Next Article in Journal
Insights into Bacterial Extracellular Vesicle Biogenesis, Functions, and Implications in Plant–Microbe Interactions
Previous Article in Journal
Characterization of Antibiotic-Resistance Antarctic Pseudomonas That Produce Bacteriocin-like Compounds
Previous Article in Special Issue
Extracellular Vesicles: A Novel Mode of Viral Propagation Exploited by Enveloped and Non-Enveloped Viruses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

State of the Art on the Role of Staphylococcus aureus Extracellular Vesicles in the Pathogenesis of Atopic Dermatitis

by
Marina Passos Torrealba
1,
Fabio Seiti Yamada Yoshikawa
2,
Valeria Aoki
1,
Maria Notomi Sato
1 and
Raquel Leão Orfali
1,*
1
Laboratory of Medical Investigation in Dermatology and Immunodeficiencies (LIM-56), Department of Dermatology, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, SP, Brazil
2
Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba 260-8670, Japan
*
Author to whom correspondence should be addressed.
Microorganisms 2024, 12(3), 531; https://doi.org/10.3390/microorganisms12030531
Submission received: 15 February 2024 / Revised: 1 March 2024 / Accepted: 5 March 2024 / Published: 6 March 2024
(This article belongs to the Special Issue Extracellular Vesicles in Pathogens)

Abstract

:
Atopic dermatitis (AD) is a chronic and relapsing inflammatory cutaneous disease. The role of host defense and microbial virulence factors in Staphylococcus aureus skin colonization, infection, and inflammation perpetuation in AD remains an area of current research focus. Extracellular vesicles (EV) mediate cell-to-cell communication by transporting and delivering bioactive molecules, such as nucleic acids, proteins, and enzymes, to recipient cells. Staphylococcus aureus spontaneously secretes extracellular vesicles (SA-derived EVs), which spread throughout the skin layers. Previous research has shown that SA-derived EVs from AD patients can trigger cytokine secretion in keratinocytes, shape the recruitment of neutrophils and monocytes, and induce inflammatory AD-type lesions in mouse models, in addition to their role as exogenous worsening factors for the disease. In this review article, we aim to examine the role of SA-derived EVs in AD physiopathology and its progression, highlighting the recent research in the field and exploring the potential crosstalk between the host and the microbiota.

Graphical Abstract

1. Introduction

Atopic dermatitis (AD) is a chronic and relapsing inflammatory skin disease. It shows worldwide occurrence, affecting both children and adults, whose prevalence is approximately 15–20% and 1–5%, respectively [1,2,3]. Although a common disease, the pathogenesis of AD is a complex network of skin barrier defects, imbalance in adaptive and innate immunity, and chronic skin colonization by Staphylococcus aureus (S. aureus) [4,5,6], all of which drive an inflammatory vicious cycle culminating in the main symptom: pruritus [7,8,9,10]. But among the variables associated with AD development, the interaction between the host defense system and the microbial virulence factors involved in S. aureus skin colonization, infection and the perpetuation of inflammation are gathering increased attention [11], mostly due to clinical evidence that, in AD flares, decreased bacterial diversity associated with increased S. aureus abundance has been described and related to disease severity [11,12,13,14].
The virulence factors of S. aureus include the secretion of numerous exotoxins, comprising an assembly of polypeptides capable of injuring the host cell plasma membrane, the called pore-forming toxins: α-hemolysin and the bi-component leukocidins γ-hemolysin, the Panton Valentine leukocidin, LukED, and LukGH/AB, β-hemolysin (neutral sphingomyelinase), and the phenol soluble modulins (small amphipathic peptides), as well as staphylococcal enterotoxins (SEA to SEE, SEG to SEJ, SEL to SEQ and SER to SET), and 11 staphylococcal superantigen-like (SSL) toxins (SEIK to SEIQ, SEIU to SEIX) [15,16].
Almost all cell types release double-lipid membrane structures termed “extracellular vesicles (EVs)” into the extracellular lumen. EV production is a conserved biological phenomenon, considered a fundamental feature in all three domains of life: eukaryotic, prokaryotic and archaea cells [17,18,19]. In Eukaryotes, EVs are named based on their size, function, and biogenesis. Apoptotic bodies are very large vesicles with an average diameter of 1000–5000 nm, formed through the disassembly of the cell membrane. Ectosomes, also known as microvesicles, exhibit 100 to 1000 nm of diameter and are formed by the shedding of the plasma membrane. Exosomes are smaller vesicles ranging from 40 to 150 nm and originating from endosomal generation. Despite this categorization, the International Society for Extracellular Vesicles (ISEV) recommends the use of “extracellular vesicles” as a communal term for “particles naturally released from the cell that are delimited by a lipid bilayer and cannot replicate” [20,21,22]. These spherical structures play an essential role in cell communication by acting as biological carriers between different cells and tissues. Moreover, surface molecules on EVs can target specific cells and tissues, providing specificity to the recipient cell. The cargo includes acid nucleic molecules, such as DNA and RNA, proteins, lipids, enzymes, and any other molecules found in the donor cell.
EVs exert a physiological role in cellular communication but have also been found to be important drivers in the pathophysiology of many diseases, including AD in recent years. Thus, the mechanism of action by which microbe-derived EVs may contribute to immune modulation, the delivery of virulence factors, the enhancement of antibiotic resistance, as well as the facilitation of biofilm production, thereby impacting the physiopathology of inflammatory diseases [20], are still being uncovered. The aim of this review is to examine the role of S. aureus-derived EVs (SA-derived EVs) in the pathophysiology and progression of AD.

2. Review Strategy

The authors performed a review in the PubMed, Embase, Google Scholar, and Web of Science databases. The search terms were: (‘staphylococcus aureus extracellular vesicles’ OR ((‘staphylococcus’/exp OR staphylococcus) AND aureus AND extracellular AND (‘vesicles’/exp OR vesicles)) OR ‘staphylococcus aureus exosomes’ OR ((‘staphylococcus’/exp OR staphylococcus) AND aureus AND (‘exosomes’/exp OR exosomes))) AND (‘atopic dermatitis’/exp OR ‘atopic dermatitis’ OR (atopic AND (‘dermatitis’/exp OR dermatitis))). The basis for assessment was the title and abstract for relevance to S. aureus EVs and AD. We included English-language articles published between 2009 and 2023. Figure 1 illustrates the accessed and selected papers.
The search terms resulted in 11 articles in PubMed, 23 in Embase, 48 in Web of Science, and 572 in Google Scholar, but many with topics that did not match the purpose of this article. We excluded all articles that did not fit with the aim of the current review and accessed the relevant articles related to the topic.
In Table 1, we present a timeline summarizing the main findings in the field uncovered during the last 15 years.

