Fungal Extracellular Vesicles: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Extracellular vesicles (EVs) are membranous vesicular organelles that perform a variety of biological functions including cell communication across different biological kingdoms. EVs of mammals and, to a lesser extent, bacteria have been deeply studied over the years, whereas investigations of fungal EVs are still in their infancy. Fungi, encompassing both yeast and filamentous forms, are increasingly recognized for their production of extracellular vesicles (EVs) containing a wealth of proteins, lipids, and nucleic acids. These EVs play pivotal roles in orchestrating fungal communities, bolstering pathogenicity, and mediating interactions with the environment. 

  • extracellular vesicles
  • fungi

1. Introduction

Extracellular vesicles (EVs) are defined as cup-sized nanostructures delimited by lipid bilayers [1]. The International Society for Extracellular Vesicles (ISEV) proposed classifying EVs based on their sedimentation speed [2], but most scientists and authors still use a former classification based on the origin and size of EVs, dividing them into exosomes and microvesicles (also called microparticles or ectosomes), which are shared across almost all domains of life [3], and apoptotic bodies, whose presence in fungi is currently under investigation [4]. The debate regarding EV classification is also still unresolved because there is not a universal consensus upon the most reliable methods of isolation, purification and characterisation [5][6][7][8][9][10]. The content of EVs can be very heterogeneous, and mainly includes proteins, lipids and nucleic material [11].
Since their early descriptions as “clotting factors” [12] or “platelet dust” [13], researchers' knowledge on extracellular vesicles (EVs) has tremendously expanded, such that they are currently used in biotechnological, diagnostic and therapeutic applications [14][15][16]. However, several aspects of EVs, such as biogenesis, cargo and releasing mechanisms differ among species and are still far from being wholly understood [17]. Fungal EVs are a recent research field, since they were properly identified for the first time in 2007 in Cryptococcus neoformans [18]. Thanks to their cargo, EVs are used by fungal species to modulate a series of different functions within the fungal community [19]. For instance, they participate in intercellular communication during biofilm formation [20][21], regulate the intracellular proliferation of a population [22], play a critical role in cell wall remodelling [23] and could even mediate the vertical and horizontal transfer of prion-like protein in fungi [24]. In addition to all the intra-kingdom functions, fungal EVs are known to help releasing cells during their interactions with other organisms, such as other fungi and bacteria, or with the host, including animals and plants [4][25]. The interactions between fungi and their host through fungal EVs have been studied especially during infections in mammalian cells, where fungal EVs can exacerbate or attenuate fungal infection by enhancing pathogenicity or modulating virulence strategies [2][26][27][28]. Fungal EVs show the capacity to enter host cells and start a process that can lead to the modulation of antimicrobial activities and immune responses. Several studies investigated the effects of EVs from pathogenic fungi on murine and human immune cells, including macrophages [29][30], keratinocytes [31], dendritic cells (DCs) [32] and neutrophils [33]. These interactions lead to the activation of the innate immune system, but also of the priming activity of T cells through several mechanisms, including the production of pro-inflammatory and anti-inflammatory cytokines.
In addition to fungi, the bigger portion of the literature about the interactions between microbial EVs and the host immune system relies on bacteria. In the last twenty years, there has been a great number of studies regarding EVs both from pathogens [34] and commensal bacteria [35]. Several Gram-negative commensals have been investigated [36][37], while Gram-positive studies have focused on probiotics of the genera Lactobacillus and Bifidobacterium [38][39]. Research on EVs in Gram-positive bacteria, mycobacteria and fungi was neglected until recently, due the presumption that vesicles could not traverse the thick cell walls found in these organisms [40]. Despite the later discovery, EVs from these microorganisms are now comprehensively studied, and share common features, including the delivery of multiple virulence factors that elicit strong immune responses in the host [41].
The current available information on the applications of EVs in drug delivery provides significant groundwork for their development as delivery vehicles to harness their potential for future therapeutic applications. Fungal EVs could represent novel biotechnological tools for diagnostics and immunotherapy. Fungal EVs have been shown to possess most of the characteristics for an ideal vaccine, such as nanometric sizes, being g naturally carriers of multiple antigens, and conjugation with sugars, which can contribute to cell recognition [42].
The increased number of immunocompromised patients [43], and the spread of new fungal pathogens due to environmental and climate change [44][45][46] have significantly modified the rate of fungal infections among humans around the globe, as attested by the Global Action Fund for Fungal Infections (GAFFI) [47][48]. These dramatic data, together with the low efficacy of currently approved antifungal drugs and the outbreak of resistant fungal strains [49][50][51], outline the urgent need for new strategies to both prevent and fight fungal infections.

2. Fungal Fungal Extracellular Vesicles

Fungal EVs have been described in more than twenty different species [19], and it is now assumed that they represent a universal mechanism for the transport of molecules outside the fungal cell [52][53]

2.1. Discovery and Seminal Studies

The biological entities that would later be named extracellular vesicles were first described in 1946 by Chargaff and West, who were working on blood coagulation [12]. Only in 1967 did another study by Wolf describe them as platelet-derived material [13]. In fungi, the term “extracellular vesicles” was used for the first time in 1977, in a study on Candida tropicalis [54]. Before that, both Gibson and Peberdy in 1972 [55] and Takeo and colleagues in 1973 [56] reported the presence of outer membranous particles in Aspergillus nidulans and Cryptococcus neoformans, respectively. Despite the fact that in 1990 Anderson and colleagues found wall-crossing vesicles in Candida albicans [57], all of the aforementioned studies did not promote research on fungal EVs, mostly because the thick cell wall of fungi seemed to preclude the production of vesicles destined to the extracellular space [40]. The first set of experimental evidence suggesting the production of EVs by fungi was carried out at the end of the last century, when an attempt to develop monoclonal antibodies against Cryptococcus neoformans was made by Arturo Casadevall, as reported in the footnote of the first book on fungal EVs [58]. Since lipid structures were found in the fungal cell wall, Rodrigues and Nimrichter hypothesised the presence of hydrophobic structures that could reach the cell wall [59]. Following this hypothesis, in 2007 Rodrigues and colleagues isolated EVs from C. neoformans cultures [18], and right after this they demonstrated that these vesicles contain glucuronoxylomannan (GXM), which is the major virulence factor of C. neoformans [60]. Around the same period, Vallejo and colleagues showed that EV production occurs in Paracoccidioides brasiliensis [61]. To date, EVs have been found in more than 20 fungal species, in both yeast-like and filamentous forms, such that the production of EVs is now assumed to be a communication mechanism shared by the whole fungal kingdom. An almost complete list of these species can be found in previous comprehensive reviews [4][19], except (as far as researchers know) for the pathogenic yeast Candida auris, whose EVs have been characterised in the last year [62][63][64]. Fungal EVs have been investigated mostly in human infections and, more generally, in the context of interactions with the human host. Nonetheless, fungal EVs have been studied also regarding their functions in parasitic or mutualistic relationships with plants [25][65][66], defence from predators [67], cell wall synthesis [68] and remodelling [23][61][69], and other physiological roles within fungal communities [70], including morphological transitions during biofilm formation [20][21], the regulation of protein cargo related to nutrients [71][72][73][74] and adaptation to the matrix [75][76][77].

