effectiveness and buoyant density-based fractionation [131]. However, they require longer centrifugation time (> 16-20 h) because of their high viscosity [46, 132]. Furthermore, manual preparation can introduce variability in sucrose concentration, as minor inconsistencies in dissolution can affect density uniformity between experiments [130]. Moreover, sucrose aggregates may form during dissolution and prolonged storage, even under refrigeration, might increase the risk of microbial contamination. Compared to sucrose gradients, OptiPrep (iodixanol-based) gradients provide a more precise and reproducible approach for EV purification [133]. Iodixanol can form isosmotic solutions at all densities, preserve EV integrity, minimize aggregation, and ensure high-purity vesicle recovery [131]. It can also improve the separation of heterogeneous EV populations while avoiding density shifts observed in sucrose gradients during storage. Note that commercially available (ready-made) OptiPrep stock solution (Sigma-Aldrich) is supplied at 60% (w/v), not 100%, with a density of 1.320 ± 0.001 g/ml [134]. Therefore, precise dilution calculations are essential for gradient preparation. The working solution (50%) should be prepared first using a diluent buffer (Table 2) instead of PBS, as the use of PBS may lead to variations in salt concentration across gradient fractions [135]. All dilution solutions should be filtered with 0.45 μm membrane filters before use. A standard iodixanol gradient typically ranges from 0% to 50% (v/v), although specific concentrations should be optimized based on bacterial strain and EV density [131].
Table 2.
Preparation of a stepwise Iodixanol (OptiPrep) gradient (v/v).
|
Briefly, bacterial EVs are resuspended in an OptiPrep working solution to adjust concentration to 40-50% at the bottom, followed by sequential layering of lower concentrations (5-30%) (Table 2). The gradient is then centrifuged at 100,000 ×g to 200,000 ×g at 4°C for 2-3 h using an ultracentrifuge [8, 78]. Optimizing centrifugation parameters (e.g., force, time, and rotor type) for specific bacterial strains and EV density is crucial for reproducibility. While some protocols use longer centrifugation time (up to 16-20 h) to generate a linear continuous gradient, excessive spins may lead to multiple bands due to vesicle heterogeneity [136]. Continuous gradients allow finer separation of EV subpopulations. However, selecting the appropriate fraction requires extensive QC and validation. For efficient EV recovery, step gradients are preferred over continuous gradients, as they can concentrate EVs into distinct bands, thus minimizing vesicle loss (Table 1). If the goal is to isolate a homogenous vesicle subtype, flow cytometry such as CytoFLEX (Beckman) which can dissolve nano-sized vesicles based on their size and antigen expression might be a more suitable alternative [137, 138].
After centrifugation, fractions are collected from the top and analyzed for EV presence. Fraction collection should be tailored to EV characteristics and intended application. Strategies such as combining several fractions or adjusting the step gradient percentage can improve yield and purity. If EV amounts are sufficient, a visible band at the interface (typically at 1.1-1.24 g/ml, although it is strain-dependent) can be harvested directly [139, 140]. Otherwise, EV presence should be traced using density measurements, protein quantification, or further QC.
While density gradient centrifugation can significantly enhance EV purity, it does not fully eliminate protein complexes or bacterial components with similar densities. Frequently observed contaminants in EV proteomics studies include ribosomes, multi-tRNA synthetases, and heat-shock proteins [141]. These entities may co-purify with EV due to their overlapping buoyant densities, complex formation, or high abundance in bacterial cultures rather than representing genuine EV components [142]. Accordingly, to achieve the maximum purity and reproducibility for EV preparations, combining ultrafiltration and SEC with density gradient centrifugation is highly recommended [143]. This integrated approach can effectively remove aggregated vesicles and unwanted components, thus ensuring greater consistency in EV isolations.
QC and Storage of Purified EVs
For reproducible EV experiments, proper QC and storage methods are critical. Formally, purified EVs are characterized using TEM and NTA to confirm their nanoscale size and purity. However, unlike human EVs, bacterial EV lack universal markers such as CD63 and CD81, which are commonly used for human EV validation [144]. Consequently, bacterial EV studies tend to prioritize passing QC criteria, such as meeting size and purity thresholds, rather than ensuring consistency in methodology due to species diversity, contamination risks, and scalability issues [1]. This discrepancy highlights an urgent need for bacterial EV-specific reporting standards. Efforts akin to MISEV guidelines for eukaryotic EVs are essential.
A key challenge in bacterial EV isolation is the high cost of immunoaffinity-based methods, which can further complicate QC consistency (Table 3). While defining common molecular signatures including PAMPs such as Gram-positive LTA and Gram-negative LPS can help identify EVs, these PAMPs can also label contaminants from cell debris or aggregates, thus limiting their reliability as EV-specific markers [145, 146]. Instead, outer membrane proteins and surface-exposed proteins provide more specific validation targets. For example, an antibody against outer membrane protein A (OmpA) of E. coli or Acinetobacter baumannii is commercially available. It can be used to confirm EV QC via Western blotting or gold-labeling TEM [147, 148]. Similarly, S. aureus EVs contain IgG-binding protein (SbI), which can be easily detected using anti-IgG-gold labeling, thus offering a reliable validation marker [8]. Nevertheless, certain proteins (e.g., GroEL or flagellin) are inconsistently classified as either EV-enriched or EV-depleted, underlining the context-dependent nature of EV markers [139]. Although such protein-based markers are not universal, some microbial epitopes are shared among certain bacterial groups, which can facilitate bacterial EV identification while helping researchers distinguish them from non-vesicular contaminants and host-derived EVs in biological samples.
Table 3.
Representative bacterial EV markers and their validation methods.
|
Quantification and normalization strategies are also important. Protein concentration (e.g., BCA assay), lipid content, and vesicle counts with NTA, dynamic light scattering, or flow cytometry are commonly employed for standardization [149]. Depending on the study objective, different approaches can be used to compare total EV counts between samples. SDS-PAGE with Coomassie Brilliant Blue or silver staining can be used to assess protein pattern differences between whole cells and EV fractions, thus providing additional QC measures [46, 150].
As a final consideration, proper storage of bacterial EVs ensures reliable experimental outcomes. EVs are inherently sensitive to environmental changes due to their phospholipid bilayer structure and cargo of diverse biomolecules [151]. The MISEV2023 guidelines for eukaryotic EV storage strategies might also be applicable to bacterial EVs, as they emphasize standardized conditions for preserving vesicle integrity, functionality, and biomolecular content [1]. However, further validation is needed to confirm their suitability given that eukaryotic and bacterial EVs show structural and compositional differences. To preserve EV integrity and functionality while avoiding damage, aliquoting EV samples before storage can minimize freeze-thaw cycles. Numerous studies have demonstrated that repeated freeze-thaw events can lead to significant degradation of EVs, including reduction in particle number, increase of aggregation, and loss of bioactivity [149, 152, 153]. Therefore, EVs should be ideally stored at -80°C in single-use aliquots. Rapid freezing techniques such as immersion in dry ice or liquid nitrogen immediately after EV isolation can help preserve their properties. Alternatively, freeze-drying (lyophilization) has emerged as a potential strategy for enhancing long-term stability and transportability of EVs [91]. Recent studies have shown that freeze-drying with appropriate cryo- and lyoprotectants such as disaccharides (e.g., trehalose) combined with dextran or glycine can successfully maintain the physicochemical integrity of human EVs [154, 155]. Furthermore, biological excipients such as EV-depleted serum and secretome can enhance EV preservation and cellular uptake, indicating their dual roles as stabilizers and functional enhancers [91, 156]. Based on these promising results, the impact of lyophilization on functional activity of bacterial EVs should be explored post-reconstitution, particularly regarding immune modulation and host-pathogen interaction.