3. A Brief Background on SA-Derived EVs

Any cell has the potential to produce EVs. EVs released from Gram-negative bacteria range between 10–500 nm and were first described in 1966, named as outer membrane vesicles (OMVs) due to their origin from the bacterial outer membrane layer [37,38]. Curiously, the production of EVs by Gram-positive bacteria was only confirmed in 2009 [23], mostly due to a misconception; once they do not possess an outer membrane, they would be unable to liberate such structures [39]. Nonetheless, it was uncovered that Gram-positive bacteria-derived EVs are released from the single cytoplasmic cell membrane surrounded by a peptidoglycan-rich cell wall, and exhibit a diameter of 20–400 nm, and are therefore named membrane vesicles (MVs) [40]. In this review, however, we will keep the term “extracellular vesicles/EVs” regardless of the source organism.
S. aureus is also capable of producing and releasing EVs (here termed SA-derived EVs) that contain membrane proteins, as well as superantigens and enzymes, but which do not seem to display significant cytotoxicity after being internalized by the host cell [24,32]. In 2009, Lee et al. identified 90 proteins in S. aureus EVs through proteomic approaches, marking also the first observation of S. aureus spontaneously producing these structures [23]. More recently, another research group evaluated five different S. aureus strains obtained from various hosts, including humans, bovine, and ovine animals, and demonstrated the presence of a highly conserved EV core proteome for SA-derived EV [41]. This core comprises 119 proteins, including toxins such as phenol-soluble modulins, haemolysin, and leukocidin, invasion factors like elastin-binding protein, autolysin, and extracellular adherence protein, enzymes such as nuclease and proteases, surface antigens, and immunoglobulin-binding proteins [41]. Since then, SA-derived EVs have been implicated as an exogenous worsening factor of AD and other allergic diseases by mechanisms as increasing the expression of pro-inflammatory cytokines [27,35]. Certain components from SA-derived EVs have been directly associated with extracellular matrix degradation, bacterial antibiotic resistance, and bacterial invasion [42,43,44], providing strong evidence for their role in virulence.
Among the many factors that stimulate the release of SA-derived EVs, one can cite the exposure to DNA-damaging agents or UV radiation as a relevant trigger [45]. Antibiotics such as the β-lactam flucloxacillin and ceftaroline are capable of stimulating their secretion through different pathways, such as DNA-damaging agents and antibiotics that induce signaling responses, initiating vesicle formation in lysogenic strains of S. aureus, or in a prophage-independent manner by affecting the peptidoglycan layer [45].

4. SA-Derived EVs and AD Pathophysiology

In 2011, Hong et al. [24] conducted the first study to propose the role of S. aureus-derived EV in AD pathophysiology. The authors reported that applying SA-derived EVs in tape-stripping mouse skin resulted in skin inflammation similar to that observed in AD patients, displaying a profile encompassed by (i) thickened epidermis, (ii) dermal infiltration of polymorphonuclear cells, (iii) an increase in Th1/Th17 inflammatory cytokines, and (iv) elevated serum IgE [24]. Importantly, these modifications were observed without the presence of live bacterial cells [24].
S. aureus colonization and infection evoke an immune response from the host by secreting pathogenic molecules and toxins (reviewed in Seiti Yamada Yoshikawa et al., 2019) [46]. Among these, we emphasize α-hemolysin, a cytotoxic protein secreted by S. aureus which induces cell death [47] on different cell types [48,49], including keratinocytes [23]. S. aureus from colonized AD skin releases greater amounts of α-hemolysin compared to S. aureus from the skin of healthy individuals. Furthermore, there are reports suggesting S. aureus α-hemolysin production is linked to AD severity [25].
An in vitro analysis revealed that EVs-associated α-hemolysin was more cytotoxic, and more effective to induce HaCaT keratinocytes death than soluble α-hemolysin, producing necrosis and inducing epidermal thickening and eosinophilic inflammation in the dermis [25,34], which could be explained by their better delivery to the host cell when encapsulated inside EVs. In addition, despite in vivo studies that indicate that soluble and EV-associated α-hemolysin induced skin barrier disruption and epidermal hyperplasia, an EV-associated toxin was the responsible for the induction of dermal infiltration [25].
Some S. aureus strains, such as ATCC14458 [24], a related cytotoxin producer, can release cytotoxins into the lumen of the EVs and induce their secretion to the extracellular space. Besides the assistance in the killing of the host cells by transferring these cytotoxic factors inside EVs, which favors their integration into the target cell cytoplasm [25], their association also shields the toxins from neutralization by the host immune system [24,25,50].
Mutually, in vitro and in vivo studies indicate that SA-derived EVs can upregulate pro-inflammatory mediators that elicit the Th17 response with augmented production of IgE, triggering AD-like inflammation [21,24]. Although AD is a disease ruled by type 2 immune responses (Th2, IL-4 and IL-13), IL-17 contributes to the worsening of the symptoms by enhancing IL-4 production from Th2 cells [51]. Moreover, SA-derived EVs induce the secretion of CXCL8 and TNF-α by primary human keratinocytes, the recruitment of neutrophils, and the formation of neutrophil extracellular traps, leading to better S. aureus skin colonization. The stimulation of CXCL8 is TLR2- and NFκB-dependent, and the induction level positively correlates with the membrane lipid and protein A in a similar quantity as those from pathogenic S. aureus [33].
SA-derived EVs can mediate inflammatory responses in AD pathogenesis; Kim et al. [30] examined the effect of these EVs on human dermal microvascular endothelial cells (HDMECs). HDMECs treated with SA-derived EVs increased the expression of cell adhesion molecules such as E-selectin, VCAM1, and ICAM1, and IL-6 with improved recruitment of monocytes in a TLR-4/NFκB-dependent signaling pathway [30]. All these findings together suggest that SA-derived EVs are key molecules in AD pathogenesis and are potential therapeutic targets. Figure 2 illustrates the role of SA-derived EVs in AD and some key biological effects are summarized in Table 2.