2.2. Biogenesis and Release of Fungal EVs

Despite the fact that studies on the production of fungal EVs have been conducted for more than a decade, the mechanisms of their biogenesis are still unknown. Nevertheless, proteomic, genetic and in silico analyses have provided insights into certain molecular patterns related to EV trafficking, which are already extensively discussed elsewhere [4][19][27][78][79][80][81].
Mammalian vesicles, which have been far more frequently investigated than fungal ones have, are finely classified in different subpopulations, depending on the size and origin [82][83][84][85]. Currently, studies on fungal EVs also divide them into exosomes, which correspond roughly to small EVs in the ISEV classification and are smaller than 150 nm, and microvesicles, which correspond to medium EVs and are larger than 150 nm [27]. The most important difference between exosomes and microvesicles, apart from size, is the mechanism of biogenesis. Exosomes derive from the endosomal pathway, through the formation of multivesicular bodies (MVBs) which contain several vesicles. They fuse with the plasma membrane releasing them into the intraluminal space. On the other hand, microvesicles (or ectosomes) originate directly from projections of the plasma membrane [78].
Exosome biogenesis, release and cargo selection have been shown to be related with both conventional and unconventional secretory pathways, thanks to genetic deletions, but none of the studied mutations were able to completely cease EV production, suggesting a co-occurrence of the multiple known pathways or the presence of currently unknown ones. Firstly, mutations in SEC genes, which play a role in the post-Golgi conventional secretion pathway, cause an accumulation of vesicles in the cytoplasm [86][87] or vesicles of altered diameters [88] in S. cerevisiae, and a lack of EV detection in C. neoformans [89]. Regarding the unconventional secretion pathways, the most important regulators of EVs production known so far are involved in MVB formation and release. Specifically, a lack of expression of Golgi reassembly and stacking protein (GRASP) causes a reduction in EV release in S. cerevisiae [88] and different EV sizes in C. neoformans [90]. Moreover, a class of proteins of vacuolar protein sorting (VPS) named E-VPS is crucial in the endosomal sorting complexes required for transport (ESCRT) machinery [91][92][93], which coordinate MVB release [94]. Therefore, it has been shown that the deletion of different VPS genes strongly affects the biogenesis of EVs in S. cerevisiae [23][88], C. neoformans [95][96][97][98] and C. albicans [21][99].
The production and release of microvesicles via the invagination of the plasma membrane was shown two decades ago [59], together with observations of cytoplasmic volume loss [88] and cytoplasmic proteins in fungal EVs [100]. Moreover, mutations in genes involved in the composition of the plasma membrane affect EV formation and content [76][101][102][103]
Another fungal EV-related process that still needs to be fully understood is cell wall crossing. The fungal cell wall is composed of a core set of molecules that are conserved across the fungal kingdom, mainly beta-glucans, mannoproteins and chitin [104], and of components that are species-specific, such that the wall is probably the part of the cell that exhibits the most diversity and plasticity [105]. Fungal cell walls are nowadays considered dynamic structures whose compositions and pores’ dimensions are highly regulated via environmental conditions [106][107]. Despite this, they have been shown to act as barriers to the passage of EVs, since mutations in cell wall-remodelling genes increase their release [23][98], and an increase in their thickness can lead to an accumulation of vesicles in the area between the plasma membrane and cell wall [108]. Moreover, it still remains poorly understood how vesicles as large as almost 1 μm could pass through these dense polysaccharide/protein matrices. Three nonexclusive hypotheses, already reviewed elsewhere [40][106], are currently under investigation: (i) vesicles can pass the cell wall through guide channels; (ii) there are specific enzymes that could open areas in the wall; (iii) turgor pressure and/or other physical forces could force the passage of EVs through cell wall pores. The last hypothesis has received support via a recent study which demonstrated that the cell wall has viscoelastic properties, which could allow the passage of EVs [109]. However, since that study utilised small liposomes (60–80 nm), the full explanation for the passage of EVs with a broad spectrum of sizes has yet to be found.

2.3. Composition of Fungal EVs

The process of the production and release of EVs is conserved across kingdoms even if it has a considerable energetic cost for the cell, and the reason for that has to be found in the multiple advantages of vesicle packaging compared to soluble secretion directly to the extracellular milieu. Firstly, vesicles protect easily degradable molecules, such as RNAs. Secondly, loading molecules into a confined space prevents the loss of them. Finally, the vesicle co-transportation of more molecules enables their synergistic effect over distances [110]. As stated before, a large part of the knowledge on fungal EVs is derived from studies on mammals, since there are several resemblances between them, including their content in terms of classes of molecules [106]. The most abundant molecules packed the into EVs of both mammals and fungi belong to proteins, lipids and nucleic acids, even though there are other molecules specific to fungi, such as prions. 

2.3.1. Proteins

Proteomic studies on fungal EVs showed a common set of proteins between different species, such as those involved in the stress response, oxidation, metabolic pathways, transport and signalling, while others have proven to be species-specific [53][110][111].
EVs from pathogenic species carry proteins associated with virulence. In C. neoformans, laccases, ureases, phosphatases and heat shock proteins were found [60]; in Paracoccidioides brasiliensis, phosphatases [111]; in Histoplasma capsulatum, catalases, superoxide dismutases and cell-wall hydrolyzing enzymes [69]; in Malassezia sympodialis, multiple allergens [112]; and in C. albicans, numerous proteins related mainly to pathogenesis, cell organisation and the response to stress [113][114].
The protein content of non-pathogenic yeast EVs have also been investigated, especially in S. cerevisiae, Torulaspora delbrueckii, Hanseniaspora uvarum, Candida sake and Metschnikowia pulcherrima [88][115]. Researchers have found proteins implicated in almost every cell pathway, but the most present were heat shock and stress-related proteins, and those involved in cell wall metabolism [116]. Moreover, proteins contained in EVs from Pichia fermentans were shown to be related to dimorphic transition [20].

2.3.2. Lipids

Lipids are the major components of vesicles’ membranes, and also part of their cargo. Several lipids found in fungal EVs have important functions in pathogenesis, biofilm building and membrane formation [110][117][118]. For example, ergosterol, which is commonly found in EVs from all fungal species, is known to be crucial for biofilm building [118]. EVs from C. albicans and C. neoformans are enriched in ergosterol and glucosylceramide [102][114][119], an important immunogenic compound which has been proven to be a good target for inhibiting fungal hyphal formation during pathogenesis [59]. EVs from H. capsulatum and P. brasiliensis were found to mainly contain sterols and phospholipids, such as phosphatidylethanolamine, phosphatidylcholine, and phosphatidylserine, which are key elements in the formation of fungal lipid bilayers [69][117][120].

