Download PDF
Review  |  Open Access  |  12 Sep 2024

Role of probiotic extracellular vesicles in inter-kingdom communication and current technical limitations in advancing their therapeutic utility

Views: 331 |  Downloads: 63 |  Cited:  0
Extracell Vesicles Circ Nucleic Acids 2024;5:609-26.
10.20517/evcna.2024.39 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

Diverse functions of probiotic extracellular vesicles (EVs) have been extensively studied over the past decade, proposing their role in inter-kingdom communication. Studies have explored their therapeutic role in pathophysiological processes ranging from cancer, immunoregulation, and ulcerative colitis to stress-induced depression. These studies have highlighted the significant and novel potential of probiotic EVs for therapeutic applications, offering immense promise in addressing several unmet clinical needs. Additionally, probiotic EVs are being explored as vehicles for targeted delivery approaches. However, the realization of clinical utility of probiotic EVs is hindered by several knowledge gaps, pitfalls, limitations, and challenges, which impede their wider acceptance by the scientific community. Among these, limited knowledge of EV biogenesis, markers and regulators in bacteria, variations in cargo due to culture conditions or EV isolation method, and lack of proper understanding of gut uptake and demonstration of in vivo effect are some important issues. This review aims to summarize the diverse roles of probiotic EVs in health and disease conditions. More importantly, it discusses the significant knowledge gaps and limitations that stand in the way of the therapeutic utility of probiotic EVs. Furthermore, the importance of addressing these gaps and limitations with technical advances such as rigorous omics has been discussed.

Keywords

Extracellular vesicles, probiotic extracellular vesicles, bacterial extracellular vesicles, outer membrane vesicles, membrane vesicles, species crosstalk, inter-kingdom communication

INTRODUCTION

Probiotics are live microorganisms that confer beneficial effects on the host, typically by mediating changes to the gut and intestinal microbiota[1]. Over the last few decades, their popularity and consumption have surged due to reported benefits[2]. Additionally, probiotics have been shown to positively impact interactions within the gut-brain axis[3]. Changes in microbiota composition have also been associated with various diseases, including inflammatory bowel disease (IBD), obesity, and irritable bowel syndrome (IBS)[4-6]. However, there are conflicting reports on the clinical efficacy of supplemented probiotics based on the pre-microbiota of patients[7], as well as in murine models of colitis[8]. To address these discrepancies, future research must further investigate the complex factors influencing individual responses, such as gut microbiota composition, diet, and age. Regardless, the implication of altered microbiota during disease progression has spurred interest in exploring the therapeutic potential of probiotic strains. Recent studies have demonstrated that the administration of specific probiotic species can elicit beneficial responses in models of colitis, IBS, and colorectal tumorigenesis[9-11]. Therefore, identifying the specific factors most responsible for these observed changes has become an area of intrigue. Given the importance of intercellular communication in maintaining microbiome symbiosis, probiotic extracellular vesicles (EVs) are thought to play a role in mediating these benefits[12]. The well-established role of EVs in intercellular communication and their reported benefits in complex disease models make them a promising area of study. Thus, recent investigations have expanded to explore the role of probiotic EVs in a wider range of functions by influencing the surrounding gut microbiota[13].

EVs have been generally defined as lipid bilayer structures that contain membrane proteins and a diverse range of cargo, including proteins, metabolites, DNA, and RNA[14]. Most EV subtypes range in size from 30 nm to 10 µm and are typically classified based on distinct characteristics such as biogenesis, size, content, and cell of origin[15]. Gram-positive and Gram-negative bacteria release distinct types of EVs, generally smaller than 500 nm, termed membrane vesicles (MVs) and outer membrane vesicles (OMVs), respectively[16,17]. MVs consist of a lipid bilayer enclosing cytoplasmic material, while OMVs are enclosed by the outer membrane and, therefore, also contain periplasmic material in addition to cytoplasmic contents. Additionally, the overall surface and embedded membrane proteins are distinct between these two types of EVs[18]. Despite this, the mechanisms and regulation of EV biogenesis in bacteria remain poorly understood[19]. Consequently, the lack of reliable EV markers hinders thorough characterization in the bacterial EV field[20]. Notably, it has been observed that the DNA/RNA ratio in probiotic-derived EVs can vary significantly among different probiotic species[21]. Identifying bacterial EV markers will likely lead to improved purity and characterization, potentially enabling researchers to pinpoint specific subtypes responsible for particular effects. Therefore, in this review, the term “probiotic EVs” will be employed to refer to vesicles from various sources, given the current lack of complete understanding and characterization.

Although probiotic EVs are biochemically lipid bilayers encapsulating a diverse range of biomolecules, they may possess several unique attributes that distinguish them from other EVs. A recent study characterized EVs from Lactobacillus species, revealing an enrichment of proteins with a serine-rich motif[22]. Additionally, EVs secreted by Lactobacillus species were found to be rich in proteins and enzymes involved in metabolic processes such as gluconeogenesis, glycolysis, amino acid and carbohydrate metabolism[22-24]. This enrichment suggests that probiotic bacteria may employ EVs as conduits to convey systemic benefits or regulate pathophysiological processes. For instance, studies have shown that Lactobacillus species secrete EVs enriched with antimicrobials, proteases, and proteins p40 and p75, which have been identified as mediators of probiotic effects[22,25-27]. Further research could uncover additional properties of probiotic EVs that differentiate them from other EVs.

THE PATHOPHYSIOLOGICAL ROLE OF PROBIOTIC EVs THROUGH INTER-KINGDOM AND SPECIES CROSSTALK

Probiotics have been shown to elicit a wide range of beneficial effects in various disease states, including cancer, IBS, and upper respiratory tract infections[28-30]. However, the underlying mechanisms of how probiotics mediate these effects remain poorly understood. Recently, EVs derived from probiotic strains have been implicated in important functional roles in pathophysiology [Figure 1]. Several studies have presented evidence that probiotic EVs laden with a variety of functional cargos may be responsible for initiating or mediating crucial signaling events that can alleviate disease conditions [Table 1].

Role of probiotic extracellular vesicles in inter-kingdom communication and current technical limitations in advancing their therapeutic utility

Figure 1. Implicated beneficial effects of probiotic extracellular vesicles in pathophysiological conditions. Schematic representation of the implicated roles that probiotic-derived EVs play in a plethora of pathophysiological processes. Several studies so far have demonstrated the potential of probiotic EVs in modulating the immune system and alleviating pathological conditions such as IBD, diabetes, and ulcerative colitis. The suggested health benefits of probiotic EVs range from improving gut health to inducing chemosensitivity and apoptosis in resistant cancer cells. Their role in several other diseases such as asthma, metabolic disorders including diabetes and obesity and their antimicrobial properties have been investigated in several studies. Table 1 provides a detailed summary of studies where the role of probiotic EVs in pathophysiology and disease alleviation has been suggested. IBD: Inflammatory bowel disease; EVs: extracellular vesicles; HIV: human immunodeficiency virus.