5. SA-Derived EVs and Microbiota in the Context of AD

Several studies have described alterations in the microbiome composition and Shannon’s microbial diversity index in AD skin, demonstrating a prevalence of S. aureus strains in flares [52,53,54]. Kim et al. [26] demonstrated the recovery of the reduced microbial diversity AD skin after treatment and detected SA-derived EVs in serum samples of AD patients, suggesting a novel host-microbiota crosstalk via microbe-derived EVs [20]. Likewise, a pilot study targeting the EV microbiome in the serum of AD patients identified biomarkers based on metagenomic analysis of serum-derived microbial EVs reinforcing that they may be a helpful tool for diagnosis, prognosis, and treatment prediction [31].
Besides S. aureus, commensal microorganisms on the skin also release EVs. For example, the EVs from the commensal yeast Malassezia sympodialis, associated with episodes of atopic dermatitis, can work as vessels for delivering small RNAs [55]. On the other hand, EV derived from skin commensals can protect against S. aureus colonization by conditioning the human skin to enhance innate defenses and by interfering with S. aureus keratinocyte attachment [33].
Recent research has highlighted how microbe-derived EVs can help to regulate immune responses in allergic diseases [20]. EVs derived from the probiotic Lactobacillus plantarum have demonstrated the ability to restore cell viability in keratinocytes and reduce levels of secreted IL-6 from both keratinocytes and macrophages in response to SA-derived EVs in vitro [29]. Moreover, Lactobacillus plantarum-derived EVs administered orally in a mouse model of AD induced with SA-derived EVs inhibited IL-4 production and its consequent skin inflammation in vivo, resulting in decreased epidermal thickness, albeit no changes in the eosinophilic infiltration [29]. These findings point out L. plantarum-derived EVs as a possible candidate for preventing skin inflammation [29,35].
Reports show the successful treatment of AD-like skin lesions induced by SA-derived EV inoculation in mice ears with monoterpenoid thymol. Both the topical application of thymol-treated SA-derived EVs or treatment with thymol after intact EV administration reduced AD inflammation, decreased epidermal/dermal thickness, attenuated the infiltration of eosinophils/mast cells, and reduced the expression of proinflammatory cytokine/chemokine genes [28]. Furthermore, thymol inhibited Th1, Th2, and Th17-mediated inflammatory responses as well as reduced IgG2a, mite-specific IgE, and total IgE serum levels [28].
Staphylococcus epidermidis dysbiosis is associated with AD disease, but in the opposite way to S. aureus. Byrd et al. [56] demonstrated in 2017 that S. epidermidis predominance is associated with less severe AD disease in some patients, while its lower abundance is associated with a more severe clinical presentation. In fact, EVs from skin commensal microbes, including S. epidermidis, S. lugdunensis, and S. hominis play a potentially protective role against S. aureus skin colonization by decreasing the adherence of S. aureus to keratinocytes [31]. Interestingly, a recent study has reported that EVs derived from S. epidermidis can reduce the expression of proinflammatory genes (TNF-α, IL-1β, IL-6, IL-8, and iNOS) in MC903-treated HaCaT cells [36]. Moreover, S. epidermidis EVs induced the expression of human β-defensins and promoted epidermal cell renewal in HaCaT cells, in addition to suppressing proinflammatory genes [36]. Finally, in vivo studies showed that the topical administration of S. epidermidis EVs in an AD-like dermatitis model attenuated lesion formation and reduced AD-like inflammatory features, such as epidermal thickness, dryness, transepidermal water loss, pruritus, epidermal cell infiltration (CD4+ T cells and Gr1+ cells), Th2 cytokine gene expression (IL-4, IL-13 and TLSP), and IgE levels [36].

6. Future Research Questions

In this review, we describe an updated panorama of the research regarding SA-derived EVs and AD. Even though the subject is still in its initial approach, we believe that SA-derived EVs may have a role as novel players in AD pathogenesis. Overall, EVs are essential virulence factors for S. aureus, highlighted by the higher toxicity of the pathogen toxins observed when associated with/carried within EVs and their ability to disrupt the host immune system in favor of skin colonization.
Kobiela et al. [57] demonstrated that supernatants from S. aureus cultures hijack the EV secretion machinery of keratinocytes, increasing the content of filaggrin eliminated inside these vesicles. This would deplete the keratinocyte reservoir of this protein, which could possibly impair the antimicrobial functions of the epidermal layer, favoring bacteria colonization. Since the authors did not identify the component(s) responsible for this activity, we may hypothesize that SA-derived EVs in those supernatants could be involved in delivering the bacterial factors to the keratinocytes. Indeed, the proportion of factors/toxins that S. aureus might release in the free-form and in an EV-encapsulated manner remains to be explored. It would be even more interesting to assess whether AD-associated S. aureus strains are more prone to EV production than the ones colonizing healthy individuals, which could help to explain the dysbiosis observed in AD patients.
Although not the focus of this review, it is relevant to point out that the host has its own source of EVs, which exert a role in the shaping of the host immune profile, therefore counterbalancing the AD polarization driven by SA-derived EVs [58]. Cho et al. [59] showed that EVs from adipose tissue-derived mesenchymal stem cells ameliorate AD symptoms in an experimental model through undetermined effector mechanism(s). Furthermore, IFN-γ-primed mesenchymal stem cells secrete EVs that dampen AD symptoms by repressing Th2 responses [60]. Therefore, the phenotype that prevails would be determined by the relative contributions of SA-derived versus host-derived EVs. We still have a poor understanding of if and how the EV profile changes between atopic and non-atopic individuals; one can speculate that AD patients could even display different EV kinetics between resting and flare states. Thus, whether an “atopy-prone EV profile” can be delineated is an open question for future investigations.
The search for EV markers of AD would be very beneficial for early diagnosis, disease monitoring, or even therapeutic targets. One important advantage is the easy accessibility of EV samples from the blood. Indeed, some authors propose the use of blood fluids as a source of EVs with the aim of improving and facilitating AD diagnosis [26]. Although plasma and serum are the main sources for EV studies due to their abundance in liquid biopsies representing a low-invasive harvesting procedure [61], they include a diverse mixture of EVs from different cell types and tissues, but also from different species [62,63,64]. This might not be a concern in a laboratory routine, but some limitations are present when considering research purposes. Fortunately, the discrimination of the source of EVs seems to be technically possible, as exemplified by Yang et al. (2020) [31], who assessed the diversity of microbial EVs in the serum of AD patients by a metagenomic approach to establish a diagnostic model. Great advances in the use of mass spectrometry and computational biology should harbor the answer for faster and more detailed EV profiling in the context of patient screening [65].
Finally, it is important to keep in mind the bidirectional nature of this subject. Despite the great focus on the ways that SA-derived EVs can affect the host to benefit pathogen establishment, one might wonder whether host EVs could not affect S. aureus behavior. Recently, Wang et al. [66] demonstrated that the model plant Arabidopis thaliana uses its EVs to deliver mRNAs to the fungal pathogen Botrytis cinerea, attenuating its virulence. Although this idea remains largely unappreciated in the AD field, it opens new venues of investigation for exploring host—S. aureus interaction.
Among the many possibilities in the field of SA-derived EVs in atopic dermatitis, we proposed some drivers for a better understanding of AD, including its pathogenesis, identifying new biomarkers and targets for clinical intervention.
Is it possible to establish an AD-linked, SA-derived EV profile?
-
Are AD-associated S. aureus strains related to different EV features?
-
Is the EV production machinery a viable target for targeted pharmacological intervention?