2.3.3. Nucleic Acids

Concerning nucleic acids, fungal EVs show strong resemblance to those of mammals, transporting several types of both coding and non-coding RNA, which have been compared and reviewed elsewhere [4][121][122][123]. RNAs are receiving special attention in the context of intercellular communication due to the several mechanisms of post-transcriptional regulation, such as those mediated by microRNAs (miRNAs), short interfering RNAs (siRNAs), tRNA-derived fragments (tRFs) and long non-coding RNAs (lncRNAs) [124][125]. However, they are commonly found in EVs across all biological kingdoms [25][126][127][128]. For instance, several studies showed cross-kingdom interactions between fungi and plants through RNA-containing EVs. Pathogen-induced gene silencing (PIGS) is a mechanism of RNAi used by several fungal pathogens [125], such as Botrytis cinerea [129], Verticillium dahliae [130], Puccinia striiformis f.sp. tritici [131] and Fusarium graminearum [132].
Apart from studies on plants, several studies were conducted on yeast species related to humans. The first one was a comparison between the RNA content of five different species, as reported by Da Silva and colleagues in 2015 [133]. These authors found that the most common shared small RNA classes in EVs belong to small nucleolar RNAs (snoRNAs), small nuclear RNA (snRNAs) and tRNA-derived fragments (tRFs). Then, EVs from Malassezia sympodialis were shown to contain small RNAs similar to miRNAs [134], which was also the case in Pichia fermentans [20], while Histoplasma capsulatum EVs contained 25 nt long anti-sense RNAs [135]. Moreover, a few studies have indicated that the EV-RNA composition differs from the cellular composition [90][136]. The RNA content of EVs from P. brasiliensis and P. lutzii shared mRNA sequences related to protein modification and DNA metabolism, as well as small non-coding RNAs, some of which could potentially modulate the murine immune response [137]. A study on Cryptococcus gattii focused on the impact of the membrane architecture on the formation and content of EVs revealed that the RNA cargo was strongly altered in mutant cells [76]. Finally, a recent study comparing the EV cargos and functions of Candida albicans, a human-related yeast, and Candida auris, a new fungal pathogen that has emerged as a result of global warming [45], found significant differences in RNA content and speculated that these differences could explain the phenotypic changes induced by these EVs in human cells during immunological assays [62].

2.3.4. Other Molecules

Other than proteins, lipids and nucleic acids, the most abundant molecules found in fungal EVs are carbohydrates, pigments and prions. Glucuronoxylomannan (GXM) has been found in EVs from C. neoformans [18], and in C. gattii [76][138]. Moreover, other polysaccharides have been found in EVs from P. brasiliensis [61], P. lutzii [139] and C. albicans [21]. C. neoformans EVs were also found to carry melanin in an older study [140]. Prions have been found in EVs from S. cerevisiae, both in soluble and aggregated forms [24][141], and it has been hypothesised that they can play a role in vertical and horizontal transmission [142].