Table 1

Role of probiotic EVs in pathophysiology

Source of EVsFunction observedModelFunctional cargoReference
Antimicrobial activity
Lactobacillus acidophilusBacteriocin delivery and antimicrobial activityIn vitroNot stated[26]
Bacteroides thetaiotaomicron, Enterobacter cloacae, Lactobacillus acidophilusAltered EV cargo, biogenesis, size and quantityIn vitroNot stated[31]
Lactobacillus paracaseiPEGylated MoS2-ZnO containing EVs used as antimicrobial agents to restrict the growth of Staphylococcus aureusIn vitroNot stated[32]
Lactobacillus plantarum WCFS1EVs promoted expression of host defense genes and inhibited growth of vancomycin-resistant Enterococcus faeciumIn vivoNot stated[33]
Lactobacillus plantarumEVs reduced bacterial levels of Shewanella putrefaciens and maintained better food quality, increasing shelf lifeEx vivoNot stated[34]
Lactobacillus crispatus BC5, Lactobacillus gasseri BC12Probiotic EVs maintained healthy vaginal homeostasis by supporting colonization of beneficial bacterial species and preventing attachment of opportunistic pathogensIn vitroNot stated[35]
Lactobacillus plantarumProbiotic EVs displayed protective effects in atopic dermatitis induced by Staphylococcus aureus EVsIn vivoNot stated[36]
Antiviral activity
Staphylococcus aureus, Gardnerella vaginalis, Enterococcus faecium, Enterococcus faecalisEVs prevented HIV-1 infection in tissues ex vivo by preventing virus-cell receptor interactionEx vivoNot stated[37]
Lactobacillus crispatus BC3 and Lactobacillus gasseri BC12Probiotic EVs prevented HIV-1 infection in cervico-vaginal and tonsillar tissues ex vivoEx vivoNot stated[38]
Immunomodulatory effects
Lactobacillus sakei subsp. sakei NBRC 15893Enhanced IgA production in Peyer’s patch cells in response to membrane vesiclesIn vivoNot stated[39,40]
Lactobacillus rhamnosus GG and Lactobacillus reuteri DSM 17938Lactobacilli MVs dampen pro-inflammatory cytokine responses in a monocyte-dependent manner.In vitroNot stated[41]
Lactobacillus plantarumRegulation of pro and anti-inflammatory responses; innate and acquired. Enhanced IgA production in Peyer’s patch cellsIn vitroN-acylated peptides from lipoprotein19180[42]
Escherichia coli Nissle 1917E. coli Nissle 1917 enhanced secretion levels of pro- and anti-inflammatory cytokines in monocyte-derived dendritic cellsIn vitroNot stated[43]
Lactobacillus plantarumEVs promoted differentiation of human monocytic THP1 cells toward an anti-inflammatory M2 phenotypeIn vitroNot stated[44]
Lactobacillus reuteri BBC3EVs maintained intestinal immune homeostasis against LPS-induced inflammatory responsesIn vivoNot stated[45]
Lactobacillus caseiEVs exhibited anti-inflammatory immunomodulatory effects in intestinal epithelial cellsIn vitroNot stated[46]
Bifidobacterium longum, Clostridium butyricum, Lactobacillus plantarum WCFS1EVs stimulated the innate immune system and exhibited adjuvant-like propertiesIn vitroNot stated[47]
Bifidobacterium longum and Lactobacillus plantarum WCFS1Immune cells produced pro-inflammatory cytokines in response to EVsIn vitroNot stated[48]
Lactobacillus strains (L. rhamnosus NutRes 1, L. plantarum NutRes 8 and L. caseï CNCM I-1518) and Bifidobacterium strains (B. longum NutRes 266, B. breve NutRes 200 and B. animalis DN173010)Effect of EVs (TLR2 activity and phagocytosis ability) characterized on immune cellsIn vitroNot stated[49]
Lactobacillus plantarumpH-based stimulation of bacterial cells led to secretion of EVs with enhanced anti-inflammatory propertiesIn vitroNot stated[50]
Lactobacillus johnsonii N6.2EVs led to antibody and immune response generationIn vivoNot stated[51]
Escherichia coli Nissle 1917EVs caused RAW 264.7 macrophages to have increased phagocytosis, proliferation, and shifted their phenotype to more anti-inflammatoryIn vitroNot stated[52]
Bifidobacterium longumEVs induced apoptosis of mast cells. IP injection of ESBP led to reduced diarrhea occurrence and abundance of mast cellsIn vivoESBP[53]
Propionibacterium freudenreichii CIRM-BIA 129EVs exhibited anti-inflammatory effect by modulation of the NF-κB pathway in a dose-dependent manner, partly but not completely dependent on surface proteinsIn vitroNot stated[54]
Leuconostoc mesenteroides, Latilactobacillus curvatus, and Lactiplantibacillus plantarumEVs exerted anti-inflammatory effects on microglial cells and macrophagesIn vivoNot stated[55]
Escherichia coli Nissle 1917NOD1 signaling activated by EVs in intestinal epithelial cellsIn vitroNot stated[56]
Escherichia coli Nissle 1917EV-treated macrophages exhibited anti-inflammatory effects and increased anti-bacterial activityIn vitroNot stated[52]
Lacticaseibacillus rhamnosus JB-1EVs led to immunomodulatory effects in dendritic cells mediated by TLR2In vitroLipoteichoic acid[57]
Lactobacillus, Bifidobacterium, and LactococcusProbiotic EV administration post hepatic surgery reduced adhesion molecule expression and immune cell invasion in liver and contributed to improved livery recoveryIn vivoNot stated[58]
Escherichia coli Nissle 1917EVs treatment led to an altered miRNA profile in dendritic cellsIn vitroNot stated[59]
Bacillus amyloliquefaciens SC06Probiotic EV-treated porcine intestinal epithelial cells shed EVs, which led to improved macrophage functionIn vitroNot stated[60]
Gut health
Lactobacillus plantarum Q7EVs induced a shift in the gut microbiota to a more anti-inflammatory community, reduction of pro-inflammatory cytokine expression in colon tissue, and alleviated DSS-induced colitis symptomsIn vivoNot stated[61]
Lactobacillus rhamnosus GGEVs modulate gut microbiota and attenuated inflammation and ulcerative colitisIn vivoNot stated[62]
Lactobacillus paracaseiEV-mediated anti-inflammatory effects maintained gut health in response to LPS treatment and provided protection against ulcerative colitisIn vivoNot stated[63]
Clostridium butyricumEVs protected against DSS-induced colitis by regulating the repolarization of M2-macrophages and remodeling gut microbiotaIn vivoNot stated[64]
Lactobacillus kefirgranum PRCC-1301EVs exhibited anit-inflammatory effects by inhibiting the NF-κB pathway and improving intestinal barrier functionIn vivoNot stated[65]
Lactobacillus kefir KCTC 3611, L. kefiranofaciens KCTC 5075, and L. kefirgranum KCTC 5086EVs exhibited anti-inflammatory effects and alleviated IBD symptomsIn vivoNot stated[66]
Lactobacillus reuteri DSM-17938EVs mediate gut motility effectIn vivoNot stated[67]
Escherichia coli Nissle 1917Modulation of intestinal tight junctions that were disrupted via enteropathogenic E. coli. Specifically, by maintaining important tight junction proteins and preventing F-actin disorganizationIn vivoNot stated[68]
Clostridium butyricumEVs alleviated colitis symptoms, improved gut barrier integrity, and restored gut microbiota homeostasisIn vivoNot stated[69]
Escherichia coli Nissle 1917EVs exhibited gut barrier protective effects in enteropathogenic E. coli-infected intestinal epithelial cellsIn vitroNot stated[68]
Clostridium butyricumEVs demonstrated anti-inflammatory effects, alleviated bacterial dysbiosis, reduced abundance of pathogens, improved gut barrier integrity, and regulated metabolism of gut microbiota in colitis miceIn vivoNot stated[70]
Lactobacillus plantarumEngineered EVs loaded with fucoxanthin had beneficial effects in colitis mice as they polarized macrophages and alleviated colitis symptoms and bacterial dysbiosisIn vivoNot stated[71]
Cancer
Lactobacillus plantarumEVs restored chemosensitivity in 5-FU resistant colorectal cancer cellsIn vitroNot stated[72]
Lactobacillus rhamnosus GGEVs inhibited growth of colorectal cancer cellsIn vitroNot stated[73]
Lactobacillus rhamnosus GGEVs inhibited hepatic cancer cell growth and induced apoptosisIn vitroNot stated[74]
Lacticaseibacillus paracaseiEVs had an anti-tumor effect in vitro on colorectal cancer cell proliferation, invasion and migration and promoted apoptosis in vitro and in vivoIn vivoNot stated[75]
Other diseases/pathophysiology
Akkermansia muciniphilaAdministration of A.muciniphila EVs decreased weight, improved glucose tolerance, and enhanced tight junction function in a HFD-induced mouse model. This treatment also improved gut permeability in HFD miceIn vivoNot stated[76]
Akkermansia muciniphilaA. muciniphila EVs prevented gut dysbiosis in a high-fat diet-induced mouse model. They also reduced the levels of pro-inflammatory cytokines in plasma, reduced food intake, and decreased the expression of genes involved in lipid metabolism and inflammationIn vivoNot stated[77]
Akkermansia muciniphilaEVs ameliorated HFD-induced obesity, and improved the intestinal barrier integrity, inflammation, energy balance, and blood parametersIn vivoNot stated[78]
Lactobacillus plantarumEVs inhibited neuron apoptosis and protected against ischemic brain injuryIn vivoNot stated[79]
Lactobacillus plantarum, Akkermansia muciniphila, Bacillus subtilisEV treatment led to anti-depressive effects and alleviated stress-induced depressive behavior in miceIn vivoNot stated[80]
Lactobacillus plantarumEVs displayed anti-depressant activity by rescuing reduced expression of BDNF and inhibited stress-induced depressive-like behaviorIn vivoNot stated[81]
Lactobacillus animalisEVs prevented the development of glucocorticoid-induced ONFHIn vivoNot stated[82]
Lactobacillus reuteriNo effect of EVs on osteoclastogenesisIn vitroNot stated[83]
Lactobacillus druckeriiEVs inhibited hypertrophic scar fibrosisIn vivoNot stated[84]
Lactobacillus plantarum and Lactobacillus caseiEVs from both species were coupled with microparticles; these bacteriomimetics were then embedded into a hydrogel mixture; the resulting combination improved wound healingIn vivoNot stated[85]
Lactobacillus plantarumEVs regulated extracellular matrix-related genes and suppressed wrinkle formation and pigmentation in womenIn vivoNot stated[86]
Lactobacillus casei BL23EGFR pathway stimulation by EVsIn vitroP40 and P75[87]
Akkermansia muciniphilaEVs caused the regression of hepatic stellate cell activation in a liver injury mouse model. They also ameliorated levels of inflammatory cytokines, improved liver histopathological damages, and reduced the expression of fibrosis biomarkersIn vivoNot stated[88]
Akkermansia muciniphilaOral administration of A. muciniphila EVs improved intestinal integrity and anti-inflammatory responses in a mouse model of liver injuryIn vivoNot stated[89]
Lactococcus lactisEVs exhibited an immunoregulatory effect on airway inflammation in allergic asthma by activating dendritic cellsIn vivoNot stated[90]
Leuconostoc holzapfeliiEVs promoted proliferation, migration and regulated cell cycle in human hair follicle dermal papilla cellsIn vitroNot stated[91]
Akkermansia muciniphila and Faecalibacterium prausnitziiTreatment with EVs led to increased serotonin production in intestinal epithelial cellsIn vitroNot stated[92]