7. Conclusions

Microbe-derived EVs have various functions linked to cell-to-cell communication and are currently potential tools for diagnostic and therapeutic purposes. Microbe-derived EV studies show novel approaches for diseases such as atopic dermatitis. Future studies will elucidate gaps in the understanding of host and microbiota crosstalk, further facilitating novel diagnostic tools and therapeutic targets.

Author Contributions

Writing—original draft preparation—R.L.O., F.S.Y.Y. and M.P.T.; web search—R.L.O. and M.P.T.; conceptualization, writing—review and editing—R.L.O., M.P.T., M.N.S. and V.A. All authors have read and agreed to the published version of the manuscript.

Funding

Fundação de Amparo à Pesquisa do Estado de São Paulo—FAPESP (Grant # 2016/24161-1).

Data Availability Statement

Data sharing not applicable.

Acknowledgments

We acknowledge the Laboratory of Dermatology and Immunodeficiencies (LIM-56), Department of Dermatology, University of Sao Paulo Medical School.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sacotte, R.; Silverberg, J.I. Epidemiology of adult atopic dermatitis. Clin. Dermatol. 2018, 36, 595–605. [Google Scholar] [CrossRef]
  2. Asher, M.I.; Montefort, S.; Björkstén, B.; Lai, C.K.; Strachan, D.P.; Weiland, S.K.; Williams, H. Worldwide time trends in the prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and eczema in childhood: ISAAC Phases One and Three repeat multicountry cross-sectional surveys. Lancet 2006, 368, 733–743. [Google Scholar] [CrossRef]
  3. Bylund, S.; Kobyletzki, L.B.; Svalstedt, M.; Svensson, A. Prevalence and Incidence of Atopic Dermatitis: A Systematic Review. Acta Derm. Venereol. 2020, 100, adv00160. [Google Scholar] [CrossRef] [PubMed]
  4. Blicharz, L.; Szymanek-Majchrzak, K.; Mlynarczyk, G.; Czuwara, J.; Waskiel-Burnat, A.; Goldust, M.; Samochocki, Z.; Rudnicka, L. Multilocus-sequence typing reveals clonality of Staphylococcus aureus in atopic dermatitis. Clin. Exp. Dermatol. 2023, 48, 1341–1346. [Google Scholar] [CrossRef]
  5. Orfali, R.L.; Yoshikawa, F.S.Y.; Oliveira, L.; Pereira, N.Z.; de Lima, J.F.; Ramos YÁ, L.; Duarte, A.; Sato, M.N.; Aoki, V. Staphylococcal enterotoxins modulate the effector CD4+ T cell response by reshaping the gene expression profile in adults with atopic dermatitis. Sci. Rep. 2019, 9, 13082. [Google Scholar] [CrossRef]
  6. Orfali, R.L.; da Silva Oliveira, L.M.; de Lima, J.F.; de Carvalho, G.C.; Ramos, Y.A.L.; Pereira, N.Z.; Pereira, N.V.; Zaniboni, M.C.; Sotto, M.N.; da Silva Duarte, A.J.; et al. Staphylococcus aureus enterotoxins modulate IL-22-secreting cells in adults with atopic dermatitis. Sci. Rep. 2018, 8, 6665. [Google Scholar] [CrossRef] [PubMed]
  7. Garcovich, S.; Maurelli, M.; Gisondi, P.; Peris, K.; Yosipovitch, G.; Girolomoni, G. Pruritus as a Distinctive Feature of Type 2 Inflammation. Vaccines 2021, 9, 303. [Google Scholar] [CrossRef]
  8. Steinhoff, M.; Ahmad, F.; Pandey, A.; Datsi, A.; AlHammadi, A.; Al-Khawaga, S.; Al-Malki, A.; Meng, J.; Alam, M.; Buddenkotte, J. Neuroimmune communication regulating pruritus in atopic dermatitis. J. Allergy Clin. Immunol. 2022, 149, 1875–1898. [Google Scholar] [CrossRef]
  9. Agelopoulos, K.; Renkhold, L.; Wiegmann, H.; Dugas, M.; Suer, A.; Zeidler, C.; Schmelz, M.; Pereira, M.P.; Stander, S. Transcriptomic, Epigenomic, and Neuroanatomic Signatures Differ in Chronic Prurigo, Atopic Dermatitis, and Brachioradial Pruritus. J. Investig. Dermatol. 2023, 143, 264–272.e263. [Google Scholar] [CrossRef] [PubMed]
  10. Simpson, E.L.; Tom, W.L.; Bushmakin, A.G.; Cappelleri, J.C.; Yosipovitch, G.; Stander, S.; Luger, T.; Sanders, P.; Gerber, R.A.; Myers, D.E. Relationship Among Treatment, Pruritus, Investigator’s Static Global Assessment, and Quality of Life in Patients with Atopic Dermatitis. Dermatol. Ther. 2021, 11, 587–598. [Google Scholar] [CrossRef]
  11. Alexander, H.; Paller, A.S.; Traidl-Hoffmann, C.; Beck, L.A.; De Benedetto, A.; Dhar, S.; Girolomoni, G.; Irvine, A.D.; Spuls, P.; Su, J.; et al. The role of bacterial skin infections in atopic dermatitis: Expert statement and review from the International Eczema Council Skin Infection Group. Br. J. Dermatol. 2019, 182, 1331–1342. [Google Scholar] [CrossRef]
  12. Kim, J.; Kim, B.E.; Leung, D.Y.M. Pathophysiology of atopic dermatitis: Clinical implications. Allergy Asthma Proc. 2019, 40, 84–92. [Google Scholar] [CrossRef]
  13. Simpson, E.L.; Villarreal, M.; Jepson, B.; Rafaels, N.; David, G.; Hanifin, J.; Taylor, P.; Boguniewicz, M.; Yoshida, T.; De Benedetto, A.; et al. Patients with Atopic Dermatitis Colonized with Staphylococcus aureus Have a Distinct Phenotype and Endotype. J. Invest. Dermatol. 2018, 138, 2224–2233. [Google Scholar] [CrossRef]
  14. Kim, J.; Kim, B.E.; Ahn, K.; Leung, D.Y.M. Interactions Between Atopic Dermatitis and Staphylococcus aureus Infection: Clinical Implications. Allergy Asthma Immunol. Res. 2019, 11, 593–603. [Google Scholar] [CrossRef]
  15. Oliveira, D.; Borges, A.; Simoes, M. Staphylococcus aureus Toxins and Their Molecular Activity in Infectious Diseases. Toxins 2018, 10, 252. [Google Scholar] [CrossRef]