This entry is adapted from the peer-reviewed paper 10.3390/biom13101487

References

  1. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228.
  2. Mateescu, B.; Kowal, E.J.K.; Van Balkom, B.W.M.; Bartel, S.; Bhattacharyya, S.N.; Buzás, E.I.; Buck, A.H.; de Candia, P.; Chow, F.W.N.; Das, S.; et al. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA—An ISEV position paper. J. Extracell. Vesicles 2017, 6, 1286095.
  3. Gill, S.; Catchpole, R.; Forterre, P. Extracellular membrane vesicles in the three domains of life and beyond. FEMS Microbiol. Rev. 2019, 43, 273–303.
  4. Liebana-Jordan, M.; Brotons, B.; Falcon-Perez, J.M.; Gonzalez, E. Extracellular Vesicles in the Fungi Kingdom. Int. J. Mol. Sci. 2021, 22, 7221.
  5. Ayala-Mar, S.; Donoso-Quezada, J.; Gallo-Villanueva, R.C.; Perez-Gonzalez, V.H.; González-Valdez, J. Recent advances and challenges in the recovery and purification of cellular exosomes. Electrophoresis 2019, 40, 3036–3049.
  6. Hartjes, T.; Mytnyk, S.; Jenster, G.W.; van Steijn, V.; van Royen, M. Extracellular Vesicle Quantification and Characterization: Common Methods and Emerging Approaches. Bioengineering 2019, 6, 7.
  7. Doyle, L.; Wang, M. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727.
  8. Szatanek, R.; Baj-Krzyworzeka, M.; Zimoch, J.; Lekka, M.; Siedlar, M.; Baran, J. The Methods of Choice for Extracellular Vesicles (EVs) Characterization. Int. J. Mol. Sci. 2017, 18, 1153.
  9. Coumans, F.A.W.; Brisson, A.R.; Buzas, E.I.; Dignat-George, F.; Drees, E.E.E.; El-Andaloussi, S.; Emanueli, C.; Gasecka, A.; Hendrix, A.; Hill, A.F.; et al. Methodological Guidelines to Study Extracellular Vesicles. Circ. Res. 2017, 120, 1632–1648.
  10. Rodrigues, M.L.; Oliveira, D.L.; Vargas, G.; Girard-Dias, W.; Franzen, A.J.; Frasés, S.; Miranda, K.; Nimrichter, L. Analysis of Yeast Extracellular Vesicles. In Unconventional Protein Secretion; Pompa, A., De Marchis, F., Eds.; Springer: New York, NY, USA, 2016; pp. 175–190.
  11. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066.
  12. Chargaff, E.; West, R. The biological significance of the thromboplastic protein of blood. J. Biol. Chem. 1946, 166, 189–197.
  13. Wolf, P. The Nature and Significance of Platelet Products in Human Plasma. Br. J. Haematol. 1967, 13, 269–288.
  14. Wiklander, O.P.B.; Brennan, M.Á.; Lötvall, J.; Breakefield, X.O.; EL Andaloussi, S. Advances in therapeutic applications of extracellular vesicles. Sci. Transl. Med. 2019, 11, eaav8521.
  15. Agrahari, V.; Agrahari, V.; Burnouf, P.-A.; Chew, C.H.; Burnouf, T. Extracellular Microvesicles as New Industrial Therapeutic Frontiers. Trends Biotechnol. 2019, 37, 707–729.
  16. Elsharkasy, O.M.; Nordin, J.Z.; Hagey, D.W.; De Jong, O.G.; Schiffelers, R.M.; Andaloussi, S.E.; Vader, P. Extracellular vesicles as drug delivery systems: Why and how? Adv. Drug Deliv. Rev. 2020, 159, 332–343.
  17. Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell. Mol. Neurobiol. 2016, 36, 301–312.
  18. Rodrigues, M.L.; Nimrichter, L.; Oliveira, D.L.; Frases, S.; Miranda, K.; Zaragoza, O.; Alvarez, M.; Nakouzi, A.; Feldmesser, M.; Casadevall, A. Vesicular Polysaccharide Export in Cryptococcus neoformans Is a Eukaryotic Solution to the Problem of Fungal Trans-Cell Wall Transport. Eukaryot. Cell 2007, 6, 48–59.
  19. Rizzo, J.; Rodrigues, M.L.; Janbon, G. Extracellular Vesicles in Fungi: Past, Present, and Future Perspectives. Front. Cell. Infect. Microbiol. 2020, 10, 346.
  20. Leone, F.; Bellani, L.; Mucciflora, S.; Giorgetti, L.; Bongioanni, P.; Simili, M.; Maserti, B.; Del Carratore, R. Analysis of extracellular vesicles produced in the biofilm by the dimorphic yeast Pichia fermentans. J. Cell. Physiol. 2017, 233, 2759–2767.
  21. Zarnowski, R.; Sanchez, H.; Covelli, A.S.; Dominguez, E.; Jaromin, A.; Bernhardt, J.; Mitchell, K.F.; Heiss, C.; Azadi, P.; Mitchell, A.; et al. Candida albicans biofilm–induced vesicles confer drug resistance through matrix biogenesis. PLoS Biol. 2018, 16, e2006872.
  22. Voelz, K.; Johnston, S.A.; Smith, L.M.; Hall, R.A.; Idnurm, A.; May, R.C. ‘Division of labour’ in response to host oxidative burst drives a fatal Cryptococcus gattii outbreak. Nat. Commun. 2014, 5, 5194.
  23. Zhao, K.; Bleackley, M.; Chisanga, D.; Gangoda, L.; Fonseka, P.; Liem, M.; Kalra, H.; Al Saffar, H.; Keerthikumar, S.; Ang, C.-S.; et al. Extracellular vesicles secreted by Saccharomyces cerevisiae are involved in cell wall remodelling. Commun. Biol. 2019, 2, 305.
  24. Kabani, M.; Melki, R. Sup35p in Its Soluble and Prion States Is Packaged inside Extracellular Vesicles. mBio 2015, 6, e01017-15.
  25. Cai, Q.; He, B.; Weiberg, A.; Buck, A.H.; Jin, H. Small RNAs and extracellular vesicles: New mechanisms of cross-species communication and innovative tools for disease control. PLoS Pathog. 2019, 15, e1008090.
  26. Freitas, M.S.; Bonato, V.L.D.; Pessoni, A.M.; Rodrigues, M.L.; Casadevall, A.; Almeida, F. Fungal Extracellular Vesicles as Potential Targets for Immune Interventions. mSphere 2019, 4, e00747-19.
  27. Bielska, E.; May, R.C. Extracellular vesicles of human pathogenic fungi. Curr. Opin. Microbiol. 2019, 52, 90–99.
  28. Marina, C.L.; Bürgel, P.H.; Agostinho, D.P.; Zamith-Miranda, D.; Las-Casas, L.O.; Tavares, A.H.; Nosanchuk, J.D.; Bocca, A.L. Nutritional Conditions Modulate C. neoformans Extracellular Vesicles’ Capacity to Elicit Host Immune Response. Microorganisms 2020, 8, 1815.
  29. Oliveira, D.L.; Freire-De-Lima, C.G.; Nosanchuk, J.D.; Casadevall, A.; Rodrigues, M.L.; Nimrichter, L. Extracellular Vesicles from Cryptococcus neoformans Modulate Macrophage Functions. Infect. Immun. 2010, 78, 1601–1609.
  30. Da Silva, T.A.; Roque-Barreira, M.C.; Casadevall, A.; Almeida, F. Extracellular vesicles from Paracoccidioides brasiliensis induced M1 polarization in vitro. Sci. Rep. 2016, 6, 35867.
  31. Bitencourt, T.A.; Rezende, C.P.; Quaresemin, N.R.; Moreno, P.; Hatanaka, O.; Rossi, A.; Martinez-Rossi, N.M.; Almeida, F. Extracellular Vesicles from the Dermatophyte Trichophyton interdigitale Modulate Macrophage and Keratinocyte Functions. Front. Immunol. 2018, 9, 2343.