Ultimately, regardless of the chosen preservation method, minimizing freeze-thaw cycles at the earliest stage of EV handling is essential. By incorporating standardized QC metrics and optimized storage protocols, bacterial EV research can achieve greater reproducibility, ensuring high-quality EV for biological relevance, including functional assays and molecular characterization.
Concluding Remarks
Over the past few decades, the bacterial EV field has made remarkable strides, yet a standard EV isolation protocol remains a major challenge. Unlike human EVs, bacterial EVs rely heavily on physical separation rather than biochemical approaches due to species diversity and the absence of universal microbial markers. To ensure reproducibility, establishing minimal yet effective purification guidelines is essential. As a step toward future standardization, we propose a unified and practical EV isolation workflow with minimal equipment requirements (Fig. 3, Table 4). Such streamlined guidelines will support reproducibility across varying laboratory settings and help bridge resource gaps in bacterial EV research.
Fig. 3. Minimal-optimal suggested protocol for bacterial EV isolation.
Stepwise workflow integrating minimal essential methods with optimal components. Designed to support reproducible and scalable EV purification across diverse laboratory settings.
Table 4.
Minimal equipment and materials required for each step of bacterial EV purification.
|
Beyond in vitro isolation, distinguishing bacterial EVs from host biological fluids, stools, and tissues remains a hurdle as they share overlapping physicochemical properties [1, 4]. Refining strategies for accurate differentiation will provide clearer insights for clinical studies, particularly for microbiome research [157-159]. Leveraging insights from human EV studies, such as single-vesicle profiling (e.g., sequencing-based approaches), can help us define the heterogeneity of bacterial EV subtypes and improve differentiation strategies [160]. Improving bacterial culture conditions and optimizing isolation methods will enhance EV quality and consistency, which in turn can unlock their therapeutic potential as non-replicative yet immune-modulatory agents to enhance clinical safety [38]. Their promise as vaccine platforms and diagnostic biomarkers further reinforces the importance of establishing robust purification standards.
Acknowledgments
This study was supported by a grant (NRF-2023R1A2C1006040 and RS-2023-00219563) of the National Research Foundation funded by the Korea government (MSIT), and by the Soonchunhyang University Research Fund.
Footnotes
Conflict of Interest
The authors have no financial conflicts of interest to declare.
References
- 1.Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J. Extracell. Vesicles. 2024;13:e12404. doi: 10.1002/jev2.12451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Beveridge TJ. Structures of gram-negative cell walls and their derived membrane vesicles. J. Bacteriol. 1999;181:4725–4733. doi: 10.1128/JB.181.16.4725-4733.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Schwechheimer C, Kuehn MJ. Outer-membrane vesicles from Gram-negative bacteria: biogenesis and functions. Nat. Rev. Microbiol. 2015;13:605–619. doi: 10.1038/nrmicro3525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Qian D, Xu P, Wang X, Du C, Zhao X, Xu J. Bacterial extracellular vesicles for gut microbiome-host communication and drug development. Acta Pharm. Sin. B. 2025;15:1816–1840. doi: 10.1016/j.apsb.2025.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018;19:213–228. doi: 10.1038/nrm.2017.125. [DOI] [PubMed] [Google Scholar]
- 6.Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367:eaau6977. doi: 10.1126/science.aau6977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Toyofuku M, Nomura N, Eberl L. Types and origins of bacterial membrane vesicles. Nat. Rev. Microbiol. 2019;17:13–24. doi: 10.1038/s41579-018-0112-2. [DOI] [PubMed] [Google Scholar]
- 8.Lee EY, Choi DY, Kim DK, Kim JW, Park JO, Kim S, et al. Gram-positive bacteria produce membrane vesicles: proteomicsbased characterization of Staphylococcus aureus-derived membrane vesicles. Proteomics. 2009;9:5425–5436. doi: 10.1002/pmic.200900338. [DOI] [PubMed] [Google Scholar]
- 9.Prados-Rosales R, Baena A, Martinez LR, Luque-Garcia J, Kalscheuer R, Veeraraghavan U, et al. Mycobacteria release active membrane vesicles that modulate immune responses in a TLR2-dependent manner in mice. J. Clin. Invest. 2011;121:1471–1483. doi: 10.1172/JCI44261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Chernov VM, Mouzykantov AA, Baranova NB, Medvedeva ES, Grygorieva TY, Trushin MV, et al. Extracellular membrane vesicles secreted by mycoplasma Acholeplasma laidlawii PG8 are enriched in virulence proteins. J. Proteomics. 2014;110:117–128. doi: 10.1016/j.jprot.2014.07.020. [DOI] [PubMed] [Google Scholar]
- 11.Kabani M, Melki R. More than just trash bins? Potential roles for extracellular vesicles in the vertical and horizontal transmission of yeast prions. Curr. Genet. 2016;62:265–270. doi: 10.1007/s00294-015-0534-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kang CS, Ban M, Choi EJ, Moon HG, Jeon JS, Kim DK, et al. Extracellular vesicles derived from gut microbiota, especially Akkermansia muciniphila, protect the progression of dextran sulfate sodium-induced colitis. PLoS One. 2013;8:e76520. doi: 10.1371/journal.pone.0076520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Luo ZW, Xia K, Liu YW, Liu JH, Rao SS, Hu XK, et al. Extracellular vesicles from Akkermansia muciniphila elicit antitumor immunity against prostate cancer via modulation of CD8+ T cells and macrophages. Int. J. Nanomedicine. 2021;16:2949–2963. doi: 10.2147/IJN.S304515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chu H, Khosravi A, Kusumawardhani IP, Kwon AH, Vasconcelos AC, Cunha LD, et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science. 2016;352:1116–1120. doi: 10.1126/science.aad9948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Shen Y, Giardino Torchia ML, Lawson GW, Karp CL, Ashwell JD, Mazmanian SK. Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host Microbe. 2012;12:509–520. doi: 10.1016/j.chom.2012.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gul L, Modos D, Fonseca S, Madgwick M, Thomas JP, Sudhakar P, et al. Extracellular vesicles produced by the human commensal gut bacterium Bacteroides thetaiotaomicron affect host immune pathways in a cell-type specific manner that are altered in inflammatory bowel disease. J. Extracell. Vesicles. 2022;11:e12189. doi: 10.1002/jev2.12189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Thery C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles. 2018;7:1535750. doi: 10.1080/20013078.2018.1535750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wang Y, Duan C, Yu X. Extracellular vesicles of Gram-negative and Gram-positive probiotics. J. Microbiol. 2025;63:e2506005. doi: 10.71150/jm.2506005. [DOI] [PubMed] [Google Scholar]