  16. Vandenesch, F.; Lina, G.; Henry, T. Staphylococcus aureus hemolysins, bi-component leukocidins, and cytolytic peptides: A redundant arsenal of membrane-damaging virulence factors? Front. Cell Infect. Microbiol. 2012, 2, 12. [Google Scholar] [CrossRef]
  17. Kim, J.H.; Lee, J.; Park, J.; Gho, Y.S. Gram-negative and Gram-positive bacterial extracellular vesicles. Semin. Cell Dev. Biol. 2015, 40, 97–104. [Google Scholar] [CrossRef]
  18. Gill, S.; Catchpole, R.; Forterre, P. Extracellular membrane vesicles in the three domains of life and beyond. FEMS Microbiol. Rev. 2019, 43, 273–303. [Google Scholar] [CrossRef] [PubMed]
  19. Ellen, A.F.; Albers, S.V.; Huibers, W.; Pitcher, A.; Hobel, C.F.; Schwarz, H.; Folea, M.; Schouten, S.; Boekema, E.J.; Poolman, B.; et al. Proteomic analysis of secreted membrane vesicles of archaeal Sulfolobus species reveals the presence of endosome sorting complex components. Extremophiles 2009, 13, 67–79. [Google Scholar] [CrossRef] [PubMed]
  20. Dagnelie, M.A.; Corvec, S.; Khammari, A.; Dreno, B. Bacterial extracellular vesicles: A new way to decipher host-microbiota communications in inflammatory dermatoses. Exp. Dermatol. 2020, 29, 22–28. [Google Scholar] [CrossRef] [PubMed]
  21. Bose, S.; Aggarwal, S.; Singh, D.V.; Acharya, N. Extracellular vesicles: An emerging platform in gram-positive bacteria. Microb. Cell 2020, 7, 312–322. [Google Scholar] [CrossRef]
  22. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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. [Google Scholar] [CrossRef] [PubMed]
  23. Lee, E.Y.; Choi, D.Y.; Kim, D.K.; Kim, J.W.; Park, J.O.; Kim, S.; Kim, S.H.; Desiderio, D.M.; Kim, Y.K.; Kim, K.P.; et al. Gram-positive bacteria produce membrane vesicles: Proteomics-based characterization of Staphylococcus aureus-derived membrane vesicles. Proteomics 2009, 9, 5425–5436. [Google Scholar] [CrossRef] [PubMed]
  24. Hong, S.W.; Kim, M.R.; Lee, E.Y.; Kim, J.H.; Kim, Y.S.; Jeon, S.G.; Yang, J.M.; Lee, B.J.; Pyun, B.Y.; Gho, Y.S.; et al. Extracellular vesicles derived from Staphylococcus aureus induce atopic dermatitis-like skin inflammation. Allergy 2011, 66, 351–359. [Google Scholar] [CrossRef] [PubMed]
  25. Hong, S.W.; Choi, E.B.; Min, T.K.; Kim, J.H.; Kim, M.H.; Jeon, S.G.; Lee, B.J.; Gho, Y.S.; Jee, Y.K.; Pyun, B.Y.; et al. An important role of α-hemolysin in extracellular vesicles on the development of atopic dermatitis induced by Staphylococcus aureus. PLoS ONE 2014, 9, e100499. [Google Scholar] [CrossRef] [PubMed]
  26. Kim, M.H.; Rho, M.; Choi, J.P.; Choi, H.I.; Park, H.K.; Song, W.J.; Min, T.K.; Cho, S.H.; Cho, Y.J.; Kim, Y.K.; et al. A Metagenomic Analysis Provides a Culture-Independent Pathogen Detection for Atopic Dermatitis. Allergy Asthma Immunol. Res. 2017, 9, 453–461. [Google Scholar] [CrossRef] [PubMed]
  27. Jun, S.H.; Lee, J.H.; Kim, S.I.; Choi, C.W.; Park, T.I.; Jung, H.R.; Cho, J.W.; Kim, S.H.; Lee, J.C. Staphylococcus aureus-derived membrane vesicles exacerbate skin inflammation in atopic dermatitis. Clin. Exp. Allergy 2017, 47, 85–96. [Google Scholar] [CrossRef] [PubMed]
  28. Kwon, H.I.; Jeong, N.H.; Jun, S.H.; Son, J.H.; Kim, S.; Jeon, H.; Kang, S.C.; Kim, S.H.; Lee, J.C. Thymol attenuates the worsening of atopic dermatitis induced by Staphylococcus aureus membrane vesicles. Int. Immunopharmacol. 2018, 59, 301–309. [Google Scholar] [CrossRef]
  29. Kim, M.H.; Choi, S.J.; Choi, H.I.; Choi, J.P.; Park, H.K.; Kim, E.K.; Kim, M.J.; Moon, B.S.; Min, T.K.; Rho, M.; et al. Lactobacillus plantarum-derived Extracellular Vesicles Protect Atopic Dermatitis Induced by Staphylococcus aureus-derived Extracellular Vesicles. Allergy Asthma Immunol. Res. 2018, 10, 516–532. [Google Scholar] [CrossRef]
  30. Kim, J.; Bin, B.H.; Choi, E.J.; Lee, H.G.; Lee, T.R.; Cho, E.G. Staphylococcus aureus-derived extracellular vesicles induce monocyte recruitment by activating human dermal microvascular endothelial cells in vitro. Clin. Exp. Allergy 2019, 49, 68–81. [Google Scholar] [CrossRef]
  31. Yang, J.; McDowell, A.; Seo, H.; Kim, S.; Min, T.K.; Jee, Y.K.; Choi, Y.; Park, H.S.; Pyun, B.Y.; Kim, Y.K. Diagnostic Models for Atopic Dermatitis Based on Serum Microbial Extracellular Vesicle Metagenomic Analysis: A Pilot Study. Allergy Asthma Immunol. Res. 2020, 12, 792–805. [Google Scholar] [CrossRef]
  32. Wang, X.; Koffi, P.F.; English, O.F.; Lee, J.C. Staphylococcus aureus Extracellular Vesicles: A Story of Toxicity and the Stress of 2020. Toxins 2021, 13, 75. [Google Scholar] [CrossRef]
  33. Staudenmaier, L.; Focken, J.; Schlatterer, K.; Kretschmer, D.; Schittek, B. Bacterial membrane vesicles shape Staphylococcus aureus skin colonization and induction of innate immune responses. Exp. Dermatol. 2022, 31, 349–361. [Google Scholar] [CrossRef] [PubMed]
  34. Yang, J.; Shin, T.S.; Kim, J.S.; Jee, Y.K.; Kim, Y.K. A new horizon of precision medicine: Combination of the microbiome and extracellular vesicles. Exp. Mol. Med. 2022, 54, 466–482. [Google Scholar] [CrossRef] [PubMed]
  35. Choi, Y.; Park, H.S.; Kim, Y.K. Bacterial Extracellular Vesicles: A Candidate Molecule for the Diagnosis and Treatment of Allergic Diseases. Allergy Asthma Immunol. Res. 2023, 15, 279–289. [Google Scholar] [CrossRef]