  32. Ikeda, M.A.K.; De Almeida, J.R.F.; Jannuzzi, G.P.; Cronemberger-Andrade, A.; Torrecilhas, A.C.T.; Moretti, N.S.; da Cunha, J.P.C.; De Almeida, S.R.; Ferreira, K.S. Extracellular Vesicles from Sporothrix brasiliensis Are an Important Virulence Factor That Induce an Increase in Fungal Burden in Experimental Sporotrichosis. Front. Microbiol. 2018, 9, 2286.
  33. Brauer, V.S.; Pessoni, A.M.; Bitencourt, T.A.; de Paula, R.G.; de Oliveira Rocha, L.; Goldman, G.H.; Almeida, F. Extracellular Vesicles from Aspergillus flavus Induce M1 Polarization In Vitro. mSphere 2020, 5, e00190-20.
  34. Kaparakis-Liaskos, M.; Ferrero, R.L. Immune modulation by bacterial outer membrane vesicles. Nat. Rev. Immunol. 2015, 15, 375–387.
  35. Macia, L.; Nanan, R.; Hosseini-Beheshti, E.; Grau, G.E. Host- and Microbiota-Derived Extracellular Vesicles, Immune Function, and Disease Development. Int. J. Mol. Sci. 2019, 21, 107.
  36. Sartorio, M.G.; Pardue, E.J.; Feldman, M.F.; Haurat, M.F. Bacterial Outer Membrane Vesicles: From Discovery to Applications. Annu. Rev. Microbiol. 2021, 75, 609–630.
  37. Jan, A.T. Outer Membrane Vesicles (OMVs) of Gram-negative Bacteria: A Perspective Update. Front. Microbiol. 2017, 8, 1053.
  38. Liu, Y.; Defourny, K.A.Y.; Smid, E.J.; Abee, T. Gram-Positive Bacterial Extracellular Vesicles and Their Impact on Health and Disease. Front. Microbiol. 2018, 9, 1502.
  39. 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.
  40. Brown, L.; Wolf, J.M.; Prados-Rosales, R.; Casadevall, A. Through the wall: Extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi. Nat. Rev. Microbiol. 2015, 13, 620–630.
  41. Coelho, C.; Casadevall, A. Answers to naysayers regarding microbial extracellular vesicles. Biochem. Soc. Trans. 2019, 47, 1005–1012.
  42. Honorato, L.; Bonilla, J.J.A.; Piffer, A.C.; Nimrichter, L. Fungal Extracellular Vesicles as a Potential Strategy for Vaccine Development. In Fungal Extracellular Vesicles; Rodrigues, M., Janbon, G., Eds.; Springer International Publishing: Cham, Switzerland, 2021; pp. 121–138.
  43. Köhler, J.R.; Hube, B.; Puccia, R.; Casadevall, A.; Perfect, J.R. Fungi that Infect Humans. Microbiol. Spectr. 2017, 5, 813–843.
  44. Du, H.; Bing, J.; Hu, T.; Ennis, C.L.; Nobile, C.J.; Huang, G. Candida auris: Epidemiology, biology, antifungal resistance, and virulence. PLoS Pathog. 2020, 16, e1008921.
  45. Casadevall, A.; Kontoyiannis, D.P.; Robert, V. On the Emergence of Candida auris: Climate Change, Azoles, Swamps, and Birds. mBio 2019, 10, e01397-19.
  46. Gremião, I.D.F.; Miranda, L.H.M.; Reis, E.G.; Rodrigues, A.M.; Pereira, S.A. Zoonotic Epidemic of Sporotrichosis: Cat to Human Transmission. PLoS Pathog. 2017, 13, e1006077.
  47. Siscar-Lewin, S.; Hube, B.; Brunke, S. Emergence and evolution of virulence in human pathogenic fungi. Trends Microbiol. 2022, 30, 693–704.
  48. Bongomin, F.; Gago, S.; Oladele, R.O.; Denning, D.W. Global and Multi-National Prevalence of Fungal Diseases—Estimate Precision. J. Fungi 2017, 3, 57.
  49. Fisher, M.C.; Hawkins, N.J.; Sanglard, D.; Gurr, S.J. Worldwide emergence of resistance to antifungal drugs challenges human health and food security. Science 2018, 360, 739–742.
  50. Farmakiotis, D.; Kontoyiannis, D.P. Epidemiology of antifungal resistance in human pathogenic yeasts: Current viewpoint and practical recommendations for management. Int. J. Antimicrob. Agents 2017, 50, 318–324.
  51. Robbins, N.; Caplan, T.; Cowen, L.E. Molecular Evolution of Antifungal Drug Resistance. Annu. Rev. Microbiol. 2017, 71, 753–775.
  52. Nimrichter, L.; de Souza, M.M.; Del Poeta, M.; Nosanchuk, J.D.; Joffe, L.; Tavares, P.d.M.; Rodrigues, M.L. Extracellular Vesicle-Associated Transitory Cell Wall Components and Their Impact on the Interaction of Fungi with Host Cells. Front. Microbiol. 2016, 7, 1034.
  53. Rodrigues, M.L.; Nakayasu, E.S.; Almeida, I.C.; Nimrichter, L. The impact of proteomics on the understanding of functions and biogenesis of fungal extracellular vesicles. J. Proteom. 2014, 97, 177–186.
  54. Chigaleĭchik, A.G.; Belova, L.A.; Grishchenko, V.M.; Rylkin, S.S. Several properties of the extracellular vesicles of Candida tropicalis yeasts grown on n-alkanes. Mikrobiologiia 1977, 46, 467–471.
  55. Gibson, R.K.; Peberdy, J.F. Fine Structure of Protoplasts of Aspergillus nidulans. J. Gen. Microbiol. 1972, 72, 529–538.
  56. Takeo, K.; Uesaka, I.; Uehira, K.; Nishiura, M. Fine Structure of Cryptococcus neoformans Grown In Vitro as Observed by Freeze-Etching. J. Bacteriol. 1973, 113, 1442–1448.
  57. Anderson, J.; Mihalik, R.; Soll, D.R. Ultrastructure and antigenicity of the unique cell wall pimple of the Candida opaque phenotype. J. Bacteriol. 1990, 172, 224–235.
  58. Rodrigues, M.; Janbon, G. Fungal Extracellular Vesicles: Biological Roles; Springer International Publishing: Cham, Switzerland, 2021.
  59. Rodrigues, M.L.; Travassos, L.R.; Miranda, K.R.; Franzen, A.J.; Rozental, S.; de Souza, W.; Alviano, C.S.; Barreto-Bergter, E. Human Antibodies against a Purified Glucosylceramide from Cryptococcus neoformans Inhibit Cell Budding and Fungal Growth. Infect. Immun. 2000, 68, 7049–7060.
  60. Rodrigues, M.L.; Nakayasu, E.S.; Oliveira, D.L.; Nimrichter, L.; Nosanchuk, J.D.; Almeida, I.C.; Casadevall, A. Extracellular Vesicles Produced by Cryptococcus neoformans Contain Protein Components Associated with Virulence. Eukaryot. Cell 2008, 7, 58–67.
  61. Vallejo, M.C.; Matsuo, A.L.; Ganiko, L.; Medeiros, L.C.S.; Miranda, K.; Silva, L.S.; Freymüller-Haapalainen, E.; Sinigaglia-Coimbra, R.; Almeida, I.C.; Puccia, R. The Pathogenic Fungus Paracoccidioides brasiliensis Exports Extracellular Vesicles Containing Highly Immunogenic α-Galactosyl Epitopes. Eukaryot. Cell 2011, 10, 343–351.
  62. Zamith-Miranda, D.; Heyman, H.M.; Couvillion, S.P.; Cordero, R.J.B.; Rodrigues, M.L.; Nimrichter, L.; Casadevall, A.; Amatuzzi, R.F.; Alves, L.R.; Nakayasu, E.S.; et al. Comparative Molecular and Immunoregulatory Analysis of Extracellular Vesicles from Candida albicans and Candida auris. mSystems 2021, 6, e0082221.
  63. Chan, W.; Chow, F.W.-N.; Tsang, C.-C.; Liu, X.; Yao, W.; Chan, T.T.-Y.; Siu, G.K.-H.; Ho, A.Y.-M.; Luk, K.S.; Lau, S.K.-P.; et al. Induction of amphotericin B resistance in susceptible Candida auris by extracellular vesicles. Emerg. Microbes Infect. 2022, 11, 1900–1909.