- 19.Munoz-Echeverri LM, Benavides-Lopez S, Geiger O, Trujillo-Roldan MA, Valdez-Cruz NA. Bacterial extracellular vesicles: biotechnological perspective for enhanced productivity. World J. Microbiol. Biotechnol. 2024;40:174. doi: 10.1007/s11274-024-03963-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Liang A, Korani L, Yeung CLS, Tey SK, Yam JWP. The emerging role of bacterial extracellular vesicles in human cancers. J. Extracell. Vesicles. 2024;13:e12521. doi: 10.1002/jev2.12521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Liu Y, Chen J, Raj K, Baerg L, Nathan N, Philpott DJ, et al. A universal strategy to promote secretion of G+/G- bacterial extracellular vesicles and its application in host innate immune responses. ACS Synth. Biol. 2023;12:319–328. doi: 10.1021/acssynbio.2c00583. [DOI] [PubMed] [Google Scholar]
- 22.Hong SW, Kim MR, Lee EY, Kim JH, Kim YS, Jeon SG, et al. Extracellular vesicles derived from Staphylococcus aureus induce atopic dermatitis-like skin inflammation. Allergy. 2011;66:351–359. doi: 10.1111/j.1398-9995.2010.02483.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lee J, Lee EY, Kim SH, Kim DK, Park KS, Kim KP, et al. Staphylococcus aureus extracellular vesicles carry biologically active beta-lactamase. Antimicrob. Agents Chemother. 2013;57:2589–2595. doi: 10.1128/AAC.00522-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kim W, Lee EJ, Bae IH, Myoung K, Kim ST, Park PJ, et al. Lactobacillus plantarum-derived extracellular vesicles induce antiinflammatory M2 macrophage polarization in vitro. J. Extracell. Vesicles. 2020;9:1793514. doi: 10.1080/20013078.2020.1793514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Park JH, Song S, Kim S, Kim M, Kim KS. Optimizing conditions for the production of bacterial extracellular vesicles of Vibrio vulnificus and analysis of the inner small RNA compositions. J. Microbiol. Biotechnol. 2024;34:29–38. doi: 10.4014/jmb.2310.10002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Guo J, Huang Z, Wang Q, Wang M, Ming Y, Chen W, et al. Opportunities and challenges of bacterial extracellular vesicles in regenerative medicine. J. Nanobiotechnology. 2025;23:4. doi: 10.1186/s12951-024-02935-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Huang X, Hu Z, Shang W, Chen J, Hu Q, Zhou Y, et al. Beta-lactam antibiotics promote extracellular vesicle production of Staphylococcus aureus through ROS-mediated lipid metabolic reprogramming. J. Extracell. Vesicles. 2025;14:e70077. doi: 10.1002/jev2.70077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Lotvall J, Hill AF, Hochberg F, Buzas EI, Di Vizio D, Gardiner C, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J. Extracell. Vesicles. 2014;3:26913. doi: 10.3402/jev.v3.26913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Russell AE, Sneider A, Witwer KW, Bergese P, Bhattacharyya SN, Cocks A, et al. Biological membranes in EV biogenesis, stability, uptake, and cargo transfer: an ISEV position paper arising from the ISEV membranes and EVs workshop. J. Extracell. Vesicles. 2019;8:1684862. doi: 10.1080/20013078.2019.1684862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25:364–372. doi: 10.1016/j.tcb.2015.01.004. [DOI] [PubMed] [Google Scholar]
- 31.Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J. Cell Biol. 1983;97:329–339. doi: 10.1083/jcb.97.2.329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. USA. 2016;113:E968–977. doi: 10.1073/pnas.1521230113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Blijdorp CJ, Tutakhel OAZ, Hartjes TA, van den Bosch TPP, van Heugten MH, Rigalli JP, et al. Comparing approaches to normalize, quantify, and characterize urinary extracellular vesicles. J. Am. Soc. Nephrol. 2021;32:1210–1226. doi: 10.1681/ASN.2020081142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Mathieu M, Nevo N, Jouve M, Valenzuela JI, Maurin M, Verweij FJ, et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nat. Commun. 2021;12:4389. doi: 10.1038/s41467-021-24384-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Juodeikis R, Carding SR. Outer membrane vesicles: biogenesis, functions, and issues. Microbiol. Mol. Biol. Rev. 2022;86:e0003222. doi: 10.1128/mmbr.00032-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Perez-Cruz C, Carrion O, Delgado L, Martinez G, Lopez-Iglesias C, Mercade E. New type of outer membrane vesicle produced by the Gram-negative bacterium Shewanella vesiculosa M7T: implications for DNA content. Appl. Environ. Microbiol. 2013;79:1874–1881. doi: 10.1128/AEM.03657-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Suri K, D'Souza A, Huang D, Bhavsar A, Amiji M. Bacterial extracellular vesicle applications in cancer immunotherapy. Bioact. Mater. 2023;22:551–566. doi: 10.1016/j.bioactmat.2022.10.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sandanusova M, Turkova K, Pechackova E, Kotoucek J, Roudnicky P, Sindelar M, et al. Growth phase matters: boosting immunity via Lacticasebacillus-derived membrane vesicles and their interactions with TLR2 pathways. J. Extracell. Biol. 2024;3:e169. doi: 10.1002/jex2.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Mannik J, Kar P, Amarasinghe C, Amir A, Mannik J. Determining the rate-limiting processes for cell division in Escherichia coli. Nat. Commun. 2024;15:9948. doi: 10.1038/s41467-024-54242-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Din MO, Danino T, Prindle A, Skalak M, Selimkhanov J, Allen K, et al. Synchronized cycles of bacterial lysis for in vivo delivery. Nature. 2016;536:81–85. doi: 10.1038/nature18930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hecht A, Filliben J, Munro SA, Salit M. A minimum information standard for reproducing bench-scale bacterial cell growth and productivity. Commun. Biol. 2018;1:219. doi: 10.1038/s42003-018-0220-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Govantes F. Serial dilution-based growth curves and growth curve synchronization for high-resolution time series of bacterial biofilm growth. Methods Mol. Biol. 2018;1734:159–169. doi: 10.1007/978-1-4939-7604-1_13. [DOI] [PubMed] [Google Scholar]
- 43.Mao H, Gong T, Sun Y, Yang S, Qiao X, Yang D. Bacterial growth stage determines the yields, protein composition, and periodontal pathogenicity of Porphyromonas gingivalis outer membrane vesicles. Front. Cell. Infect. Microbiol. 2023;13:1193198. doi: 10.3389/fcimb.2023.1193198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Chen J, Zhang H, Wang S, Du Y, Wei B, Wu Q, et al. Inhibitors of bacterial extracellular vesicles. Front. Microbiol. 2022;13:835058. doi: 10.3389/fmicb.2022.835058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Deatherage BL, Lara JC, Bergsbaken T, Rassoulian Barrett SL, Lara S, Cookson BT. Biogenesis of bacterial membrane vesicles. Mol. Microbiol. 2009;72:1395–1407. doi: 10.1111/j.1365-2958.2009.06731.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lee EY, Bang JY, Park GW, Choi DS, Kang JS, Kim HJ, et al. Global proteomic profiling of native outer membrane vesicles derived from Escherichia coli. Proteomics. 2007;7:3143–3153. doi: 10.1002/pmic.200700196. [DOI] [PubMed] [Google Scholar]