  36. Zhou, H.; Tan, X.; Chen, G.; Liu, X.; Feng, A.; Liu, Z.; Liu, W. Extracellular Vesicles of Commensal Skin Microbiota Alleviate Cutaneous Inflammation in Atopic Dermatitis Mouse Model by Re-Establishing Skin Homeostasis. J. Investig. Dermatol. 2023. ahead of printing. [Google Scholar] [CrossRef]
  37. Jan, A.T. Outer Membrane Vesicles (OMVs) of Gram-negative Bacteria: A Perspective Update. Front. Microbiol. 2017, 8, 1053. [Google Scholar] [CrossRef] [PubMed]
  38. Knox, K.W.; Vesk, M.; Work, E. Relation between excreted lipopolysaccharide complexes and surface structures of a lysine-limited culture of Escherichia coli. J. Bacteriol. 1966, 92, 1206–1217. [Google Scholar] [CrossRef]
  39. Briaud, P.; Carroll, R.K. Extracellular Vesicle Biogenesis and Functions in Gram-Positive Bacteria. Infect. Immun. 2020, 88, 10–1128. [Google Scholar] [CrossRef]
  40. Mobarak, H.; Javid, F.; Narmi, M.T.; Mardi, N.; Sadeghsoltani, F.; Khanicheragh, P.; Narimani, S.; Mahdipour, M.; Sokullu, E.; Valioglu, F.; et al. Prokaryotic microvesicles Ortholog of eukaryotic extracellular vesicles in biomedical fields. Cell Commun. Signal 2024, 22, 80. [Google Scholar] [CrossRef]
  41. Tartaglia, N.R.; Nicolas, A.; Rodovalho, V.R.; Luz, B.S.R.D.; Briard-Bion, V.; Krupova, Z.; Thierry, A.; Coste, F.; Burel, A.; Martin, P.; et al. Extracellular vesicles produced by human and animal Staphylococcus aureus strains share a highly conserved core proteome. Sci. Rep. 2020, 10, 8467. [Google Scholar] [CrossRef]
  42. Wang, X.; Eagen, W.J.; Lee, J.C. Orchestration of human macrophage NLRP3 inflammasome activation by. Proc. Natl. Acad. Sci. USA 2020, 117, 3174–3184. [Google Scholar] [CrossRef] [PubMed]
  43. Tartaglia, N.R.; Breyne, K.; Meyer, E.; Cauty, C.; Jardin, J.; Chrétien, D.; Dupont, A.; Demeyere, K.; Berkova, N.; Azevedo, V.; et al. Extracellular Vesicles Elicit an Immunostimulatory Response. Front. Cell Infect. Microbiol. 2018, 8, 277. [Google Scholar] [CrossRef] [PubMed]
  44. Lee, J.; Lee, E.Y.; Kim, S.H.; Kim, D.K.; Park, K.S.; Kim, K.P.; Kim, Y.K.; Roh, T.Y.; Gho, Y.S. Staphylococcus aureus extracellular vesicles carry biologically active β-lactamase. Antimicrob. Agents Chemother. 2013, 57, 2589–2595. [Google Scholar] [CrossRef]
  45. Andreoni, F.; Toyofuku, M.; Menzi, C.; Kalawong, R.; Mairpady Shambat, S.; Francois, P.; Zinkernagel, A.S.; Eberl, L. Antibiotics Stimulate Formation of Vesicles in Staphylococcus aureus in both Phage-Dependent and -Independent Fashions and via Different Routes. Antimicrob. Agents Chemother. 2019, 63, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  46. Seiti Yamada Yoshikawa, F.; Feitosa de Lima, J.; Notomi Sato, M.; Álefe Leuzzi Ramos, Y.; Aoki, V.; Leao Orfali, R. Exploring the Role of Staphylococcus aureus Toxins in Atopic Dermatitis. Toxins 2019, 11, 321. [Google Scholar] [CrossRef] [PubMed]
  47. Bantel, H.; Sinha, B.; Domschke, W.; Peters, G.; Schulze-Osthoff, K.; Jänicke, R.U. alpha-Toxin is a mediator of Staphylococcus aureus-induced cell death and activates caspases via the intrinsic death pathway independently of death receptor signaling. J. Cell Biol. 2001, 155, 637–648. [Google Scholar] [CrossRef] [PubMed]
  48. Essmann, F.; Bantel, H.; Totzke, G.; Engels, I.H.; Sinha, B.; Schulze-Osthoff, K.; Janicke, R.U. Staphylococcus aureus alpha-toxin-induced cell death: Predominant necrosis despite apoptotic caspase activation. Cell Death Differ. 2003, 10, 1260–1272. [Google Scholar] [CrossRef] [PubMed]
  49. Jia, N.; Li, G.; Wang, X.; Cao, Q.; Chen, W.; Wang, C.; Chen, L.; Ma, X.; Zhang, X.; Tao, Y.; et al. Staphylococcal superantigen-like protein 10 induces necroptosis through TNFR1 activation of RIPK3-dependent signal pathways. Commun. Biol. 2022, 5, 813. [Google Scholar] [CrossRef]
  50. Kulp, A.; Kuehn, M.J. Biological functions and biogenesis of secreted bacterial outer membrane vesicles. Annu. Rev. Microbiol. 2010, 64, 163–184. [Google Scholar] [CrossRef]
  51. Sugaya, M. The Role of Th17-Related Cytokines in Atopic Dermatitis. Int. J. Mol. Sci. 2020, 21, 1314. [Google Scholar] [CrossRef]
  52. Grice, E.A.; Kong, H.H.; Conlan, S.; Deming, C.B.; Davis, J.; Young, A.C.; Program, N.C.S.; Bouffard, G.G.; Blakesley, R.W.; Murray, P.R.; et al. Topographical and temporal diversity of the human skin microbiome. Science 2009, 324, 1190–1192. [Google Scholar] [CrossRef]
  53. Kong, H.H.; Oh, J.; Deming, C.; Conlan, S.; Grice, E.A.; Beatson, M.A.; Nomicos, E.; Polley, E.C.; Komarow, H.D.; Program, N.C.S.; et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012, 22, 850–859. [Google Scholar] [CrossRef] [PubMed]
  54. Alenius, H.; Sinkko, H.; Moitinho-Silva, L.; Rodriguez, E.; Broderick, C.; Alexander, H.; Reiger, M.; Hjelmso, M.H.; Fyhrquist, N.; Olah, P.; et al. The power and potential of BIOMAP to elucidate host-microbiome interplay in skin inflammatory diseases. Exp. Dermatol. 2021, 30, 1517–1531. [Google Scholar] [CrossRef]
  55. Rayner, S.; Bruhn, S.; Vallhov, H.; Andersson, A.; Billmyre, R.B.; Scheynius, A. Identification of small RNAs in extracellular vesicles from the commensal yeast Malassezia sympodialis. Sci. Rep. 2017, 7, 39742. [Google Scholar] [CrossRef]
  56. Byrd, A.L.; Deming, C.; Cassidy, S.K.B.; Harrison, O.J.; Ng, W.I.; Conlan, S.; Belkaid, Y.; Segre, J.A.; Kong, H.H. Staphylococcus aureus and Staphylococcus epidermidis strain diversity underlying pediatric atopic dermatitis. Sci. Transl. Med. 2017, 9, eaal4651. [Google Scholar] [CrossRef] [PubMed]
  57. Kobiela, A.; Hovhannisyan, L.; Jurkowska, P.; de la Serna, J.B.; Bogucka, A.; Deptula, M.; Paul, A.A.; Panek, K.; Czechowska, E.; Rychlowski, M.; et al. Excess filaggrin in keratinocytes is removed by extracellular vesicles to prevent premature death and this mechanism can be hijacked by Staphylococcus aureus in a TLR2-dependent fashion. J. Extracell. Vesicles 2023, 12, e12335. [Google Scholar] [CrossRef]
  58. Shao, S.; Fang, H.; Li, Q.; Wang, G. Extracellular vesicles in Inflammatory Skin Disorders: From Pathophysiology to Treatment. Theranostics 2020, 10, 9937–9955. [Google Scholar] [CrossRef] [PubMed]
  59. Cho, B.S.; Kim, J.O.; Ha, D.H.; Yi, Y.W. Exosomes derived from human adipose tissue-derived mesenchymal stem cells alleviate atopic dermatitis. Stem Cell Res. Ther. 2018, 9, 187. [Google Scholar] [CrossRef]
  60. Kim, J.; Lee, S.K.; Jung, M.; Jeong, S.Y.; You, H.; Won, J.Y.; Han, S.D.; Cho, H.J.; Park, S.; Park, J.; et al. Extracellular vesicles from IFN-gamma-primed mesenchymal stem cells repress atopic dermatitis in mice. J. Nanobiotechnol. 2022, 20, 526. [Google Scholar] [CrossRef]
  61. Alberro, A.; Iparraguirre, L.; Fernandes, A.; Otaegui, D. Extracellular Vesicles in Blood: Sources, Effects, and Applications. Int. J. Mol. Sci. 2021, 22, 8163. [Google Scholar] [CrossRef] [PubMed]
  62. Schaack, B.; Hindre, T.; Quansah, N.; Hannani, D.; Mercier, C.; Laurin, D. Microbiota-Derived Extracellular Vesicles Detected in Human Blood from Healthy Donors. Int. J. Mol. Sci. 2022, 23, 3787. [Google Scholar] [CrossRef] [PubMed]
  63. Jones, E.; Stentz, R.; Telatin, A.; Savva, G.M.; Booth, C.; Baker, D.; Rudder, S.; Knight, S.C.; Noble, A.; Carding, S.R. The Origin of Plasma-Derived Bacterial Extracellular Vesicles in Healthy Individuals and Patients with Inflammatory Bowel Disease: A Pilot Study. Genes 2021, 12, 1636. [Google Scholar] [CrossRef] [PubMed]
  64. Laakmann, K.; Eckersberg, J.M.; Hapke, M.; Wiegand, M.; Bierwagen, J.; Beinborn, I.; Preusser, C.; Pogge von Strandmann, E.; Heimerl, T.; Schmeck, B.; et al. Bacterial extracellular vesicles repress the vascular protective factor RNase1 in human lung endothelial cells. Cell Commun. Signal 2023, 21, 111. [Google Scholar] [CrossRef]
  65. Morales-Sanfrutos, J.; Munoz, J. Unraveling the complexity of the extracellular vesicle landscape with advanced proteomics. Expert. Rev. Proteom. 2022, 19, 89–101. [Google Scholar] [CrossRef]
  66. Wang, S.; He, B.; Wu, H.; Cai, Q.; Ramirez-Sanchez, O.; Abreu-Goodger, C.; Birch, P.R.J.; Jin, H. Plant mRNAs move into a fungal pathogen via extracellular vesicles to reduce infection. Cell Host Microbe 2024, 32, 93–105 e106. [Google Scholar] [CrossRef]
Figure 1. Review search terms. Graphical strategy and search results. The applied keywords and the main results of the search.
Figure 1. Review search terms. Graphical strategy and search results. The applied keywords and the main results of the search.
Microorganisms 12 00531 g001
Figure 2. S. Aureus-derived EVs in AD pathogenesis. The effects of S. aureus-derived extracellular vesicles on skin cells are diverse. They can induce fibroblasts to secrete IL-6, CCL2, TSLP, and Eotaxin (1), and keratinocytes (KC) to secrete CXCL-8, IL-6, IL-1β, and TNF-α (2), all of which are proinflammatory mediators. Dermal microvascular endothelial cells upregulate the expression of the adhesion molecules E-selectin, VCAM1, and ICAM1 (3), which contribute to leukocyte migration (4). Secretion of CXCL-8 by KC also contributes to the cell migration, resulting in an increased polymorphonuclear cell (PMN) infiltration (5). Additionally, S. aureus-derived EVs can have a cytotoxic effect on KC, culminating in necrosis and skin barrier disruption (6). All these events together lead to eosinophilic inflammation in the dermis (7), epidermal hyperplasia (8), upregulation of proinflammatory mediators that elicit the Th2/Th17/Th1 response and augmented production of IgE. ↑: increased expression. Figure created with BioRender.com (accessed on 9 February 2024).
Figure 2. S. Aureus-derived EVs in AD pathogenesis. The effects of S. aureus-derived extracellular vesicles on skin cells are diverse. They can induce fibroblasts to secrete IL-6, CCL2, TSLP, and Eotaxin (1), and keratinocytes (KC) to secrete CXCL-8, IL-6, IL-1β, and TNF-α (2), all of which are proinflammatory mediators. Dermal microvascular endothelial cells upregulate the expression of the adhesion molecules E-selectin, VCAM1, and ICAM1 (3), which contribute to leukocyte migration (4). Secretion of CXCL-8 by KC also contributes to the cell migration, resulting in an increased polymorphonuclear cell (PMN) infiltration (5). Additionally, S. aureus-derived EVs can have a cytotoxic effect on KC, culminating in necrosis and skin barrier disruption (6). All these events together lead to eosinophilic inflammation in the dermis (7), epidermal hyperplasia (8), upregulation of proinflammatory mediators that elicit the Th2/Th17/Th1 response and augmented production of IgE. ↑: increased expression. Figure created with BioRender.com (accessed on 9 February 2024).
Microorganisms 12 00531 g002
Table 1. Milestones in SA-derived EVs and AD research.
Table 1. Milestones in SA-derived EVs and AD research.
YearMain FindingsReference
2009First description that S. aureus spontaneously producing EVsLee et al. [23]
2011SA-derived EVs induce AD-like inflammation in the skin and should be considered as novel diagnostic and therapeutic target for the control of ADHong et al. [24]
2014EV-associated α-hemolysin induces necrosis of keratinocytes, skin barrier disruption and epidermal hyperplasiaHong et al. [25]
2017Metagenomic analysis together with serum detection of pathogen-specific EVs provides a model for identification and diagnosis of pathogens of ADKim et al. [26]
2017SA-derived EVs as potent mediator for exacerbation of AD severityJun et al. [27]
2018Thymol disrupts SA-derived EVs and suppresses inflammatory responses in AD-like skin lesions aggravated by S. aureus EVsKwon et al. [28]
2018L. plantarum-derived EVs might help prevent skin inflammationKim et al. [29]
2019SA-derived EVs as proinflammatory factors could mediate immune cell infiltration in AD by efficiently inducing endothelial cell activation and monocyte recruitmentKim et al. [30]
2020Bacterial EVs carry several types of molecules: proteins, glycoproteins, mRNAs and small RNA species, as mammalian EVs do, but also carbohydratesDagnelie et al. [20]
2020A pilot study indicates microbial EVs as potential biomarkers for AD diagnosisYang et al. [31]
2020EVs from Gram-positive bacteria carry a diversity of cargo compounds that have a role in bacterial competition, survival, invasion, host immune evasion, and infectionBose et al. [21]
2021EVs represent a novel S. aureus secretory system that is affected by a variety of stress responses and allows the delivery of biologically active pore-forming toxins and other virulence determinants to host cellsWang et al. [32]
2021EVs of S. aureus strains from the lesional skin of AD patients show an enhanced membrane lipid and protein A content compared to the strains from the non-lesional sites with enhanced proinflammatory potentialStaudenmaier et al. [33]
2022Microbial EV therapy may offer a variety of benefits over live biotherapeutics and human cell EV (or exosome) therapy for the treatment of intractable diseasesYang et al. [34]
2023Bacterial EVs may exert diverse effects on immune responses both beneficial or pathogenic role in patients with allergic and immunologic diseasesChoi et al. [35]
2023SA-derived EVs reduced AD-like skin inflammation in mice and may potentially be a bioactive nanocarrier for the treatment of ADZhou et al. [36]
AD: atopic dermatitis; EVs: extracellular vesicles; S. aureus: Staphylococcus aureus; L. plantarum: Lactobacillus plantarum.
Table 2. Biological effects of SA-derived EVs.
Table 2. Biological effects of SA-derived EVs.
S. aureus StrainStudy TypeExperimental ModelInflammatory
Effector
Molecules
Upregulated
Histological
Features
Others
Observed Effects
Ref.
ATCC14458In vivoEV were applied by tape stripping mouse skinIL-4, IL-5, IL-17, IFN-γInfiltration of polymorphonuclear cells and epidermal thickness-[24]
03ST17In vivoTopical application of EVs into DFE induced lesions on AD-like mouse model;IL-13, IL-31, CXCL8, CCL2 and CCL3Infiltration of polymorphonuclear cells and epidermal thicknessSevere eczematous dermatitis, swelling, redness, bullae, and eschar formation[27]
USA300In vitroPrimary human keratinocytesCXCL8 and TNF-α-Recruitment of neutrophils and induction of NETs[33]
ATCC 6538In vitroImmortalized human dermal microvascular endothelial cells E-selectin, VCAM1, ICAM1 and IL-6-Recruitment of monocytes[30]
ATCC14458 and from AD patientsIn vitroImmortalized human keratinocytesIL-1β and IL-6-Cytotoxic effect associated with EV-α-Hemolysin[25]
ATCC14458In vitroPrimary mouse dermal fibroblastsIL-6, TSLP, CCL2 and Eotaxin--[24]
03ST17In vitroImmortalized human keratinocytesIL-6--[27]
S. aureus: Staphylococcus aureus; AD: atopic dermatitis; EV: extracellular vesicles; NETs: neutrophil extracellular traps; Ref.: references; DFE: Dermatophagoides farinae extract.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Torrealba, M.P.; Yoshikawa, F.S.Y.; Aoki, V.; Sato, M.N.; Orfali, R.L. State of the Art on the Role of Staphylococcus aureus Extracellular Vesicles in the Pathogenesis of Atopic Dermatitis. Microorganisms 2024, 12, 531. https://doi.org/10.3390/microorganisms12030531

AMA Style

Torrealba MP, Yoshikawa FSY, Aoki V, Sato MN, Orfali RL. State of the Art on the Role of Staphylococcus aureus Extracellular Vesicles in the Pathogenesis of Atopic Dermatitis. Microorganisms. 2024; 12(3):531. https://doi.org/10.3390/microorganisms12030531

Chicago/Turabian Style

Torrealba, Marina Passos, Fabio Seiti Yamada Yoshikawa, Valeria Aoki, Maria Notomi Sato, and Raquel Leão Orfali. 2024. "State of the Art on the Role of Staphylococcus aureus Extracellular Vesicles in the Pathogenesis of Atopic Dermatitis" Microorganisms 12, no. 3: 531. https://doi.org/10.3390/microorganisms12030531

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

Article Metrics

Back to TopTop