  64. Amatuzzi, R.F.; Zamith-Miranda, D.; da Rocha, I.F.M.; Lucena, A.C.R.; de Toledo Martins, S.; Streit, R.; Staats, C.C.; Trentin, G.; Almeida, F.; Rodrigues, M.L.; et al. Caspofungin Affects Extracellular Vesicle Production and Cargo in Candida auris. J. Fungi 2022, 8, 990.
  65. Bleackley, M.R.; Samuel, M.; Garcia-Ceron, D.; McKenna, J.A.; Lowe, R.G.T.; Pathan, M.; Zhao, K.; Ang, C.-S.; Mathivanan, S.; Anderson, M.A. Extracellular Vesicles from the Cotton Pathogen Fusarium oxysporum f. sp. vasinfectum Induce a Phytotoxic Response in Plants. Front. Plant Sci. 2020, 10, 1610.
  66. Costa, J.H.; Bazioli, J.M.; Barbosa, L.D.; dos Santos Júnior, P.L.T.; Reis, F.C.G.; Klimeck, T.; Crnkovic, C.M.; Berlinck, R.G.S.; Sussulini, A.; Rodrigues, M.L.; et al. Phytotoxic Tryptoquialanines Produced In Vivo by Penicillium digitatum Are Exported in Extracellular Vesicles. mBio 2021, 12, e03393-20.
  67. Rizzo, J.; Albuquerque, P.C.; Wolf, J.M.; Nascimento, R.; Pereira, M.D.; Nosanchuk, J.D.; Rodrigues, M.L. Analysis of multiple components involved in the interaction between Cryptococcus neoformans and Acanthamoeba castellanii. Fungal Biol. 2017, 121, 602–614.
  68. Rizzo, J.; Chaze, T.; Miranda, K.; Roberson, R.W.; Gorgette, O.; Nimrichter, L.; Matondo, M.; Latgé, J.-P.; Beauvais, A.; Rodrigues, M.L. Characterization of Extracellular Vesicles Produced by Aspergillus fumigatus Protoplasts. mSphere 2020, 5, e00476-20.
  69. Albuquerque, P.C.; Nakayasu, E.S.; Rodrigues, M.L.; Frases, S.; Casadevall, A.; Zancope-Oliveira, R.M.; Almeida, I.C.; Nosanchuk, J.D. Vesicular transport in Histoplasma capsulatum: An effective mechanism for trans-cell wall transfer of proteins and lipids in ascomycetes. Cell. Microbiol. 2008, 10, 1695–1710.
  70. Bitencourt, T.A.; Hatanaka, O.; Pessoni, A.M.; Freitas, M.S.; Trentin, G.; Santos, P.; Rossi, A.; Martinez-Rossi, N.M.; Alves, L.L.; Casadevall, A.; et al. Fungal Extracellular Vesicles Are Involved in Intraspecies Intracellular Communication. mBio 2022, 13, e0327221.
  71. Giardina, B.J.; Stein, K.; Chiang, H.-L. The endocytosis gene END3 is essential for the glucose-induced rapid decline of small vesicles in the extracellular fraction in Saccharomyces cerevisiae. J. Extracell. Vesicles 2014, 3, 23497.
  72. Stein, K.; Chiang, H.-L. Exocytosis and Endocytosis of Small Vesicles across the Plasma Membrane in Saccharomyces cerevisiae. Membranes 2014, 4, 608–629.
  73. Stein, K.; Winters, C.; Chiang, H.-L. Vps15p regulates the distribution of cup-shaped organelles containing the major eisosome protein Pil1p to the extracellular fraction required for endocytosis of extracellular vesicles carrying metabolic enzymes: Roles of Vps15p and Pil1p in vesicle endocytosis. Biol. Cell 2017, 109, 190–209.
  74. Kenno, S.; Speth, C.; Rambach, G.; Binder, U.; Chatterjee, S.; Caramalho, R.; Haas, H.; Lass-Flörl, C.; Shaughnessy, J.; Ram, S.; et al. Candida albicans Factor H Binding Molecule Hgt1p–A Low Glucose-Induced Transmembrane Protein Is Trafficked to the Cell Wall and Impairs Phagocytosis and Killing by Human Neutrophils. Front. Microbiol. 2019, 9, 3319.
  75. Dawson, C.S.; Garcia-Ceron, D.; Rajapaksha, H.; Faou, P.; Bleackley, M.R.; Anderson, M.A. Protein markers for Candida albicans EVs include claudin-like Sur7 family proteins. J. Extracell. Vesicles 2020, 9, 1750810.
  76. Reis, F.C.G.; Borges, B.S.; Jozefowicz, L.J.; Sena, B.A.G.; Garcia, A.W.A.; Medeiros, L.C.; Martins, S.T.; Honorato, L.; Schrank, A.; Vainstein, M.H.; et al. A Novel Protocol for the Isolation of Fungal Extracellular Vesicles Reveals the Participation of a Putative Scramblase in Polysaccharide Export and Capsule Construction in Cryptococcus gattii. mSphere 2019, 4, e00080-19.
  77. Cleare, L.G.; Zamith, D.; Heyman, H.M.; Couvillion, S.P.; Nimrichter, L.; Rodrigues, M.L.; Nakayasu, E.S.; Nosanchuk, J.D. Media matters! Alterations in the loading and release of Histoplasma capsulatum extracellular vesicles in response to different nutritional milieus. Cell. Microbiol. 2020, 22, e13217.
  78. De Oliveira, H.C.; Kato, A.F.; Sena, B.A.G.; Duarte, I.; Jozefowicz, L.J.; Castelli, R.F.; Kuczera, D.; Reis, F.C.G.; Alves, L.R.; Rodrigues, M.L. Biogenesis of Fungal Extracellular Vesicles: What Do We Know? In Fungal Extracellular Vesicles; Rodrigues, M., Janbon, G., Eds.; Springer International Publishing: Cham, Switzerland, 2021; pp. 1–11.
  79. Oliveira, D.L.; Nakayasu, E.S.; Joffe, L.S.; Guimarães, A.J.; Sobreira, T.J.P.; Nosanchuk, J.D.; Cordero, R.J.B.; Frases, S.; Casadevall, A.; Almeida, I.C.; et al. Biogenesis of extracellular vesicles in yeast: Many questions with few answers. Commun. Integr. Biol. 2010, 3, 533–535.
  80. Rodrigues, M.L.; Nosanchuk, J.D.; Schrank, A.; Vainstein, M.H.; Casadevall, A.; Nimrichter, L.; Lai, R.C.; Chen, T.S.; Lim, S.K.; Hodge, T.W.; et al. Vesicular transport systems in fungi. Future Microbiol. 2011, 6, 1371–1381.
  81. Oliveira, D.L.; Rizzo, J.; Joffe, L.S.; Godinho, R.M.C.; Rodrigues, M.L. Where Do They Come from and Where Do They Go: Candidates for Regulating Extracellular Vesicle Formation in Fungi. Int. J. Mol. Sci. 2013, 14, 9581–9603.
  82. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383.
  83. Maas, S.L.N.; Breakefield, X.O.; Weaver, A.M. Extracellular Vesicles: Unique Intercellular Delivery Vehicles. Trends Cell Biol. 2017, 27, 172–188.
  84. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289.
  85. Latifkar, A.; Hur, Y.H.; Sanchez, J.C.; Cerione, R.A.; Antonyak, M.A. New insights into extracellular vesicle biogenesis and function. J. Cell Sci. 2019, 132, jcs222406.
  86. Novick, P.; Field, C.; Schekman, R. Identification of 23 complementation groups required for post-translational events in the yeast secretory pathway. Cell 1980, 21, 205–215.
  87. Schekman, R. SEC mutants and the secretory apparatus. Nat. Med. 2002, 8, 1055–1058.
  88. Oliveira, D.L.; Nakayasu, E.S.; Joffe, L.S.; Guimarães, A.J.; Sobreira, T.J.P.; Nosanchuk, J.D.; Cordero, R.J.B.; Frases, S.; Casadevall, A.; Almeida, I.C.; et al. Characterization of Yeast Extracellular Vesicles: Evidence for the Participation of Different Pathways of Cellular Traffic in Vesicle Biogenesis. PLoS ONE 2010, 5, e11113.