- 47.Bitto NJ, Zavan L, Johnston EL, Stinear TP, Hill AF, Kaparakis-Liaskos M. Considerations for the analysis of bacterial membrane vesicles: methods of vesicle production and quantification can influence biological and experimental outcomes. Microbiol. Spectr. 2021;9:e0127321. doi: 10.1128/Spectrum.01273-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zavan L, Bitto NJ, Johnston EL, Greening DW, Kaparakis-Liaskos M. Helicobacter pylori growth stage determines the size, protein composition, and preferential cargo packaging of outer membrane vesicles. Proteomics. 2019;19:e1800209. doi: 10.1002/pmic.201970004. [DOI] [PubMed] [Google Scholar]
- 49.da Luz BSR, de Rezende Rodovalho V, Nicolas A, Chabelskaya S, Jardin J, Briard-Bion V, et al. Impact of environmental conditions on the protein content of Staphylococcus aureus and its derived extracellular vesicles. Microorganisms. 2022;10:1808. doi: 10.3390/microorganisms10091808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kim Y, Edwards N, Fenselau C. Extracellular vesicle proteomes reflect developmental phases of Bacillus subtilis. Clin. Proteomics. 2016;13:6. doi: 10.1186/s12014-016-9107-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Mehanny M, Kroniger T, Koch M, Hoppstadter J, Becher D, Kiemer AK, et al. Yields and immunomodulatory effects of pneumococcal membrane vesicles differ with the bacterial growth phase. Adv. Healthc. Mater. 2022;11:e2101151. doi: 10.1002/adhm.202101151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kram KE, Finkel SE. Rich Medium Composition Affects Escherichia coli survival, glycation, and mutation frequency during long-term batch culture. Appl. Environ. Microbiol. 2015;81:4442–4450. doi: 10.1128/AEM.00722-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wijesinghe G, Dilhari A, Gayani B, Kottegoda N, Samaranayake L, Weerasekera M. Influence of laboratory culture media on in vitro growth, adhesion, and biofilm formation of Pseudomonas aeruginosa and Staphylococcus aureus. Med. Princ. Pract. 2019;28:28–35. doi: 10.1159/000494757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Missiakas DM, Schneewind O. Growth and laboratory maintenance of Staphylococcus aureus. Curr. Protoc. Microbiol. Chapter. 2013;9:Unit. doi: 10.1002/9780471729259.mc09c01s28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Schulz E, Karagianni A, Koch M, Fuhrmann G. Hot EVs - how temperature affects extracellular vesicles. Eur. J. Pharm. Biopharm. 2020;146:55–63. doi: 10.1016/j.ejpb.2019.11.010. [DOI] [PubMed] [Google Scholar]
- 56.Hirayama S, Nakao R. Glycine significantly enhances bacterial membrane vesicle production: a powerful approach for isolation of LPS-reduced membrane vesicles of probiotic Escherichia coli. Microb. Biotechnol. 2020;13:1162–1178. doi: 10.1111/1751-7915.13572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bryant WA, Stentz R, Le Gall G, Sternberg MJE, Carding SR, Wilhelm T. In Silico analysis of the small molecule content of outer membrane vesicles produced by Bacteroides thetaiotaomicron indicates an extensive metabolic link between microbe and host. Front. Microbiol. 2017;8:2440. doi: 10.3389/fmicb.2017.02440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Baker SM, Settles EW, Davitt C, Gellings P, Kikendall N, Hoffmann J, et al. Burkholderia pseudomallei OMVs derived from infection mimicking conditions elicit similar protection to a live-attenuated vaccine. NPJ Vaccines. 2021;6:18. doi: 10.1038/s41541-021-00281-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.McBroom AJ, Kuehn MJ. Release of outer membrane vesicles by Gram-negative bacteria is a novel envelope stress response. Mol. Microbiol. 2007;63:545–558. doi: 10.1111/j.1365-2958.2006.05522.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Macdonald IA, Kuehn MJ. Stress-induced outer membrane vesicle production by Pseudomonas aeruginosa. J. Bacteriol. 2013;195:2971–2981. doi: 10.1128/JB.02267-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Caruana JC, Walper SA. Bacterial membrane vesicles as mediators of microbe - microbe and microbe - host community interactions. Front. Microbiol. 2020;11:432. doi: 10.3389/fmicb.2020.00432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Dehinwal R, Gopinath T, Smith RD, Ernst RK, Schifferli DM, Waldor MK, et al. A pH-sensitive motif in an outer membrane protein activates bacterial membrane vesicle production. Nat. Commun. 2024;15:6958. doi: 10.1038/s41467-024-51364-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Andre AC, Debande L, Marteyn BS. The selective advantage of facultative anaerobes relies on their unique ability to cope with changing oxygen levels during infection. Cell. Microbiol. 2021;23:e13338. doi: 10.1111/cmi.13338. [DOI] [PubMed] [Google Scholar]
- 64.Chelakkot C, Choi Y, Kim DK, Park HT, Ghim J, Kwon Y, et al. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp. Mol. Med. 2018;50:e450. doi: 10.1038/emm.2017.282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Zheng X, Gong T, Luo W, Hu B, Gao J, Li Y, et al. Fusobacterium nucleatum extracellular vesicles are enriched in colorectal cancer and facilitate bacterial adhesion. Sci. Adv. 2024;10:eado0016. doi: 10.1126/sciadv.ado0016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ishiai T, Subsomwong P, Narita K, Kawai N, Teng W, Suzuki S, et al. Extracellular vesicles of Pseudomonas aeruginosa downregulate pyruvate fermentation enzymes and inhibit the initial growth of Staphylococcus aureus. Curr. Res. Microb. Sci. 2023;4:100190. doi: 10.1016/j.crmicr.2023.100190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lee T, Jun SH, Choi CW, Kim SI, Lee JC, Shin JH. Salt stress affects global protein expression profiles of extracellular membrane-derived vesicles of Listeria monocytogenes. Microb. Pathog. 2018;115:272–279. doi: 10.1016/j.micpath.2017.12.071. [DOI] [PubMed] [Google Scholar]
- 68.Baumgarten T, Sperling S, Seifert J, von Bergen M, Steiniger F, Wick LY, et al. Membrane vesicle formation as a multiple-stress response mechanism enhances Pseudomonas putida DOT-T1E cell surface hydrophobicity and biofilm formation. Appl. Environ. Microbiol. 2012;78:6217–6224. doi: 10.1128/AEM.01525-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bose S, Aggarwal S, Singh DV, Acharya N. Extracellular vesicles: an emerging platform in gram-positive bacteria. Microb. Cell. 2020;7:312–322. doi: 10.15698/mic2020.12.737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Muller L, Hong CS, Stolz DB, Watkins SC, Whiteside TL. Isolation of biologically-active exosomes from human plasma. J. Immunol. Methods. 2014;411:55–65. doi: 10.1016/j.jim.2014.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kooijmans SAA, Stremersch S, Braeckmans K, de Smedt SC, Hendrix A, Wood MJA, et al. Electroporation-induced siRNA precipitation obscures the efficiency of siRNA loading into extracellular vesicles. J. Control. Release. 2013;172:229–238. doi: 10.1016/j.jconrel.2013.08.014. [DOI] [PubMed] [Google Scholar]
- 72.Rice KC, Bayles KW. Molecular control of bacterial death and lysis. Microbiol. Mol. Biol. Rev. 2008;72:85–109. doi: 10.1128/MMBR.00030-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lungu B, Ricke SC, Johnson MG. Growth, survival, proliferation and pathogenesis of Listeria monocytogenes under low oxygen or anaerobic conditions: a review. Anaerobe. 2009;15:7–17. doi: 10.1016/j.anaerobe.2008.08.001. [DOI] [PubMed] [Google Scholar]
- 74.Drasar BS. Cultivation of anaerobic intestinal bacteria. J. Pathol. Bacteriol. 1967;94:417–427. doi: 10.1002/path.1700940223. [DOI] [PubMed] [Google Scholar]
- 75.Speers AM, Cologgi DL, Reguera G. 2009. Anaerobic cell culture. Curr. Protoc. Microbiol. Appendix 4: Appendix 4F. 10.1002/9780471729259.mca04fs12 [DOI] [PubMed]
- 76.De Sousa KP, Rossi I, Abdullahi M, Ramirez MI, Stratton D, Inal JM. Isolation and characterization of extracellular vesicles and future directions in diagnosis and therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2023;15:e1835. doi: 10.1002/wnan.1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Jang KS, Sweredoski MJ, Graham RL, Hess S, Clemons WM., Jr Comprehensive proteomic profiling of outer membrane vesicles from Campylobacter jejuni. J. Proteomics. 2014;98:90–98. doi: 10.1016/j.jprot.2013.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kim OY, Park HT, Dinh NTH, Choi SJ, Lee J, Kim JH, et al. Bacterial outer membrane vesicles suppress tumor by interferongamma-mediated antitumor response. Nat. Commun. 2017;8:626. doi: 10.1038/s41467-017-00729-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Mauerhofer LM, Pappenreiter P, Paulik C, Seifert AH, Bernacchi S, Rittmann SKR. Methods for quantification of growth and productivity in anaerobic microbiology and biotechnology. Folia Microbiol. (Praha) 2019;64:321–360. doi: 10.1007/s12223-018-0658-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Liu C, Yazdani N, Moran CS, Salomon C, Seneviratne CJ, Ivanovski S, et al. Unveiling clinical applications of bacterial extracellular vesicles as natural nanomaterials in disease diagnosis and therapeutics. Acta Biomater. 2024;180:18–45. doi: 10.1016/j.actbio.2024.04.022. [DOI] [PubMed] [Google Scholar]
- 81.Choi HI, Choi JP, Seo J, Kim BJ, Rho M, Han JK, et al. Helicobacter pylori-derived extracellular vesicles increased in the gastric juices of gastric adenocarcinoma patients and induced inflammation mainly via specific targeting of gastric epithelial cells. Exp. Mol. Med. 2017;49:e330. doi: 10.1038/emm.2017.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Cui L, Yang R, Huo D, Li L, Qu X, Wang J, et al. Streptococcus pneumoniae extracellular vesicles aggravate alveolar epithelial barrier disruption via autophagic degradation of OCLN (occludin) Autophagy. 2024;20:1577–1596. doi: 10.1080/15548627.2024.2330043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Carnino JM, Lee H. Extracellular vesicles in respiratory disease. Adv. Clin. Chem. 2022;108:105–127. doi: 10.1016/bs.acc.2021.07.008. [DOI] [PubMed] [Google Scholar]
- 84.Sharifian Gh M, Norouzi F. Guidelines for an optimized differential centrifugation of cells. Biochem. Biophys. Rep. 2023;36:101585. doi: 10.1016/j.bbrep.2023.101585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Witwer KW, Buzas EI, Bemis LT, Bora A, Lasser C, Lotvall J, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J. Extracell. Vesicles. 2013;27:2. doi: 10.3402/jev.v2i0.20360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hahn MW. Broad diversity of viable bacteria in 'sterilé (0.2 microm) filtered water. Res. Microbiol. 2004;155:688–691. doi: 10.1016/j.resmic.2004.05.003. [DOI] [PubMed] [Google Scholar]
- 87.Mandelbaum N, Zhang L, Carasso S, Ziv T, Lifshiz-Simon S, Davidovich I, et al. Extracellular vesicles of the Gram-positive gut symbiont Bifidobacterium longum induce immune-modulatory, anti-inflammatory effects. NPJ Biofilms Microbiomes. 2023;9:30. doi: 10.1038/s41522-023-00400-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Mannik J, Driessen R, Galajda P, Keymer JE, Dekker C. Bacterial growth and motility in sub-micron constrictions. Proc. Natl. Acad. Sci. USA. 2009;106:14861–14866. doi: 10.1073/pnas.0907542106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Wang Y, Hammes F, Boon N, Egli T. Quantification of the filterability of freshwater bacteria through 0.45, 0.22, and 0.1 microm pore size filters and shape-dependent enrichment of filterable bacterial communities. Environ. Sci. Technol. 2007;41:7080–7086. doi: 10.1021/es0707198. [DOI] [PubMed] [Google Scholar]
- 90.Gupta D, Zickler AM, El Andaloussi S. Dosing extracellular vesicles. Adv. Drug Deliv. Rev. 2021;178:113961. doi: 10.1016/j.addr.2021.113961. [DOI] [PubMed] [Google Scholar]
- 91.Susa F, Limongi T, Borgione F, Peiretti S, Vallino M, Cauda V, et al. Comparative studies of different preservation methods and relative freeze-drying formulations for extracellular vesicle pharmaceutical applications. ACS Biomater. Sci. Eng. 2023;9:5871–5885. doi: 10.1021/acsbiomaterials.3c00678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Konoshenko MY, Lekchnov EA, Vlassov AV, Laktionov PP. Isolation of extracellular vesicles: general methodologies and latest trends. Biomed Res. Int. 2018;2018:8545347. doi: 10.1155/2018/8545347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Bosch S, de Beaurepaire L, Allard M, Mosser M, Heichette C, Chretien D, et al. Trehalose prevents aggregation of exosomes and cryodamage. Sci. Rep. 2016;6:36162. doi: 10.1038/srep36162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kusuma GD, Barabadi M, Tan JL, Morton DAV, Frith JE, Lim R. To protect and to preserve: novel preservation strategies for extracellular vesicles. Front. Pharmacol. 2018;9:1199. doi: 10.3389/fphar.2018.01199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Klimentova J, Stulik J. Methods of isolation and purification of outer membrane vesicles from gram-negative bacteria. Microbiol. Res. 2015;170:1–9. doi: 10.1016/j.micres.2014.09.006. [DOI] [PubMed] [Google Scholar]
- 96.Vergauwen G, Dhondt B, Van Deun J, De Smedt E, Berx G, Timmerman E, et al. Confounding factors of ultrafiltration and protein analysis in extracellular vesicle research. Sci. Rep. 2017;7:2704. doi: 10.1038/s41598-017-02599-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Liangsupree T, Multia E, Riekkola ML. Modern isolation and separation techniques for extracellular vesicles. J. Chromatogr. A. 2021;1636:461773. doi: 10.1016/j.chroma.2020.461773. [DOI] [PubMed] [Google Scholar]
- 98.Busatto S, Vilanilam G, Ticer T, Lin WL, Dickson DW, Shapiro S, et al. Tangential flow filtration for highly efficient concentration of extracellular vesicles from large volumes of fluid. Cells. 2018;7:273. doi: 10.3390/cells7120273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Katsura K, Matsuda T, Tomabechi Y, Yonemochi M, Hanada K, Ohsawa N, et al. A reproducible and scalable procedure for preparing bacterial extracts for cell-free protein synthesis. J. Biochem. 2017;162:357–369. doi: 10.1093/jb/mvx039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kapoor KS, Harris K, Arian KA, Ma L, Schueng Zancanela B, Church KA, et al. High throughput and rapid isolation of extracellular vesicles and exosomes with purity using size exclusion liquid chromatography. Bioact. Mater. 2024;40:683–695. doi: 10.1016/j.bioactmat.2024.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Fernandez-Cerezo L, Wismer MK, Han I, Pollard JM. High throughput screening of ultrafiltration and diafiltration processing of monoclonal antibodies via the ambr(R) crossflow system. Biotechnol. Prog. 2020;36:e2929. doi: 10.1002/btpr.2929. [DOI] [PubMed] [Google Scholar]
- 102.Kwon H, Lee EH, Park SY, Park JY, Hong JH, Kim EK, et al. Lactobacillus-derived extracellular vesicles counteract Abeta42-induced abnormal transcriptional changes through the upregulation of MeCP2 and Sirt1 and improve Abeta pathology in Tg-APP/PS1 mice. Exp. Mol. Med. 2023;55:2067–2082. doi: 10.1038/s12276-023-01084-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Kornilov R, Puhka M, Mannerstrom B, Hiidenmaa H, Peltoniemi H, Siljander P, et al. Efficient ultrafiltration-based protocol to deplete extracellular vesicles from fetal bovine serum. J. Extracell. Vesicles. 2018;7:1422674. doi: 10.1080/20013078.2017.1422674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lang JB, Buck MC, Riviere J, Stambouli O, Sachenbacher K, Choudhary P, et al. Comparative analysis of extracellular vesicle isolation methods from human AML bone marrow cells and AML cell lines. Front. Oncol. 2022;12:949261. doi: 10.3389/fonc.2022.949261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Wang F, Cerione RA, Antonyak MA. Isolation and characterization of extracellular vesicles produced by cell lines. STAR Protoc. 2021;2:100295. doi: 10.1016/j.xpro.2021.100295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ansari FJ, Tafti HA, Amanzadeh A, Rabbani S, Shokrgozar MA, Heidari R, et al. Comparison of the efficiency of ultrafiltration, precipitation, and ultracentrifugation methods for exosome isolation. Biochem. Biophys. Rep. 2024;38:101668. doi: 10.1016/j.bbrep.2024.101668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Oishi S, Miyashita M, Kiso A, Kikuchi Y, Ueda O, Hirai K, et al. Cellular locations of proteinases and association with vesicles in Porphyromonas gingivalis. Eur. J. Med. Res. 2010;15:397–402. doi: 10.1186/2047-783X-15-9-397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bauman SJ, Kuehn MJ. Purification of outer membrane vesicles from Pseudomonas aeruginosa and their activation of an IL-8 response. Microbes Infect. 2006;8:2400–2408. doi: 10.1016/j.micinf.2006.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Zhou L, Srisatjaluk R, Justus DE, Doyle RJ. On the origin of membrane vesicles in gram-negative bacteria. FEMS Microbiol. Lett. 1998;163:223–228. doi: 10.1111/j.1574-6968.1998.tb13049.x. [DOI] [PubMed] [Google Scholar]
- 110.Zhao X, Wei Y, Bu Y, Ren X, Dong Z. Review on bacterial outer membrane vesicles: structure, vesicle formation, separation and biotechnological applications. Microb. Cell Fact. 2025;24:27. doi: 10.1186/s12934-025-02653-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Karttunen J, Heiskanen M, Navarro-Ferrandis V, Das Gupta S, Lipponen A, Puhakka N, et al. Precipitation-based extracellular vesicle isolation from rat plasma co-precipitate vesicle-free microRNAs. J Extracell. Vesicles. 2019;8:1555410. doi: 10.1080/20013078.2018.1555410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Gamez-Valero A, Monguio-Tortajada M, Carreras-Planella L, Franquesa M, Beyer K, Borras FE. Size-exclusion chromatographybased isolation minimally alters extracellular vesicles' characteristics compared to precipitating agents. Sci. Rep. 2016;6:33641. doi: 10.1038/srep33641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kulp A, Kuehn MJ. Biological functions and biogenesis of secreted bacterial outer membrane vesicles. Annu. Rev. Microbiol. 2010;64:163–184. doi: 10.1146/annurev.micro.091208.073413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Le LHM, Steele JR, Ying L, Schittenhelm RB, Ferrero RL. A new isolation method for bacterial extracellular vesicles providing greater purity and improved proteomic detection of vesicle proteins. J. Extracell. Biol. 2023;2:e84. doi: 10.1002/jex2.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Linares R, Tan S, Gounou C, Arraud N, Brisson AR. High-speed centrifugation induces aggregation of extracellular vesicles. J. Extracell. Vesicles. 2015;4:29509.doal. doi: 10.3402/jev.v4.29509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Mol EA, Goumans MJ, Doevendans PA, Sluijter JPG, Vader P. Higher functionality of extracellular vesicles isolated using sizeexclusion chromatography compared to ultracentrifugation. Nanomedicine. 2017;13:2061–2065. doi: 10.1016/j.nano.2017.03.011. [DOI] [PubMed] [Google Scholar]
- 117.Bergqvist M, Lasser C, Crescitelli R, Park KS, Lotvall J. A Non-centrifugation method to concentrate and purify extracellular vesicles using superabsorbent polymer followed by size exclusion chromatography. J. Extracell. Vesicles. 2025;14:e70037. doi: 10.1002/jev2.70037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Karimi N, Cvjetkovic A, Jang SC, Crescitelli R, Hosseinpour Feizi MA, Nieuwland R, et al. Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell. Mol. Life Sci. 2018;75:2873–2886. doi: 10.1007/s00018-018-2773-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Patel GK, Khan MA, Zubair H, Srivastava SK, Khushman M, Singh S, et al. Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications. Sci. Rep. 2019;9:5335. doi: 10.1038/s41598-019-41800-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Welton JL, Webber JP, Botos LA, Jones M, Clayton A. Ready-made chromatography columns for extracellular vesicle isolation from plasma. J. Extracell. Vesicles. 2015;4:27269. doi: 10.3402/jev.v4.27269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Gyorgy B, Paloczi K, Balbisi M, Turiak L, Drahos L, Visnovitz T, et al. Effect of the 35 nm and 70 nm size exclusion chromatography (SEC) column and plasma storage time on separated extracellular vesicles. Curr. Issues Mol. Biol. 2024;46:4337–4357. doi: 10.3390/cimb46050264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Laakmann K, Eckersberg JM, Hapke M, Wiegand M, Bierwagen J, Beinborn I, et al. Bacterial extracellular vesicles repress the vascular protective factor RNase1 in human lung endothelial cells. Cell Commun. Signal. 2023;21:111. doi: 10.1186/s12964-023-01131-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Yang Y, Wang Y, Wei S, Zhou C, Yu J, Wang G, et al. Extracellular vesicles isolated by size-exclusion chromatography present suitability for RNomics analysis in plasma. J. Transl. Med. 2021;19:104. doi: 10.1186/s12967-021-02775-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Yang HC, Ham YM, Kim JA, Rhee WJ. Single-step equipment-free extracellular vesicle concentration using super absorbent polymer beads. J. Extracell. Vesicles. 2021;10:e12074. doi: 10.1002/jev2.12074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Musante L, Tataruch D, Gu D, Benito-Martin A, Calzaferri G, Aherne S, et al. A simplified method to recover urinary vesicles for clinical applications, and sample banking. Sci. Rep. 2014;4:7532. doi: 10.1038/srep07532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Li K, Wong DK, Hong KY, Raffai RL. Cushioned-density gradient ultracentrifugation (C-DGUC): a refined and high performance method for the isolation, characterization, and use of exosomes. Methods Mol. Biol. 2018;1740:69–83. doi: 10.1007/978-1-4939-7652-2_7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Desgeorges A, Hollerweger J, Lassacher T, Rohde E, Helmbrecht C, Gimona M. Differential fluorescence nanoparticle tracking analysis for enumeration of the extracellular vesicle content in mixed particulate solutions. Methods. 2020;177:67–73. doi: 10.1016/j.ymeth.2020.02.006. [DOI] [PubMed] [Google Scholar]
- 128.Chen YS, Lin EY, Chiou TW, Harn HJ. Exosomes in clinical trial and their production in compliance with good manufacturing practice. Tzu Chi. Med. J. 2020;32:113–120. doi: 10.4103/tcmj.tcmj_182_19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Momen-Heravi F, Balaj L, Alian S, Mantel PY, Halleck AE, Trachtenberg AJ, et al. Current methods for the isolation of extracellular vesicles. Biol. Chem. 2013;394:1253–1262. doi: 10.1515/hsz-2013-0141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Wang Z, Zhou X, Kong Q, He H, Sun J, Qiu W, et al. Extracellular vesicle preparation and analysis: a state-of-the-art review. Adv. Sci. (Weinh) 2024;11:e2401069. doi: 10.1002/advs.202401069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Yu LL, Zhu J, Liu JX, Jiang F, Ni WK, Qu LS, et al. A comparison of traditional and novel methods for the separation of exosomes from human samples. Biomed Res. Int. 2018;2018:3634563. doi: 10.1155/2018/3634563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546:498–503. doi: 10.1038/nature22341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Cantin R, Diou J, Belanger D, Tremblay AM, Gilbert C. Discrimination between exosomes and HIV-1: purification of both vesicles from cell-free supernatants. J. Immunol. Methods. 2008;338:21–30. doi: 10.1016/j.jim.2008.07.007. [DOI] [PubMed] [Google Scholar]
- 134.Parimon T, Garrett NE, 3rd, Chen P, Antes TJ. 2018. Isolation of extracellular vesicles from murine bronchoalveolar lavage fluid using an ultrafiltration centrifugation technique. J. Vis. Exp. 9:(141): 10.3791/58310. doi: 10.3791/58310. 10.3791/58310 [DOI] [PMC free article] [PubMed]
- 135.Chutkan H, Macdonald I, Manning A, Kuehn MJ. Quantitative and qualitative preparations of bacterial outer membrane vesicles. Methods Mol. Biol. 2013;966:259–272. doi: 10.1007/978-1-62703-245-2_16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Graham JM. Formation of self-generated gradients of iodixanol. ScientificWorldJournal. 2002;2:1356–1360. doi: 10.1100/tsw.2004.284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kobayashi H, Shiba T, Yoshida T, Bolidong D, Kato K, Sato Y, et al. Precise analysis of single small extracellular vesicles using flow cytometry. Sci. Rep. 2024;14:7465. doi: 10.1038/s41598-024-57974-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Woud WW, van der Pol E, Mul E, Hoogduijn MJ, Baan CC, Boer K, et al. An imaging flow cytometry-based methodology for the analysis of single extracellular vesicles in unprocessed human plasma. Commun. Biol. 2022;5:633. doi: 10.1038/s42003-022-03569-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.De Langhe N, Van Dorpe S, Guilbert N, Vander Cruyssen A, Roux Q, Deville S, et al. Mapping bacterial extracellular vesicle research: insights, best practices and knowledge gaps. Nat. Commun. 2024;15:9410. doi: 10.1038/s41467-024-53279-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Pirolli NH, Reus LSC, Mamczarz Z, Khan S, Bentley WE, Jay SM. High performance anion exchange chromatography purification of probiotic bacterial extracellular vesicles enhances purity and anti-inflammatory efficacy. Biotechnol. Bioeng. 2023;120:3368–3380. doi: 10.1002/bit.28522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Kim DK, Lee J, Kim SR, Choi DS, Yoon YJ, Kim JH, et al. EVpedia: a community web portal for extracellular vesicles research. Bioinformatics. 2015;31:933–939. doi: 10.1093/bioinformatics/btu741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Lee EY, Choi DS, Kim KP, Gho YS. Proteomics in gram-negative bacterial outer membrane vesicles. Mass Spectrom. Rev. 2008;27:535–555. doi: 10.1002/mas.20175. [DOI] [PubMed] [Google Scholar]
- 143.Benedikter BJ, Bouwman FG, Vajen T, Heinzmann ACA, Grauls G, Mariman EC, et al. Ultrafiltration combined with size exclusion chromatography efficiently isolates extracellular vesicles from cell culture media for compositional and functional studies. Sci. Rep. 2017;7:15297. doi: 10.1038/s41598-017-15717-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Corso G, Heusermann W, Trojer D, Gorgens A, Steib E, Voshol J, et al. Systematic characterization of extracellular vesicle sorting domains and quantification at the single molecule - single vesicle level by fluorescence correlation spectroscopy and single particle imaging. J. Extracell. Vesicles. 2019;8:1663043. doi: 10.1080/20013078.2019.1663043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Sabra W, Lunsdorf H, Zeng AP. Alterations in the formation of lipopolysaccharide and membrane vesicles on the surface of Pseudomonas aeruginosa PAO1 under oxygen stress conditions. Microbiology (Reading) 2003;149:2789–2795. doi: 10.1099/mic.0.26443-0. [DOI] [PubMed] [Google Scholar]
- 146.Ellis TN, Leiman SA, Kuehn MJ. Naturally produced outer membrane vesicles from Pseudomonas aeruginosa elicit a potent innate immune response via combined sensing of both lipopolysaccharide and protein components. Infect. Immun. 2010;78:3822–3831. doi: 10.1128/IAI.00433-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Jiang Y, Ma J, Long Y, Dan Y, Fang L, Wang Z. Extracellular membrane vesicles of Escherichia coli induce apoptosis of CT26 colon carcinoma cells. Microorganisms. 2024;12:1446. doi: 10.3390/microorganisms12071446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Jin JS, Kwon SO, Moon DC, Gurung M, Lee JH, Kim SI, et al. Acinetobacter baumannii secretes cytotoxic outer membrane protein A via outer membrane vesicles. PLoS One. 2011;6:e17027. doi: 10.1371/journal.pone.0017027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Bachurski D, Schuldner M, Nguyen PH, Malz A, Reiners KS, Grenzi PC, et al. Extracellular vesicle measurements with nanoparticle tracking analysis - an accuracy and repeatability comparison between NanoSight NS300 and ZetaView. J. Extracell. Vesicles. 2019;8:1596016. doi: 10.1080/20013078.2019.1596016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Keenan JI, Allardyce RA, Bagshaw PF. Dual silver staining to characterise Helicobacter spp. outer membrane components. J. Immunol. Methods. 1997;209:17–24. doi: 10.1016/S0022-1759(97)00141-5. [DOI] [PubMed] [Google Scholar]
- 151.Guarro M, Suner F, Lecina M, Borros S, Fornaguera C. Efficient extracellular vesicles freeze-dry method for direct formulations preparation and use. Colloids Surf. B Biointerfaces. 2022;218:112745. doi: 10.1016/j.colsurfb.2022.112745. [DOI] [PubMed] [Google Scholar]
- 152.Trenkenschuh E, Richter M, Heinrich E, Koch M, Fuhrmann G, Friess W. Enhancing the stabilization potential of lyophilization for extracellular vesicles. Adv. Healthc. Mater. 2022;11:e2100538. doi: 10.1002/adhm.202100538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Frank J, Richter M, de Rossi C, Lehr CM, Fuhrmann K, Fuhrmann G. Extracellular vesicles protect glucuronidase model enzymes during freeze-drying. Sci. Rep. 2018;8:12377. doi: 10.1038/s41598-018-30786-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Charoenviriyakul C, Takahashi Y, Nishikawa M, Takakura Y. Preservation of exosomes at room temperature using lyophilization. Int. J. Pharm. 2018;553:1–7. doi: 10.1016/j.ijpharm.2018.10.032. [DOI] [PubMed] [Google Scholar]
- 155.El Baradie KBY, Nouh M, O'Brien Iii F, Liu Y, Fulzele S, Eroglu A, et al. Freeze-dried extracellular vesicles from adiposederived stem cells prevent hypoxia-induced muscle cell injury. Front. Cell. Dev. Biol. 2020;8:181. doi: 10.3389/fcell.2020.00181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Wolf M, Poupardin RW, Ebner-Peking P, Andrade AC, Blochl C, Obermayer A, et al. A functional corona around extracellular vesicles enhances angiogenesis, skin regeneration and immunomodulation. J. Extracell. Vesicles. 2022;11:e12207. doi: 10.1002/jev2.12207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Xing J, Niu T, Yu T, Zou B, Shi C, Wang Y, et al. Faecalibacterium prausnitzii-derived outer membrane vesicles reprogram gut microbiota metabolism to alleviate Porcine Epidemic Diarrhea Virus infection. Microbiome. 2025;13:90. doi: 10.1186/s40168-025-02078-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Vuong N, Alomia M, Byne A, Gade P, Philipson TR, Alhammad RI, et al. Lactobacillus rhamnosus-derived extracellular vesicles influence calcium deposition in a model of breast cancer intraductal calcium stress. iScience. 2025;28:112538. doi: 10.1016/j.isci.2025.112538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Liang X, Dai N, Sheng K, Lu H, Wang J, Chen L, et al. Gut bacterial extracellular vesicles: important players in regulating intestinal microenvironment. Gut Microbes. 2022;14:2134689. doi: 10.1080/19490976.2022.2134689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Van Deun J, Mestdagh P, Sormunen R, Cocquyt V, Vermaelen K, Vandesompele J, et al. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J. Extracell. Vesicles. 2014;18:3. doi: 10.3402/jev.v3.24858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Chen G, Gao C, Jiang S, Cai Q, Li R, Sun Q, et al. Fusobacterium nucleatum outer membrane vesicles activate autophagy to promote oral cancer metastasis. J. Adv. Res. 2024;56:167–179. doi: 10.1016/j.jare.2023.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Fan R, Zhou Y, Chen X, Zhong X, He F, Peng W, et al. Porphyromonas gingivalis outer membrane vesicles promote apoptosis via msRNA-regulated DNA methylation in periodontitis. Microbiol. Spectr. 2023;11:e0328822. doi: 10.1128/spectrum.03288-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Goman A, Ize B, Jeannot K, Pin C, Payros D, Goursat C, et al. Uncovering a new family of conserved virulence factors that promote the production of host-damaging outer membrane vesicles in gram-negative bacteria. J. Extracell. Vesicles. 2025;14:e270032. doi: 10.1002/jev2.70032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Bierwagen J, Wiegand M, Laakmann K, Danov O, Limburg H, Herbel SM, et al. Bacterial vesicles block viral replication in macrophages via TLR4-TRIF-axis. Cell Commun. Signal. 2023;21:65. doi: 10.1186/s12964-023-01086-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Potapova A, Garvey W, Dahl P, Guo S, Chang Y, Schwechheimer C, et al. Outer membrane vesicles and the outer membrane protein OmpU govern Vibrio cholerae biofilm matrix assembly. mBio. 2024;15:e0330423. doi: 10.1128/mbio.03304-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Wang X, Thompson CD, Weidenmaier C, Lee JC. Release of Staphylococcus aureus extracellular vesicles and their application as a vaccine platform. Nat. Commun. 2018;9:1379. doi: 10.1038/s41467-018-03847-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Wagner T, Joshi B, Janice J, Askarian F, Skalko-Basnet N, Hagestad OC, et al. Enterococcus faecium produces membrane vesicles containing virulence factors and antimicrobial resistance related proteins. J. Proteomics. 2018;187:28–38. doi: 10.1016/j.jprot.2018.05.017. [DOI] [PubMed] [Google Scholar]
- 168.Tang M, Tian S, Chen K, Zhang Q, Lei Y, Tang T, et al. Membrane vesicles derived from Listeria monocytogenes might be a potential antigen delivery vector. Int. J. Pharm. 2023;644:123275. doi: 10.1016/j.ijpharm.2023.123275. [DOI] [PubMed] [Google Scholar]
- 169.Reimer SL, Beniac DR, Hiebert SL, Booth TF, Chong PM, Westmacott GR, et al. Comparative analysis of outer membrane vesicle isolation methods with an Escherichia coli tolA mutant reveals a hypervesiculating phenotype with outer-inner membrane vesicle content. Front. Microbiol. 2021;12:628801. doi: 10.3389/fmicb.2021.628801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Li Q, Ou Z, Lin J, Tang D, He B, Wu Y, et al. Specific labeling of outer membrane vesicles with antibiotic-conjugated probe reveals early bacterial infections in blood. Nat. Commun. 2025;16:3535. doi: 10.1038/s41467-025-58676-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Ma D, Zhang Y, Zhang J, Shi J, Gao S, Long F, et al. Outer membrane vesicles derived from probiotic Escherichia coli Nissle 1917 promote metabolic remodeling and M1 polarization of RAW264.7 macrophages. Front. Immunol. 2025;16:1501174. doi: 10.3389/fimmu.2025.1501174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Choi DS, Kim DK, Choi SJ, Lee J, Choi JP, Rho S, et al. Proteomic analysis of outer membrane vesicles derived from Pseudomonas aeruginosa. Proteomics. 2011;11:3424–3429. doi: 10.1002/pmic.201000212. [DOI] [PubMed] [Google Scholar]
- 173.Saberi S, Esmaeili M, Saghiri R, Shekari F, Mohammadi M. Assessment of the mixed origin of the gastric epithelial extracellular vesicles in acellular transfer of Helicobacter pylori toxins and a systematic review. Microb. Pathog. 2023;177:106024. doi: 10.1016/j.micpath.2023.106024. [DOI] [PubMed] [Google Scholar]
- 174.Rompikuntal PK, Vdovikova S, Duperthuy M, Johnson TL, Ahlund M, Lundmark R, et al. Outer membrane vesicle-mediated export of processed PrtV protease from Vibrio cholerae. PLoS One. 2015;10:e0134098. doi: 10.1371/journal.pone.0134098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Brown L, Kessler A, Cabezas-Sanchez P, Luque-Garcia JL, Casadevall A. Extracellular vesicles produced by the Gram-positive bacterium Bacillus subtilis are disrupted by the lipopeptide surfactin. Mol. Microbiol. 2014;93:183–198. doi: 10.1111/mmi.12650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Biagini M, Garibaldi M, Aprea S, Pezzicoli A, Doro F, Becherelli M, et al. The human pathogen Streptococcus pyogenes releases lipoproteins as lipoprotein-rich membrane vesicles. Mol. Cell. Proteomics. 2015;14:2138–2149. doi: 10.1074/mcp.M114.045880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Resch U, Tsatsaronis JA, Le Rhun A, Stubiger G, Rohde M, Kasvandik S, et al. A two-component regulatory system impacts extracellular membrane-derived vesicle production in group A Streptococcus. mBio. 2016;7:e00207–16. doi: 10.1128/mBio.00207-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Lee J, Kim SH, Choi DS, Lee JS, Kim DK, Go G, et al. Proteomic analysis of extracellular vesicles derived from Mycobacterium tuberculosis. Proteomics. 2015;15:3331–3337. doi: 10.1002/pmic.201500037. [DOI] [PubMed] [Google Scholar]