The utility of probiotic EVs as potential antimicrobial and antiviral treatments

The current understanding that the administration of probiotic EVs can impart beneficial effects during disease has sparked interest in their potential role in cross-species and inter-kingdom communication. Recent research has highlighted the antimicrobial activity of EVs derived from probiotic sources[34-36,38]. Interestingly, it has been shown that EVs isolated from L. plantarum can reduce the levels of S. putrefaciens, a bacterium indicative of rotting fish[34]. Tuna fish stored with L. plantarum EVs had an increased shelf life and overall food quality[34]. In another recent study, the ability of bioengineered EVs from L. paracasei to target and restrict S. aureus growth was demonstrated, suggesting their utility as nano antibiotics[32]. These studies clearly highlight the potential of probiotic EVs in inter-kingdom communication. Thus, recent research has also attempted to explore the possible interactions between probiotic EVs and the host cells in various diseases and health conditions. For instance, L. plantarum EVs were able to modify host cell defenses, offering increased protection against antibiotic-resistant bacterial pathogens[33]. In this regard, a better understanding of probiotic EV cargo and the associated signaling with host cells may reveal avenues for improved treatment of various diseases. In the context of antiviral activity, one study exhibited that EVs originating from symbiotic vaginal Lactobacilli species had the ability to reduce human immunodeficiency virus-1 (HIV-1) infection[38]. Human vaginal and tonsillar tissues were infected with HIV-1 ex vivo and then treated with EVs derived from L. crispatus and L. gasseri strains. EV treatment provided protection against HIV-1 attachment and entry due to reduced exposure to envelope glycoprotein (Env), a viral envelope protein crucial for virus-host cell interaction.

The role of probiotic EVs in various disease states and the associated immunomodulatory effects

The immunomodulatory effects of probiotic EVs are their most well-characterized attribute so far [Figure 2]. Probiotic EVs from several bacterial genera, including Lactobacillus, Escherichia, and Bifidobacterium, have been associated with phenotypic changes in various immune cells[44,49,59]. EVs from L. plantarum promoted the differentiation of human leukemia monocytes toward an M2 phenotype, which is characterized by anti-inflammatory properties[44]. Moreover, probiotic EVs can also prime immune cells to mount a pro-inflammatory response in vitro. Dendritic cells treated with EVs derived from E. coli Nissle 1917, a widely studied probiotic, exhibited changes in their miRNA profile[59]. As a result, dendritic cells produced more Th1-type cytokines important for managing pathogens. Evidently, probiotic EVs can communicate with and influence a wide range of immune cells, stimulating beneficial phenotypic changes [Figure 2]. However, there have been several studies where their role in mediating a direct phenotype in disease conditions such as ulcerative colitis and IBD has been reported [Table 1].

Role of probiotic extracellular vesicles in inter-kingdom communication and current technical limitations in advancing their therapeutic utility

Figure 2. Immune regulatory effects of probiotic EVs. Schematic diagram depicting the various immunomodulatory roles of probiotic EVs, including alteration to cytokine release, activation of immune cells, surface interactions, and changes to antibody production. Table 1 provides a comprehensive summary of the literature suggesting a functional role for probiotic EVs in the immune setting. EVs: Extracellular vesicles; IgA: immunoglobulin A; TRL: toll-like receptor; LPS: lipopolysaccharide; NF-κβ: nuclear factor kappa β.

The intestinal epithelium provides a crucial physical barrier while allowing intestinal epithelial cells (IECs) to participate in crosstalk between the gut microbiota and systemic immune system[93]. There is growing evidence to support the beneficial role of probiotic EVs in modulating gut health by communicating with IECs [Table 1]. Several species of Lactobacilli have been shown to promote an anti-inflammatory intestinal phenotype in an ulcerative colitis mouse model[65,68,94], as well as reducing the expression of various pro-inflammatory cytokines. Additionally, there have been studies suggesting that alterations to DNA amount and specific immunostimulatory oligodeoxynucleotides within probiotic EVs can contribute to stronger immune activation[21,95]. Furthermore, EVs from probiotic sources have demonstrated the ability to improve gut barrier function. One study has shown that following a loss of gut barrier integrity induced by E. coli, EVs derived from E. coli Nissle 1917 improved gut barrier function[68]. Comparatively, this reduced permeability was associated with maintaining ZO-1 at tight junctions and conserving occludin and claudin-14 mRNA levels. Similarly, another study has revealed that L. kefirgranum EVs originating from kefir grain were able to conserve tight junctions and epithelial cell integrity[65]. Given this, the reported beneficial effects of probiotics EVs are not limited to solely immune modulation and gut health. Emerging evidence has demonstrated the ability of probiotic EVs to disturb detrimental signaling pathways involved in cancer, obesity, liver injury, and even depression[75,76,80,88]. A deeper understanding of the precise mechanisms and EV-associated proteins involved in pathogenic and host cell communication is required within the field of probiotic EVs. Furthermore, addressing the challenges and limitations surrounding probiotic EV application is paramount for quality future research. This improved knowledge may allow for therapeutic interventions in disease, potentially leading to prevention and/or improved treatment.

Findings in these preclinical studies present an exemplary case for the clinical utility of probiotic EVs in therapeutic applications. Several clinical studies recently have investigated the utility of pathogen-derived OMVs in designing vaccines against viral and bacterial infections[96]. Furthermore, there are clinical studies investigating the potential benefits of probiotics with promising preliminary outcomes[97-99]. However, to date, there are no clinical studies investigating the role of probiotic extracellular vesicles in combating infectious or non-infectious diseases. There are several technical limitations and challenges leading to knowledge gaps that have hindered the clinical utility of probiotic EVs.

CHALLENGES AND LIMITATIONS TO EMPLOYING PROBIOTIC EVs IN CLINICAL APPLICATIONS

EVs secreted by probiotic and commensal microbiota and derived from dietary sources have been proposed to be mediators of cross-species and inter-kingdom communication[13,100,101]. Their ability to signal in complex pathophysiological events enables them to dictate phenotype in a range of health and disease conditions [Table 1]. Bovine milk extracellular vesicles (MEV) and probiotic EVs have previously been examined for their stability and resilience to withstand harsh, degrading gut conditions, assessing their oral bioavailability and were able to reach peripheral tissues[102-104]. They were reported to withstand boiling temperatures and acidification, supporting their role as vehicles that sequester bioactive cargo in its native, functional state upon exposure to extreme conditions[104]. Similarly, in other studies, it has been reported that regular industrial processing such as heat treatment could harm the integrity and molecular composition of MEVs. However, the extent of this damage is unclear as MEVs were recovered in high abundance from milk despite the harsh processing and their surface markers could still be detected upon molecular analysis[104-107]. Furthermore, EVs from dietary sources or gut bacteria have been speculated to cross the gut barrier through either transendocytosis or paracellular translocation, entering systemic circulation and reaching peripheral tissues[23,51,102,108-110]. Although the aforementioned evidence supports the function of probiotic EVs in pathophysiology, they are not entirely convincing as several knowledge gaps persist, serving as hindrances to the application of probiotic EVs in therapy [Figure 3][111,112]. Contradictory findings have led to varying schools of thought about the potential and utility of probiotic EVs, making it imperative to address these knowledge gaps, challenges, and limitations for further advancement of the field[111].

Role of probiotic extracellular vesicles in inter-kingdom communication and current technical limitations in advancing their therapeutic utility

Figure 3. Current challenges and technical limitations hindering the therapeutic utility of probiotic EVs. Schematic illustration to depict challenges and limitations facing the utility of probiotic EVs in clinical applications. Several technical constraints such as EV cargo and yield variations due to different isolation methods, the presence of secretory factors and contaminants in the preparation, and the use of inappropriate preclinical models to study the effect and variation in phenotype based on the route of administration could prevent a detailed understanding and widespread acceptance of the role of probiotic EVs in pathophysiology. Additionally, current technical limitations prevent a better understanding of the EV cargo component responsible for the observed phenotypes. The annotated bibliography is shown in the Supplementary Materials. EVs: Extracellular vesicles.

EV biogenesis mechanisms, cargo regulation and isolation

EV biogenesis and secretion is conserved across all domains of life, with this process having only recently been identified in bacteria[19,113]. Although during the last decade, there has been a notable rise in interest in deciphering functions of probiotic EVs in inter-kingdom communication [Table 1], several gaps in our understanding of EV biogenesis in bacteria still persist[19]. Recent research has revealed that bacterial EV secretion occurs not only through membrane blebbing but also via explosive lysis[114,115]. Furthermore, several internal and external factors that could regulate or stimulate bacterial EV biogenesis and secretion are still being explored[19]. Despite the limited research on MV biogenesis and its regulators[19], it is known that variations in environmental factors such as nutrients, pH, or exposure to antibiotics can result in significant variations in EV yield, cargo, and function[31,101,116,117]. For instance, oxygen exposure and oxidative stress, as environmental factors, have been linked to changes in bacterial EV cargo[118]. Lactobacillus species in another study were observed to secrete EVs with slightly varying bacteriocin activity[26]. Therefore, environmental conditions may lead to variations in EV cargo both in vivo and in vitro, leading to varying phenotypic observations across studies. Moreover, co-cultivation of two different probiotic strains has been shown to elevate the bioactivity of EVs by increasing the secretion of interleukins from peripheral blood mononuclear cells[119]. Moreover, studies have highlighted the effect of EV isolation methods on its yield and cargo, demonstrating varying cargo and function in EVs isolated from the same source using different methods[120,121]. Evidently, improved knowledge of EV biogenesis, mechanisms at play, and regulators prior to their functional characterization is key to better understanding their function. Additionally, standardizing and optimizing the isolation method and providing detailed reporting are essential to ensure more reproducibility between groups, facilitating a better understanding of diverse EV roles.

Bacterial EV markers and characterization

Mammalian EV characterization includes confirming the presence of luminal and surface EV markers[122]. A partial understanding of EV biogenesis and other challenges, such as environmental conditions and their effect on EV cargo, along with the magnitude of species being studied, has so far prevented the community from proposing marker proteins to characterize bacterial EVs[20]. Therefore, in the absence of such makers, it is imperative to perform a thorough characterization based on particle size and morphology to ensure successful EV isolation[121,122]. Nevertheless, there is a need to perform additional rigorous high throughput omics to study EV cargo, enabling an enhanced understanding of conserved constituents and regulators of biogenesis and secretion[123].

Collective community efforts may allow for the identification of EV markers for probiotic strains and thus facilitate improved characterization. For instance, comparing protein cargo identified with proteomic analysis of EVs isolated from different species of probiotic bacteria leads to the identification of several conserved protein cargos between species and strains. Interestingly, enolase was one such protein consistently identified in EVs of all the bacterial species compared. Other proteins such as phosphoglycerate kinase, glucose-6-phosphate isomerase, glyceraldehyde-3-phosphate dehydrogenase A, and adenosine triphosphate (ATP) synthase subunits were also conserved between species [Figure 4].

Role of probiotic extracellular vesicles in inter-kingdom communication and current technical limitations in advancing their therapeutic utility

Figure 4. Omics approaches to thoroughly characterize probiotic bacterial EV cargo could aid in the identification of conserved cargo and lead to the identification of EV-enriched proteins conserved between strains and species. Venn diagrams depicting common and unique cargo in EVs of various species and strains of probiotic bacteria. Venn diagrams were generated using FunRich software (version 3.1.4)[129]. EVs: Extracellular vesicles.

EV purity

Bacterial cells actively secrete proteins and other factors in their surroundings, and several of these secretory factors could also be shed via EVs[124,125]. Consequently, the purity of EV fraction is of prime importance. While standardizing and optimizing methods for EV isolation, careful consideration is required to ensure a final EV preparation devoid of significant soluble or secretory factors. The presence of contaminants could be detrimental to reliable functional characterization[121]. For instance, the anti-cancer and apoptotic effect of cell-free supernatants from several probiotic strains has been reported[126,127]. More recently, EVs shed by probiotic strains were observed to contribute to this effect[72-75]. However, whether the effect is solely due to EVs in the conditioned medium or stems from other factors secreted by bacteria remains unknown. In this regard, Streptococcus thermophilus was reported to have a prophylactic effect in colorectal cancer development due to secreted β-galactosidase[10]. However, it has also been reported that bacterial EVs preferentially package acidic glycosidases and proteases, including β-galactosidase[128]. Thus, in addition to ensuring maximum purity of EV preparation, further experimental controls, such as a conditioned medium devoid of EVs, are needed to confidently assign a functional role to probiotic EVs.

Another aspect of EV purity to consider is to determine the subtype of EV responsible for the observed phenotype in vitro and in vivo. As previously discussed in the EV biogenesis section, bacterial EV biogenesis may occur through multiple distinct pathways. Therefore, when isolating probiotic EVs, several EV subpopulations may be present in the preparation. Thus, addressing these challenges and limitations with technical advancements is imperative to understand biogenesis pathways and distinguish EV subtypes. This may further enable a thorough understanding of the observed phenotypes and help to attribute them to specific EV subtypes, which is essential for clinical utility.

Biocompatibility

EV purity is also essential to ensure biocompatibility. The presence of immunogenic contaminants may lead to septic reactions upon systemic administration[130,131]. Bacterial toxins may have beneficial roles for the host in the organism’s native environment[132]. The biodistribution and bioavailability of EVs can change significantly when alternative routes of administration are used[133,134]. Thus, several contaminants or even cargo constituents could have a detrimental impact on host physiology as opposed to a beneficial effect observed in vitro. This also requires a thorough understanding of the cargo itself and the effects on host physiology prior to administration[135-138]. In this regard, engineering the parent bacterium to synthesize EVs devoid of toxic cargo is an attractive alternative[139]. However, efforts to knock out a cargo constituent in the parent bacterium could lead to non-related off-target effects, potentially altering EV cargo significantly and leading to a partial or complete loss of desired function[140]. Thus, depending on the route of administration of EVs, which impacts their bioavailability, biodistribution, systemic uptake, retention, clearance, and, in turn, their function, addressing the biocompatibility of EVs requires thoughtful evaluation to support their therapeutic utility[133,134].

Bioavailability and biodistribution

Probiotic EVs are naturally secreted by microbiota in the gastrointestinal tract, and their local uptake and function have been demonstrated widely in vivo in their ability to alleviate colitis symptoms and gut dysbiosis and enhance barrier integrity upon oral administration [Table 1]. Mechanisms facilitating uptake and systemic bioavailability of EVs upon oral administration are still poorly understood[111]. Recently, transcytosis of probiotic EVs in IECs has been demonstrated[23,110]. Further, elevated levels of EVs in circulation following administration of probiotic strain were observed[102]. Several studies have even demonstrated systemic function following oral administration of EVs[78,104]. However, a limited understanding of the mechanisms involved in vivo prevents wider acceptance[111]. Furthermore, plausible effects of probiotic EVs due to modulation of gut and systemic immunity and gut microbiota, thus resulting in the systemic phenotype indirectly must be examined[78,111].

CONCLUSION AND PROPOSALS

Despite their immense potential, several challenges pertaining to the acceptance and utility of probiotic EVs in clinical applications exist. The transition of probiotic EVs from bench to bedside relies on further technical developments in a bid to address the underlying limitations and knowledge gaps. Technical limitations and challenges were enlisted and discussed in the previous sections, along with proposals to address them. Further to these, a major question that remains unanswered is the delineation of EV cargo responsible for the desired effects. Most studies demonstrating the function of probiotic EVs did not link the effect to any particular cargo component [Table 1]. Comprehensive omics approaches are key in delineating the functional EV cargo and need to be prioritized to gain further momentum in this direction[141]. While doing so, it is also important to acknowledge that probiotic EVs contain diverse and rich cargos and several constraints remain to delineate the cargos responsible for the observed effects [Figure 3]. For instance, knockout, knockdown, or overexpression of a molecule of interest can have profound effects on EV cargo loading, biogenesis, and even uptake by the recipient, leading to a loss of desired function unrelated to the molecule being examined[104]. Lastly, the need to choose appropriate preclinical models is of high priority to advance the progress of the field[111] [Figure 3]. Though the potential of probiotic EVs in clinical applications is immense [Table 1 and Figure 1], several of these studies claimed an effect based on in vitro observations. Whether or not the findings would hold true in vivo remains unknown. For instance, while the prophylactic effects of probiotic EVs and their potential in treating and sensitizing cancer cells have been observed in vitro, these effects still need to be tested in vivo using appropriate models and controls[72-75]. This would aid in addressing several unanswered questions about probiotic EVs in addition to their therapeutic value such as their ability to selectively target tissues, thus enabling their use as novel adjuvants and drug delivery vehicles for a synergistic effect.

DECLARATIONS

Authors’ contributions

Made substantial contributions to the conception and design of the study and performed data analysis and interpretation: Sanwlani R, Bramich K, Mathivanan S

Performed data acquisition, as well as providing administrative, technical, and material support: Sanwlani R, Bramich K, Mathivanan S

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024.

Supplementary Materials

REFERENCES

1. Hill C, Guarner F, Reid G, et al. Expert consensus document. The International Scientific Association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 2014;11:506-14.

2. Clarke TC, Black LI, Stussman BJ, Barnes PM, Nahin and RL. Trends in the use of complementary health approaches among adults: United States, 2002-2012. Natl Health Stat Report 2015;79:1-16.

3. Banfi D, Moro E, Bosi A, et al. Impact of microbial metabolites on microbiota-gut-brain axis in inflammatory bowel disease. Int J Mol Sci 2021;22:1623.

4. Colomier E, Van Oudenhove L, Tack J, et al. Predictors of symptom-specific treatment response to dietary interventions in irritable bowel syndrome. Nutrients 2022;14:397.

5. Hall AB, Yassour M, Sauk J, et al. A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients. Genome Med 2017;9:103.

6. Ridaura VK, Faith JJ, Rey FE, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013;341:1241214.

7. Hod K, Dekel R, Aviv Cohen N, et al. The effect of a multispecies probiotic on microbiota composition in a clinical trial of patients with diarrhea-predominant irritable bowel syndrome. Neurogastroenterol Motil 2018;30:e13456.

8. Suwal S, Wu Q, Liu W, et al. The probiotic effectiveness in preventing experimental colitis is correlated with host gut microbiota. Front Microbiol 2018;9:2675.

9. Brun P, Scarpa M, Marchiori C, et al. Saccharomyces boulardii CNCM I-745 supplementation reduces gastrointestinal dysfunction in an animal model of IBS. PLoS One 2017;12:e0181863.

10. Li Q, Hu W, Liu WX, et al. Streptococcus thermophilus inhibits colorectal tumorigenesis through secreting β-Galactosidase. Gastroenterology 2021;160:1179-93.e14.

11. Wu Y, Jha R, Li A, et al. Probiotics (Lactobacillus plantarum HNU082) supplementation relieves ulcerative colitis by affecting intestinal barrier functions, immunity-related gene expression, gut microbiota, and metabolic pathways in mice. Microbiol Spectr 2022;10:e0165122.

12. Liang X, Dai N, Sheng K, et al. Gut bacterial extracellular vesicles: important players in regulating intestinal microenvironment. Gut Microbes 2022;14:2134689.

13. Díaz-Garrido N, Badia J, Baldomà L. Microbiota-derived extracellular vesicles in interkingdom communication in the gut. J Extracell Vesicles 2021;10:e12161.

14. Fonseka P, Marzan AL, Mathivanan S. Introduction to the community of extracellular vesicles. Subcell Biochem 2021;97:3-18.

15. Kang T, Atukorala I, Mathivanan S. Biogenesis of extracellular vesicles. Subcell Biochem 2021;97:19-43.

16. Toyofuku M, Schild S, Kaparakis-Liaskos M, Eberl L. Composition and functions of bacterial membrane vesicles. Nat Rev Microbiol 2023;21:415-30.

17. McBroom AJ, Johnson AP, Vemulapalli S, Kuehn MJ. Outer membrane vesicle production by Escherichia coli is independent of membrane instability. J Bacteriol 2006;188:5385-92.

18. Brown L, Wolf JM, Prados-Rosales R, Casadevall A. Through the wall: extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi. Nat Rev Microbiol 2015;13:620-30.

19. Gilmore WJ, Bitto NJ, Kaparakis-liaskos M. Pathogenesis mediated by bacterial membrane vesicles. In: Mathivanan S, Fonseka P, Nedeva C, Atukorala I, editors. New Frontiers: Extracellular Vesicles. Cham: Springer International Publishing; 2021. p. 101-50.

20. Chronopoulos A, Kalluri R. Emerging role of bacterial extracellular vesicles in cancer. Oncogene 2020;39:6951-60.

21. Morishita M, Kida M, Motomura T, et al. Elucidation of the tissue distribution and host immunostimulatory activity of exogenously administered probiotic-derived extracellular vesicles for immunoadjuvant. Mol Pharm 2023;20:6104-13.

22. Lee BH, Chen YZ, Shen TL, Pan TM, Hsu WH. Proteomic characterization of extracellular vesicles derived from lactic acid bacteria. Food Chem 2023;427:136685.

23. Bajic SS, Cañas MA, Tolinacki M, et al. Proteomic profile of extracellular vesicles released by Lactiplantibacillus plantarum BGAN8 and their internalization by non-polarized HT29 cell line. Sci Rep 2020;10:21829.

24. Huang J, Zhao A, He D, Wu X, Yan H, Zhu L. Isolation and proteomic analysis of extracellular vesicles from lactobacillus salivarius SNK-6. J Microbiol Biotechnol 2024;34:224-31.

25. Dean SN, Leary DH, Sullivan CJ, Oh E, Walper SA. Isolation and characterization of Lactobacillus-derived membrane vesicles. Sci Rep 2019;9:877.

26. Dean SN, Rimmer MA, Turner KB, et al. Lactobacillus acidophilus membrane vesicles as a vehicle of bacteriocin delivery. Front Microbiol 2020;11:710.

27. Rubio AP, Martínez JH, Martínez Casillas DC, Coluccio Leskow F, Piuri M, Pérez OE. Lactobacillus casei BL23 produces microvesicles carrying proteins that have been associated with its probiotic effect. Front Microbiol 2017;8:1783.

28. Anaya-Loyola MA, Enciso-Moreno JA, López-Ramos JE, et al. Bacillus coagulans GBI-30, 6068 decreases upper respiratory and gastrointestinal tract symptoms in healthy Mexican scholar-aged children by modulating immune-related proteins. Food Res Int 2019;125:108567.

29. Lakritz JR, Poutahidis T, Levkovich T, et al. Beneficial bacteria stimulate host immune cells to counteract dietary and genetic predisposition to mammary cancer in mice. Int J Cancer 2014;135:529-40.

30. O'Mahony L, McCarthy J, Kelly P, et al. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology 2005;128:541-51.

31. Mosby CA, Bhar S, Phillips MB, Edelmann MJ, Jones MK. Interaction with mammalian enteric viruses alters outer membrane vesicle production and content by commensal bacteria. J Extracell Vesicles 2022;11:e12172.

32. Naskar A, Cho H, Kim KS. A nanocomposite with extracellular vesicles from Lactobacillus paracasei as a bioinspired nanoantibiotic targeting staphylococcus aureus. Pharmaceutics 2022;14:2273.

33. Li M, Lee K, Hsu M, Nau G, Mylonakis E, Ramratnam B. Lactobacillus-derived extracellular vesicles enhance host immune responses against vancomycin-resistant enterococci. BMC Microbiol 2017;17:66.

34. Lee BH, Wu SC, Shen TL, Hsu YY, Chen CH, Hsu WH. The applications of Lactobacillus plantarum-derived extracellular vesicles as a novel natural antibacterial agent for improving quality and safety in tuna fish. Food Chem 2021;340:128104.

35. Croatti V, Parolin C, Giordani B, Foschi C, Fedi S, Vitali B. Lactobacilli extracellular vesicles: potential postbiotics to support the vaginal microbiota homeostasis. Microb Cell Fact 2022;21:237.

36. Kim MH, Choi SJ, Choi HI, et al. Lactobacillus plantarum-derived extracellular vesicles protect atopic dermatitis induced by staphylococcus aureus-derived extracellular vesicles. Allergy Asthma Immunol Res 2018;10:516-32.

37. Costantini PE, Vanpouille C, Firrincieli A, Cappelletti M, Margolis L, Ñahui Palomino RA. Extracellular vesicles generated by gram-positive bacteria protect human tissues ex vivo from HIV-1 infection. Front Cell Infect Microbiol 2021;11:822882.

38. Ñahui Palomino RA, Vanpouille C, Laghi L, et al. Extracellular vesicles from symbiotic vaginal lactobacilli inhibit HIV-1 infection of human tissues. Nat Commun 2019;10:5656.

39. Miyoshi Y, Saika A, Nagatake T, et al. Mechanisms underlying enhanced IgA production in Peyer’s patch cells by membrane vesicles derived from Lactobacillus sakei. Biosci Biotechnol Biochem 2021;85:1536-45.

40. Yamasaki-Yashiki S, Miyoshi Y, Nakayama T, Kunisawa J, Katakura Y. IgA-enhancing effects of membrane vesicles derived from Lactobacillus sakei subsp. sakei NBRC15893. Biosci Microbiota Food Health 2019;38:23-9.

41. Mata Forsberg M, Björkander S, Pang Y, et al. Extracellular membrane vesicles from Lactobacilli dampen IFN-γ responses in a monocyte-dependent manner. Sci Rep 2019;9:17109.

42. Kurata A, Kiyohara S, Imai T, et al. Characterization of extracellular vesicles from Lactiplantibacillus plantarum. Sci Rep 2022;12:13330.

43. Diaz-Garrido N, Badia J, Baldomà L. Modulation of dendritic cells by microbiota extracellular vesicles influences the cytokine profile and exosome cargo. Nutrients 2022;14:344.

44. Kim W, Lee EJ, Bae IH, et al. Lactobacillus plantarum-derived extracellular vesicles induce anti-inflammatory M2 macrophage polarization in vitro. J Extracell Vesicles 2020;9:1793514.

45. Hu R, Lin H, Wang M, et al. Lactobacillus reuteri-derived extracellular vesicles maintain intestinal immune homeostasis against lipopolysaccharide-induced inflammatory responses in broilers. J Anim Sci Biotechnol 2021;12:25.

46. Vargoorani ME, Modarressi MH, Vaziri F, Motevaseli E, Siadat SD. Stimulatory effects of Lactobacillus casei derived extracellular vesicles on toll-like receptor 9 gene expression and cytokine profile in human intestinal epithelial cells. J Diabetes Metab Disord 2020;19:223-31.

47. Morishita M, Horita M, Higuchi A, Marui M, Katsumi H, Yamamoto A. Characterizing different probiotic-derived extracellular vesicles as a novel adjuvant for immunotherapy. Mol Pharm 2021;18:1080-92.

48. Morishita M, Sagayama R, Yamawaki Y, Yamaguchi M, Katsumi H, Yamamoto A. Activation of host immune cells by probiotic-derived extracellular vesicles via TLR2-mediated signaling pathways. Biol Pharm Bull 2022;45:354-9.

49. Bergenhenegouwen J, Kraneveld AD, Rutten L, Kettelarij N, Garssen J, Vos AP. Extracellular vesicles modulate host-microbe responses by altering TLR2 activity and phagocytosis. PLoS One 2014;9:e89121.

50. Müller L, Kuhn T, Koch M, Fuhrmann G. Stimulation of probiotic bacteria induces release of membrane vesicles with augmented anti-inflammatory activity. ACS Appl Bio Mater 2021;4:3739-48.

51. Harrison NA, Gardner CL, da Silva DR, Gonzalez CF, Lorca GL. Identification of biomarkers for systemic distribution of nanovesicles from Lactobacillus johnsonii N6.2. Front Immunol 2021;12:723433.

52. Hu R, Lin H, Li J, et al. Probiotic Escherichia coli Nissle 1917-derived outer membrane vesicles enhance immunomodulation and antimicrobial activity in RAW264.7 macrophages. BMC Microbiol 2020;20:268.

53. Kim JH, Jeun EJ, Hong CP, et al. Extracellular vesicle-derived protein from Bifidobacterium longum alleviates food allergy through mast cell suppression. J Allergy Clin Immunol 2016;137:507-16.e8.

54. Rodovalho VR, da Luz BSR, Rabah H, et al. Extracellular vesicles produced by the probiotic propionibacterium freudenreichii CIRM-BIA 129 mitigate inflammation by modulating the NF-κB pathway. Front Microbiol 2020;11:1544.

55. Kim SH, Lee JH, Kim EH, Reaney MJT, Shim YY, Chung MJ. Immunomodulatory activity of extracellular vesicles of kimchi-derived lactic acid bacteria (Leuconostoc mesenteroides, Latilactobacillus curvatus, and Lactiplantibacillus plantarum). Foods 2022;11:313.

56. Cañas MA, Fábrega MJ, Giménez R, Badia J, Baldomà L. Outer membrane vesicles from probiotic and commensal escherichia coli activate NOD1-mediated immune responses in intestinal epithelial cells. Front Microbiol 2018;9:498.

57. Champagne-Jorgensen K, Mian MF, McVey Neufeld KA, Stanisz AM, Bienenstock J. Membrane vesicles of Lacticaseibacillus rhamnosus JB-1 contain immunomodulatory lipoteichoic acid and are endocytosed by intestinal epithelial cells. Sci Rep 2021;11:13756.

58. Cohen SJ, Meyerovich G, Blank S, et al. Microbiota transfer following liver surgery involves microbial extracellular vesicle migration that affects liver immunity. Hepatol Commun 2023;7:e0164.

59. Díaz-Garrido N, Bonnin S, Riera M, Gíménez R, Badia J, Baldomà L. Transcriptomic microRNA profiling of dendritic cells in response to gut microbiota-secreted vesicles. Cells 2020;9:1534.

60. Xu X, Liu R, Zhou X, et al. Characterization of exosomes derived from IPEC-J2 treated with probiotic Bacillus amyloliquefaciens SC06 and its regulation of macrophage functions. Front Immunol 2022;13:1033471.

61. Hao H, Zhang X, Tong L, et al. Effect of extracellular vesicles derived from Lactobacillus plantarum Q7 on gut microbiota and ulcerative colitis in mice. Front Immunol 2021;12:777147.

62. Tong L, Zhang X, Hao H, et al. Lactobacillus rhamnosus GG derived extracellular vesicles modulate gut microbiota and attenuate inflammatory in DSS-induced colitis mice. Nutrients 2021;13:3319.

63. Choi JH, Moon CM, Shin TS, et al. Lactobacillus paracasei-derived extracellular vesicles attenuate the intestinal inflammatory response by augmenting the endoplasmic reticulum stress pathway. Exp Mol Med 2020;52:423-37.

64. Liang L, Yang C, Liu L, et al. Commensal bacteria-derived extracellular vesicles suppress ulcerative colitis through regulating the macrophages polarization and remodeling the gut microbiota. Microb Cell Fact 2022;21:88.

65. Kang EA, Choi HI, Hong SW, et al. Extracellular vesicles derived from kefir grain Lactobacillus ameliorate intestinal inflammation via regulation of proinflammatory pathway and tight junction integrity. Biomedicines 2020;8:522.

66. Seo MK, Park EJ, Ko SY, Choi EW, Kim S. Therapeutic effects of kefir grain Lactobacillus-derived extracellular vesicles in mice with 2,4,6-trinitrobenzene sulfonic acid-induced inflammatory bowel disease. J Dairy Sci 2018;101:8662-71.

67. West CL, Stanisz AM, Mao YK, Champagne-Jorgensen K, Bienenstock J, Kunze WA. Microvesicles from Lactobacillus reuteri (DSM-17938) completely reproduce modulation of gut motility by bacteria in mice. PLoS One 2020;15:e0225481.

68. Alvarez CS, Giménez R, Cañas MA, et al. Extracellular vesicles and soluble factors secreted by Escherichia coli Nissle 1917 and ECOR63 protect against enteropathogenic E. coli-induced intestinal epithelial barrier dysfunction. BMC Microbiol 2019;19:166.

69. Ma L, Shen Q, Lyu W, et al. Clostridium butyricum and its derived extracellular vesicles modulate gut homeostasis and ameliorate acute experimental colitis. Microbiol Spectr 2022;10:e0136822.

70. Ma L, Lyu W, Song Y, et al. Anti-inflammatory effect of clostridium butyricum-derived extracellular vesicles in ulcerative colitis: impact on host microRNAs expressions and gut microbiome profiles. Mol Nutr Food Res 2023;67:e2200884.

71. Liang D, Liu C, Li Y, et al. Engineering fucoxanthin-loaded probiotics’ membrane vesicles for the dietary intervention of colitis. Biomaterials 2023;297:122107.

72. An J, Ha EM. Extracellular vesicles derived from Lactobacillus plantarum restore chemosensitivity through the PDK2-mediated glucose metabolic pathway in 5-FU-resistant colorectal cancer cells. J Microbiol 2022;60:735-45.

73. Keyhani G, Mahmoodzadeh Hosseini H, Salimi A. Effect of extracellular vesicles of Lactobacillus rhamnosus GG on the expression of CEA gene and protein released by colorectal cancer cells. Iran J Microbiol 2022;14:90-6.

74. Behzadi E, Mahmoodzadeh Hosseini H, Imani Fooladi AA. The inhibitory impacts of Lactobacillus rhamnosus GG-derived extracellular vesicles on the growth of hepatic cancer cells. Microb Pathog 2017;110:1-6.

75. Shi Y, Meng L, Zhang C, Zhang F, Fang Y. Extracellular vesicles of Lacticaseibacillus paracasei PC-H1 induce colorectal cancer cells apoptosis via PDK1/AKT/Bcl-2 signaling pathway. Microbiol Res 2021;255:126921.

76. Chelakkot C, Choi Y, Kim DK, et al. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp Mol Med 2018;50:e450.

77. Ashrafian F, Keshavarz Azizi Raftar S, Lari A, et al. Extracellular vesicles and pasteurized cells derived from Akkermansia muciniphila protect against high-fat induced obesity in mice. Microb Cell Fact 2021;20:219.

78. Ashrafian F, Shahriary A, Behrouzi A, et al. Akkermansia muciniphila-derived extracellular vesicles as a mucosal delivery vector for amelioration of obesity in mice. Front Microbiol 2019;10:2155.

79. Yang Z, Gao Z, Yang Z, et al. Lactobacillus plantarum-derived extracellular vesicles protect against ischemic brain injury via the microRNA-101a-3p/c-Fos/TGF-β axis. Pharmacol Res 2022;182:106332.

80. Choi J, Kwon H, Kim YK, Han PL. Extracellular vesicles from gram-positive and gram-negative probiotics remediate stress-induced depressive behavior in mice. Mol Neurobiol 2022;59:2715-28.

81. Choi J, Kim YK, Han PL. Extracellular vesicles derived from Lactobacillus plantarum increase BDNF expression in cultured hippocampal neurons and produce antidepressant-like effects in mice. Exp Neurobiol 2019;28:158-71.

82. Chen CY, Rao SS, Yue T, et al. Glucocorticoid-induced loss of beneficial gut bacterial extracellular vesicles is associated with the pathogenesis of osteonecrosis. Sci Adv 2022;8:eabg8335.

83. Kim HY, Song MK, Lim Y, et al. Effects of extracellular vesicles derived from oral bacteria on osteoclast differentiation and activation. Sci Rep 2022;12:14239.

84. Han F, Wang K, Shen K, et al. Extracellular vesicles from Lactobacillus druckerii inhibit hypertrophic scar fibrosis. J Nanobiotechnology 2023;21:113.

85. Kuhn T, Aljohmani A, Frank N, et al. A cell-free, biomimetic hydrogel based on probiotic membrane vesicles ameliorates wound healing. J Control Release 2024;365:969-80.

86. Jo CS, Myung CH, Yoon YC, et al. The effect of Lactobacillus plantarum extracellular vesicles from Korean women in their 20s on skin aging. Curr Issues Mol Biol 2022;44:526-40.

87. Bäuerl C, Coll-Marqués JM, Tarazona-González C, Pérez-Martínez G. Lactobacillus casei extracellular vesicles stimulate EGFR pathway likely due to the presence of proteins P40 and P75 bound to their surface. Sci Rep 2020;10:19237.

88. Keshavarz Azizi Raftar S, Ashrafian F, Yadegar A, et al. The protective effects of live and pasteurized Akkermansia muciniphila and its extracellular vesicles against HFD/CCl4-induced liver injury. Microbiol Spectr 2021;9:e0048421.

89. Raftar SKA, Ashrafian F, Abdollahiyan S, et al. The anti-inflammatory effects of Akkermansia muciniphila and its derivates in HFD/CCL4-induced murine model of liver injury. Sci Rep 2022;12:2453.

90. Lee DH, Park HK, Lee HR, et al. Immunoregulatory effects of Lactococcus lactis-derived extracellular vesicles in allergic asthma. Clin Transl Allergy 2022;12:e12138.

91. Yoon YC, Ahn BH, Min JW, Lee KR, Park SH, Kang HC. Stimulatory effects of extracellular vesicles derived from leuconostoc holzapfelii that exists in human scalp on hair growth in human follicle dermal papilla cells. Curr Issues Mol Biol 2022;44:845-66.

92. Yaghoubfar R, Behrouzi A, Zare Banadkoki E, et al. Effect of Akkermansia muciniphila, Faecalibacterium prausnitzii, and their extracellular vesicles on the serotonin system in intestinal epithelial cells. Probiotics Antimicrob Proteins 2021;13:1546-56.

93. Takiishi T, Fenero CIM, Câmara NOS. Intestinal barrier and gut microbiota: shaping our immune responses throughout life. Tissue Barriers 2017;5:e1373208.

94. Chen Q, Fang Z, Yang Z, et al. Lactobacillus plantarum-derived extracellular vesicles modulate macrophage polarization and gut homeostasis for alleviating ulcerative colitis. J Agric Food Chem 2024;72:14713-26.

95. Takahashi N, Kitazawa H, Iwabuchi N, et al. Oral administration of an immunostimulatory DNA sequence from Bifidobacterium longum improves Th1/Th2 balance in a murine model. Biosci Biotechnol Biochem 2006;70:2013-7.

96. Liu C, Yazdani N, Moran CS, et al. Unveiling clinical applications of bacterial extracellular vesicles as natural nanomaterials in disease diagnosis and therapeutics. Acta Biomater 2024;180:18-45.

97. Dudek-Wicher R, Junka A, Paleczny J, Bartoszewicz M. Clinical trials of probiotic strains in selected disease entities. Int J Microbiol 2020;2020:8854119.

98. Dronkers TMG, Ouwehand AC, Rijkers GT. Global analysis of clinical trials with probiotics. Heliyon 2020;6:e04467.

99. Peng Y, Ma Y, Luo Z, Jiang Y, Xu Z, Yu R. Lactobacillus reuteri in digestive system diseases: focus on clinical trials and mechanisms. Front Cell Infect Microbiol 2023;13:1254198.

100. Jian H, Liu Y, Wang X, Dong X, Zou X. Akkermansia muciniphila as a next-generation probiotic in modulating human metabolic homeostasis and disease progression: a role mediated by gut-liver-brain axes? Int J Mol Sci 2023;24:3900.

101. Díez-Sainz E, Milagro FI, Riezu-Boj JI, Lorente-Cebrián S. Effects of gut microbiota-derived extracellular vesicles on obesity and diabetes and their potential modulation through diet. J Physiol Biochem 2022;78:485-99.

102. Champagne-Jorgensen K, Jose TA, Stanisz AM, Mian MF, Hynes AP, Bienenstock J. Bacterial membrane vesicles and phages in blood after consumption of lacticaseibacillus rhamnosus JB-1. Gut Microbes 2021;13:1993583.

103. Munagala R, Aqil F, Jeyabalan J, Gupta RC. Bovine milk-derived exosomes for drug delivery. Cancer Lett 2016;371:48-61.

104. Samuel M, Fonseka P, Sanwlani R, et al. Oral administration of bovine milk-derived extracellular vesicles induces senescence in the primary tumor but accelerates cancer metastasis. Nat Commun 2021;12:3950.

105. Hansen MS, Gregersen SB, Rasmussen JT. Bovine milk processing impacts characteristics of extracellular vesicle isolates obtained by size-exclusion chromatography. International Dairy Journal 2022;127:105212.

106. Colella AP, Prakash A, Miklavcic JJ. Homogenization and thermal processing reduce the concentration of extracellular vesicles in bovine milk. Food Sci Nutr 2024;12:131-40.

107. Kleinjan M, van Herwijnen MJ, Libregts SF, van Neerven RJ, Feitsma AL, Wauben MH. Regular industrial processing of bovine milk impacts the integrity and molecular composition of extracellular vesicles. J Nutr 2021;151:1416-25.

108. Park JY, Choi J, Lee Y, et al. Metagenome analysis of bodily microbiota in a mouse model of alzheimer disease using bacteria-derived membrane vesicles in blood. Exp Neurobiol 2017;26:369-79.

109. Park J, Lee JJ, Hong Y, Seo H, Shin TS, Hong JY. Metagenomic analysis of plasma microbial extracellular vesicles in patients receiving mechanical ventilation: a pilot study. J Pers Med 2022;12:564.

110. Rubio APD, Martínez J, Palavecino M, et al. Transcytosis of Bacillus subtilis extracellular vesicles through an in vitro intestinal epithelial cell model. Sci Rep 2020;10:3120.

111. Sanwlani R, Fonseka P, Mathivanan S. Are dietary extracellular vesicles bioavailable and functional in consuming organisms? In: Mathivanan S, Fonseka P, Nedeva C, Atukorala I, editors. New Frontiers: Extracellular Vesicles. Cham: Springer International Publishing; 2021. p. 509-21.

112. Song M, Cui M, Fang Z, Liu K. Advanced research on extracellular vesicles based oral drug delivery systems. J Control Release 2022;351:560-72.

113. Sanwlani R, Fonseka P, Chitti SV, Mathivanan S. Milk-derived extracellular vesicles in inter-organism, cross-species communication and drug delivery. Proteomes 2020;8:11.

114. Schwechheimer C, Kuehn MJ. Outer-membrane vesicles from Gram-negative bacteria: biogenesis and functions. Nat Rev Microbiol 2015;13:605-19.

115. Turnbull L, Toyofuku M, Hynen AL, et al. Explosive cell lysis as a mechanism for the biogenesis of bacterial membrane vesicles and biofilms. Nat Commun 2016;7:11220.

116. Iwabuchi Y, Nakamura T, Kusumoto Y, et al. Effects of pH on the properties of membrane vesicles including glucosyltransferase in streptococcus mutans. Microorganisms 2021;9:2308.

117. Mozaheb N, Mingeot-Leclercq MP. Membrane vesicle production as a bacterial defense against stress. Front Microbiol 2020;11:600221.

118. Godlewska R, Klim J, Dębski J, Wyszyńska A, Łasica A. Influence of environmental and genetic factors on proteomic profiling of outer membrane vesicles from campylobacter jejuni. Pol J Microbiol 2019;68:255-61.

119. Ermann Lundberg L, Pallabi Mishra P, Liu P, et al. Bifidobacterium longum subsp. longum BG-L47 boosts growth and activity of Limosilactobacillus reuteri DSM 17938 and its extracellular membrane vesicles. Appl Environ Microbiol 2024;90:e0024724.

120. Takov K, Yellon DM, Davidson SM. Comparison of small extracellular vesicles isolated from plasma by ultracentrifugation or size-exclusion chromatography: yield, purity and functional potential. J Extracell Vesicles 2019;8:1560809.

121. 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.

122. Théry C, Witwer KW, Aikawa E, 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.

123. Hong J, Dauros-Singorenko P, Whitcombe A, et al. Analysis of the Escherichia coli extracellular vesicle proteome identifies markers of purity and culture conditions. J Extracell Vesicles 2019;8:1632099.

124. Kim SW, Lee JS, Park SB, et al. The importance of porins and β-Lactamase in outer membrane vesicles on the hydrolysis of β-lactam antibiotics. Int J Mol Sci 2020;21:2822.

125. Bielaszewska M, Daniel O, Nyč O, Mellmann A. In Vivo secretion of β-Lactamase-carrying outer membrane vesicles as a mechanism of β-Lactam therapy failure. Membranes (Basel) 2021;11:806.

126. Salemi R, Vivarelli S, Ricci D, et al. Lactobacillus rhamnosus GG cell-free supernatant as a novel anti-cancer adjuvant. J Transl Med 2023;21:195.

127. Escamilla J, Lane MA, Maitin V. Cell-free supernatants from probiotic Lactobacillus casei and Lactobacillus rhamnosus GG decrease colon cancer cell invasion in vitro. Nutr Cancer 2012;64:871-8.

128. Elhenawy W, Debelyy MO, Feldman MF. Preferential packing of acidic glycosidases and proteases into Bacteroides outer membrane vesicles. mBio 2014;5:e00909-14.

129. Fonseka P, Pathan M, Chitti SV, Kang T, Mathivanan S. FunRich enables enrichment analysis of OMICs datasets. J Mol Biol 2021;433:166747.

130. 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.

131. Steć A, Jońca J, Waleron K, et al. Quality control of bacterial extracellular vesicles with total protein content assay, nanoparticles tracking analysis, and capillary electrophoresis. Int J Mol Sci 2022;23:4347.

132. Ghareeb K, Awad W, Böhm J, Zebeli Q. Impact of luminal and systemic endotoxin exposure on gut function, immune response and performance of chickens. World's Poultry Science Journal 2016;72:367-80.

133. Tolomeo AM, Zuccolotto G, Malvicini R, et al. Biodistribution of intratracheal, intranasal, and intravenous injections of human mesenchymal stromal cell-derived extracellular vesicles in a mouse model for drug delivery studies. Pharmaceutics 2023;15:548.

134. Kang M, Jordan V, Blenkiron C, Chamley LW. Biodistribution of extracellular vesicles following administration into animals: a systematic review. J Extracell Vesicles 2021;10:e12085.

135. Svennerholm K, Park KS, Wikström J, et al. Escherichia coli outer membrane vesicles can contribute to sepsis induced cardiac dysfunction. Sci Rep 2017;7:17434.

136. Finethy R, Luoma S, Orench-Rivera N, et al. Inflammasome activation by bacterial outer membrane vesicles requires guanylate binding proteins. mBio 2017;8:e01188-17.

137. Klimentová J, Stulík J. Methods of isolation and purification of outer membrane vesicles from gram-negative bacteria. Microbiol Res 2015;170:1-9.

138. Kulp A, Kuehn MJ. Biological functions and biogenesis of secreted bacterial outer membrane vesicles. Annu Rev Microbiol 2010;64:163-84.

139. Kim OY, Park HT, Dinh NTH, et al. Bacterial outer membrane vesicles suppress tumor by interferon-γ-mediated antitumor response. Nat Commun 2017;8:626.

140. Yue Y, Wang C, Benedict C, et al. Interleukin-10 deficiency alters endothelial progenitor cell-derived exosome reparative effect on myocardial repair via integrin-linked kinase enrichment. Circ Res 2020;126:315-29.

141. Pathan M, Fonseka P, Chitti SV, et al. Vesiclepedia 2019: a compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res 2019;47:D516-9.

Cite This Article

Review
Open Access
Role of probiotic extracellular vesicles in inter-kingdom communication and current technical limitations in advancing their therapeutic utility
Rahul SanwlaniRahul Sanwlani, ... Suresh MathivananSuresh Mathivanan

How to Cite

Sanwlani, R.; Bramich K.; Mathivanan S. Role of probiotic extracellular vesicles in inter-kingdom communication and current technical limitations in advancing their therapeutic utility. Extracell. Vesicles. Circ. Nucleic. Acids. 2024, 5, 609-26. http://dx.doi.org/10.20517/evcna.2024.39

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click on download.

Export Citation File:

Type of Import

Tips on Downloading Citation

This feature enables you to download the bibliographic information (also called citation data, header data, or metadata) for the articles on our site.

Citation Manager File Format

Use the radio buttons to choose how to format the bibliographic data you're harvesting. Several citation manager formats are available, including EndNote and BibTex.

Type of Import

If you have citation management software installed on your computer your Web browser should be able to import metadata directly into your reference database.

Direct Import: When the Direct Import option is selected (the default state), a dialogue box will give you the option to Save or Open the downloaded citation data. Choosing Open will either launch your citation manager or give you a choice of applications with which to use the metadata. The Save option saves the file locally for later use.

Indirect Import: When the Indirect Import option is selected, the metadata is displayed and may be copied and pasted as needed.

About This Article

© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
331
Downloads
63
Citations
0
Comments
0
4

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Extracellular Vesicles and Circulating Nucleic Acids
ISSN 2767-6641 (Online)
Follow Us

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/