  89. Panepinto, J.; Komperda, K.; Frases, S.; Park, Y.-D.; Djordjevic, J.T.; Casadevall, A.; Williamson, P.R. Sec6-dependent sorting of fungal extracellular exosomes and laccase of Cryptococcus neoformans. Mol. Microbiol. 2009, 71, 1165–1176.
  90. Da Silva, R.P.; Martins, S.D.T.; Rizzo, J.; Dos Reis, F.C.G.; Joffe, L.S.; Vainstein, M.; Kmetzsch, L.; Oliveira, D.L.; Puccia, R.; Goldenberg, S.; et al. Golgi Reassembly and Stacking Protein (GRASP) Participates in Vesicle-Mediated RNA Export in Cryptococcus neoformans. Genes 2018, 9, 400.
  91. Piper, R.C.; Cooper, A.A.; Yang, H.; Stevens, T.H. VPS27 controls vacuolar and endocytic traffic through a prevacuolar compartment in Saccharomyces cerevisiae. J. Cell Biol. 1995, 131, 603–617.
  92. Hurley, J.H.; Emr, S.D. The Escrt Complexes: Structure and Mechanism of a Membrane-Trafficking Network. Annu. Rev. Biophys. Biomol. Struct. 2006, 35, 277–298.
  93. Iwaki, T.; Onishi, M.; Ikeuchi, M.; Kita, A.; Sugiura, R.; Giga-Hama, Y.; Fukui, Y.; Takegawa, K. Essential roles of class E Vps proteins for sorting into multivesicular bodies in Schizosaccharomyces pombe. Microbiology 2007, 153, 2753–2764.
  94. Chiaruttini, N.; Redondo-Morata, L.; Colom, A.; Humbert, F.; Lenz, M.; Scheuring, S.; Roux, A. Relaxation of Loaded ESCRT-III Spiral Springs Drives Membrane Deformation. Cell 2015, 163, 866–879.
  95. Hu, G.; Caza, M.; Cadieux, B.; Chan, V.; Liu, V.; Kronstad, J. Cryptococcus neoformans Requires the ESCRT Protein Vps23 for Iron Acquisition from Heme, for Capsule Formation, and for Virulence. Infect. Immun. 2013, 81, 292–302.
  96. Hu, G.; Caza, M.; Cadieux, B.; Bakkeren, E.; Do, E.; Jung, W.H.; Kronstad, J.W. The endosomal sorting complex required for transport machinery influences haem uptake and capsule elaboration in C ryptococcus neoformans: ESCRT machinery and virulence in Cryptococcus neoformans. Mol. Microbiol. 2015, 96, 973–992.
  97. Da Godinho, R.M.C.; Crestani, J.; Kmetzsch, L.; de Araujo, G.S.; Frases, S.; Staats, C.C.; Schrank, A.; Vainstein, M.H.; Rodrigues, M.L. The vacuolar-sorting protein Snf7 is required for export of virulence determinants in members of the Cryptococcus neoformans complex. Sci. Rep. 2014, 4, 6198.
  98. Park, Y.-D.; Chen, S.H.; Camacho, E.; Casadevall, A.; Williamson, P.R. Role of the ESCRT Pathway in Laccase Trafficking and Virulence of Cryptococcus neoformans. Infect. Immun. 2020, 88, e00954-19.
  99. Bruckmann, A.; Künkel, W.; Augsten, K.; Wetzker, R.; Eck, R. The deletion of CaVPS34 in the human pathogenic yeast Candida albicans causes defects in vesicle-mediated protein sorting and nuclear segregation. Yeast 2001, 18, 343–353.
  100. Rodrigues, M.L.; Franzen, A.J.; Nimrichter, L.; Miranda, K. Vesicular mechanisms of traffic of fungal molecules to the extracellular space. Curr. Opin. Microbiol. 2013, 16, 414–420.
  101. Rizzo, J.; Oliveira, D.L.; Joffe, L.S.; Hu, G.; Gazos-Lopes, F.; Fonseca, F.L.; Almeida, I.C.; Frases, S.; Kronstad, J.W.; Rodrigues, M.L. Role of the Apt1 Protein in Polysaccharide Secretion by Cryptococcus neoformans. Eukaryot. Cell 2014, 13, 715–726.
  102. Wolf, J.M.; Espadas, J.; Luque-Garcia, J.; Reynolds, T.; Casadevall, A. Lipid Biosynthetic Genes Affect Candida albicans Extracellular Vesicle Morphology, Cargo, and Immunostimulatory Properties. Eukaryot. Cell 2015, 14, 745–754.
  103. Rizzo, J.; Colombo, A.C.; Zamith-Miranda, D.; Silva, V.K.; Allegood, J.C.; Casadevall, A.; Del Poeta, M.; Nosanchuk, J.D.; Kronstad, J.W.; Rodrigues, M.L. The putative flippase Apt1 is required for intracellular membrane architecture and biosynthesis of polysaccharide and lipids in Cryptococcus neoformans. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2018, 1865, 532–541.
  104. Coronado, J.E.; Mneimneh, S.; Epstein, S.L.; Qiu, W.-G.; Lipke, P.N. Conserved Processes and Lineage-Specific Proteins in Fungal Cell Wall Evolution. Eukaryot. Cell 2007, 6, 2269–2277.
  105. Gow, N.A.R.; Latge, J.-P.; Munro, C.A. The Fungal Cell Wall: Structure, Biosynthesis, and Function. Microbiol. Spectr. 2017, 5, 28513415.
  106. Wolf, J.M.; Casadevall, A. Challenges posed by extracellular vesicles from eukaryotic microbes. Curr. Opin. Microbiol. 2014, 22, 73–78.
  107. De Souza Pereira, R.; Geibel, J. Direct observation of oxidative stress on the cell wall of Saccharomyces cerevisiae strains with atomic force microscopy. Mol. Cell. Biochem. 1999, 201, 17–24.
  108. Wolf, J.M.; Espadas-Moreno, J.; Luque-Garcia, J.L.; Casadevall, A. Interaction of Cryptococcus neoformans Extracellular Vesicles with the Cell Wall. Eukaryot. Cell 2014, 13, 1484–1493.
  109. Walker, L.; Sood, P.; Lenardon, M.D.; Milne, G.; Olson, J.; Jensen, G.; Wolf, J.; Casadevall, A.; Adler-Moore, J.; Gow, N.A.R. The Viscoelastic Properties of the Fungal Cell Wall Allow Traffic of AmBisome as Intact Liposome Vesicles. mBio 2018, 9, e02383-17.
  110. De Toledo Martins, S.; Szwarc, P.; Goldenberg, S.; Alves, L.R. Extracellular Vesicles in Fungi: Composition and Functions. In Fungal Physiology and Immunopathogenesis; Rodrigues, M.L., Ed.; Springer International Publishing: Cham, Switzerland, 2018; pp. 45–59.
  111. Vallejo, M.C.; Nakayasu, E.S.; Matsuo, A.L.; Sobreira, T.J.P.; Longo, L.V.G.; Ganiko, L.; Almeida, I.C.; Puccia, R. Vesicle and Vesicle-Free Extracellular Proteome of Paracoccidioides brasiliensis: Comparative Analysis with Other Pathogenic Fungi. J. Proteome Res. 2012, 11, 1676–1685.
  112. Johansson, H.J.; Vallhov, H.; Holm, T.; Gehrmann, U.; Andersson, A.; Johansson, C.; Blom, H.; Carroni, M.; Lehtiö, J.; Scheynius, A. Extracellular nanovesicles released from the commensal yeast Malassezia sympodialis are enriched in allergens and interact with cells in human skin. Sci. Rep. 2018, 8, 9182.
  113. Gil-Bona, A.; Llama-Palacios, A.; Parra, C.M.; Vivanco, F.; Nombela, C.; Monteoliva, L.; Gil, C. Proteomics Unravels Extracellular Vesicles as Carriers of Classical Cytoplasmic Proteins in Candida albicans. J. Proteome Res. 2015, 14, 142–153.
  114. Vargas, G.; Rocha, J.D.B.; Oliveira, D.L.; Albuquerque, P.C.; Frases, S.; Santos, S.S.; Nosanchuk, J.D.; Gomes, A.M.O.; Medeiros, L.C.A.S.; Miranda, K.; et al. Compositional and immunobiological analyses of extracellular vesicles released by Candida albicans: Extracellular vesicles from Candida albicans. Cell. Microbiol. 2014, 17, 389–407.
  115. Mencher, A.; Morales, P.; Valero, E.; Tronchoni, J.; Patil, K.R.; Gonzalez, R. Proteomic characterization of extracellular vesicles produced by several wine yeast species. Microb. Biotechnol. 2020, 13, 1581–1596.
  116. Morales, P.; Mencher, A.; Tronchoni, J.; Gonzalez, R. Fungal Extracellular Vesicles; Rodrigues, M., Janbon, G., Eds.; Springer International Publishing: Cham, Switzerland, 2021; pp. 161–170.
  117. Vallejo, M.C.; Nakayasu, E.S.; Longo, L.V.G.; Ganiko, L.; Lopes, F.G.; Matsuo, A.L.; Almeida, I.C.; Puccia, R. Lipidomic Analysis of Extracellular Vesicles from the Pathogenic Phase of Paracoccidioides brasiliensis. PLoS ONE 2012, 7, e39463.
  118. Rella, A.; Farnoud, A.M.; Del Poeta, M. Plasma membrane lipids and their role in fungal virulence. Prog. Lipid Res. 2016, 61, 63–72.
  119. Rodrigues, M.L.; Nimrichter, L.; Oliveira, D.L.; Nosanchuk, J.D.; Casadevall, A. Vesicular Trans-Cell Wall Transport in Fungi: A Mechanism for the Delivery of Virulence-Associated Macromolecules? Lipid Insights 2008, 2, LPI.S1000.
  120. Matos Baltazar, L.; Nakayasu, E.S.; Sobreira, T.J.P.; Choi, H.; Casadevall, A.; Nimrichter, L.; Nosanchuk, J.D. Antibody Binding Alters the Characteristics and Contents of Extracellular Vesicles Released by Histoplasma capsulatum. mSphere 2016, 1, e00085-15.
  121. Tsatsaronis, J.A.; Franch-Arroyo, S.; Resch, U.; Charpentier, E. Extracellular Vesicle RNA: A Universal Mediator of Microbial Communication? Trends Microbiol. 2018, 26, 401–410.
  122. Bitencourt, T.A.; Pessoni, A.M.; Oliveira, B.T.M.; Alves, L.R.; Almeida, F. The RNA Content of Fungal Extracellular Vesicles: At the “Cutting-Edge” of Pathophysiology Regulation. Cells 2022, 11, 2184.
  123. Munhoz da Rocha, I.F.; Amatuzzi, R.F.; Lucena, A.C.R.; Faoro, H.; Alves, L.R. Cross-Kingdom Extracellular Vesicles EV-RNA Communication as a Mechanism for Host–Pathogen Interaction. Front. Cell. Infect. Microbiol. 2020, 10, 593160.
  124. Holoch, D.; Moazed, D. RNA-mediated epigenetic regulation of gene expression. Nat. Rev. Genet. 2015, 16, 71–84.
  125. Weiberg, A.; Bellinger, M.; Jin, H. Conversations between kingdoms: Small RNAs. Curr. Opin. Biotechnol. 2015, 32, 207–215.
  126. Lee, H.-J. Microbe-Host Communication by Small RNAs in Extracellular Vesicles: Vehicles for Transkingdom RNA Transportation. Int. J. Mol. Sci. 2019, 20, 1487.
  127. Fang, Y.; Wang, Z.; Liu, X.; Tyler, B.M. Biogenesis and Biological Functions of Extracellular Vesicles in Cellular and Organismal Communication With Microbes. Front. Microbiol. 2022, 13, 817844.
  128. Kwon, S.; Tisserant, C.; Tulinski, M.; Weiberg, A.; Feldbrügge, M. Inside-out: From endosomes to extracellular vesicles in fungal RNA transport. Fungal Biol. Rev. 2020, 34, 89–99.
  129. Weiberg, A.; Wang, M.; Lin, F.-M.; Zhao, H.; Zhang, Z.; Kaloshian, I.; Huang, H.-D.; Jin, H. Fungal Small RNAs Suppress Plant Immunity by Hijacking Host RNA Interference Pathways. Science 2013, 342, 118–123.
  130. Wang, M.; Weiberg, A.; Lin, F.-M.; Thomma, B.P.H.J.; Huang, H.-D.; Jin, H. Bidirectional cross-kingdom RNAi and fungal uptake of external RNAs confer plant protection. Nat. Plants 2016, 2, 16151.
  131. Wang, B.; Sun, Y.; Song, N.; Zhao, M.; Liu, R.; Feng, H.; Wang, X.; Kang, Z. Puccinia striiformis f. sp. tritici mi croRNA-like RNA 1 (Pst-milR1), an important pathogenicity factor of Pst, impairs wheat resistance to Pst by suppressing the wheat pathogenesis-related 2 gene. New Phytol. 2017, 215, 338–350.
  132. Jian, J.; Liang, X. One Small RNA of Fusarium graminearum Targets and Silences CEBiP Gene in Common Wheat. Microorganisms 2019, 7, 425.
  133. Peres da Silva, R.; Puccia, R.; Rodrigues, M.L.; Oliveira, D.L.; Joffe, L.S.; César, G.V.; Nimrichter, L.; Goldenberg, S.; Alves, L.R. Extracellular vesicle-mediated export of fungal RNA. Sci. Rep. 2015, 5, 7763.
  134. 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.
  135. Alves, L.R.; da Silva, R.P.; Sanchez, D.A.; Zamith-Miranda, D.; Rodrigues, M.L.; Goldenberg, S.; Puccia, R.; Nosanchuk, J.D. Extracellular Vesicle-Mediated RNA Release in Histoplasma capsulatum. mSphere 2019, 4, e00176-19.
  136. Munhoz da Rocha, I.F.; Martins, S.T.; Amatuzzi, R.F.; Zamith-Miranda, D.; Nosanchuk, J.D.; Rodrigues, M.L.; Alves, L.R. Cellular and Extracellular Vesicle RNA Analysis in the Global Threat Fungus Candida auris. Microbiol. Spectr. 2021, 9, e0153821.
  137. Peres da Silva, R.; Longo, L.G.V.; da Cunha, J.P.C.; Sobreira, T.J.P.; Rodrigues, M.L.; Faoro, H.; Goldenberg, S.; Alves, L.R.; Puccia, R. Comparison of the RNA Content of Extracellular Vesicles Derived from Paracoccidioides brasiliensis and Paracoccidioides lutzii. Cells 2019, 8, 765.
  138. Bielska, E.; Sisquella, M.A.; Aldeieg, M.; Birch, C.; O’donoghue, E.J.; May, R.C. Pathogen-derived extracellular vesicles mediate virulence in the fatal human pathogen Cryptococcus gattii. Nat. Commun. 2018, 9, 1556.
  139. da Silva, R.P.; Heiss, C.; Black, I.; Azadi, P.; Gerlach, J.Q.; Travassos, L.R.; Joshi, L.; Kilcoyne, M.; Puccia, R. Extracellular vesicles from Paracoccidioides pathogenic species transport polysaccharide and expose ligands for DC-SIGN receptors. Sci. Rep. 2015, 5, 14213.
  140. Eisenman, H.C.; Mues, M.; Weber, S.E.; Frases, S.; Chaskes, S.; Gerfen, G.; Casadevall, A. Cryptococcus neoformans laccase catalyses melanin synthesis from both d- and l-DOPA. Microbiology 2007, 153, 3954–3962.
  141. Kabani, M.; Pilard, M.; Melki, R. Glucose availability dictates the export of the soluble and prion forms of Sup35p via periplasmic or extracellular vesicles. Mol. Microbiol. 2020, 114, 322–332.
  142. Kabani, M. Extracellular Vesicles-Encapsulated Yeast Prions and What They Can Tell Us about the Physical Nature of Propagons. Int. J. Mol. Sci. 2020, 22, 90.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations