Skip to main content
Advertisement
  • Loading metrics

Biogenesis of extracellular vesicles in protozoan parasites: The ESCRT complex in the trafficking fast lane?

Abstract

Extracellular vesicles (EVs) provide a central mechanism of cell–cell communication. While EVs are found in most organisms, their pathogenesis-promoting roles in parasites are of particular interest given the potential for medical insight and consequential therapeutic intervention. Yet, a key feature of EVs in human parasitic protozoa remains elusive: their mechanisms of biogenesis. Here, we survey the current knowledge on the biogenesis pathways of EVs secreted by the four main clades of human parasitic protozoa: apicomplexans, trypanosomatids, flagellates, and amoebae. In particular, we shine a light on findings pertaining to the Endosomal Sorting Complex Required for Transport (ESCRT) machinery, as in mammals it plays important roles in EV biogenesis. This review highlights the diversity in EV biogenesis in protozoa, as well as the related involvement of the ESCRT system in these unique organisms.

I. EV biogenesis in parasitic protozoa

Parasitic protozoa are responsible for a range of diseases in humans. Notable species include Plasmodium falciparum, responsible for the deadly malaria disease [1] (Apicomplexa phylum), Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp., responsible for African sleeping sickness, Chagas disease, and leishmaniasis, respectively [2,3], (Kinetoplastea phylum, syn. Kinetoplastida), Trichomonas vaginalis (Metamonada phylum), the most prevalent nonviral sexually transmitted human pathogen [4], and Acanthamoeba castellanii, a prevalent opportunistic disease [5] (Amoebozoa phylum). These single-celled pathogens’ capacity to cause disease in humans and other organisms is dependent on their abilities to invade tissues and cells, coordinate their actions, and manipulate host defenses [68]. Studies in recent years have implicated extracellular vesicles (EVs) as mediators of these parasitic activities.

EVs are cell-derived, membrane-enclosed vesicles that transport a variety of cargo components, such as proteins, nucleic acids, and metabolites [79]. This robust delivery system enables cell–cell communication by mediating diverse physiological and pathological processes in all kingdoms throughout evolution, from bacteria to humans [10]. EV biogenesis occurs either through the release of exosomes originated in intraluminal vesicles (ILVs) contained within multivesicular bodies (MVBs), upon the fusion of MVBs with a cell’s plasma membrane [11,12], or through direct plasma membrane budding of microvesicles (also called ectosomes) from the cell surface [9,13].

It has been demonstrated that parasitic pathogens, including P. falciparum, T. brucei, and Giardia intestinalis, produce EVs laden with proteins [6], coding and noncoding RNAs [1418], and/or DNA [1921] during infections. They do so either autonomously, by using their own machinery [2224], or possibly by exploiting their host’s system, just like viruses do [2527]. Despite the growing consensus as to the importance of EVs in protozoa pathogenesis, our understanding of the mechanisms of EV biogenesis in human parasitic protozoa is still in its infancy, with many questions remaining unanswered. With an eye to advancing this understudied aspect of parasite pathogenesis, in this review, we summarize the findings to date on the pathways involved in EV production in the major human parasitic protozoa.

We will mainly focus on the Endosomal Sorting Complex Required for Transport (ESCRT) system, a key player in the biogenesis of EVs [13]. The ESCRT system mediates several other fundamental cellular processes, including plasma membrane remodeling and maintenance during protein trafficking [28,29], cell organelle compartmentalization (e.g., lysosomes, the nucleus, autophagosomes) [30], sorting of ubiquitinated proteins into ILVs for lysosomal degradation [31], fission and repair of damaged cell membranes [32,33], cell cytokinesis [34,35], and viral egress [3638]. Composed of approximately 30 proteins, the mammalian ESCRT is a conserved machinery that is localized to endocytic compartments [13]. The ESCRT proteins assemble into five subcomplexes: ESCRT-0, -I, -II, -III, and the ESCRT machinery-associated proteins Alix/Bro1 and the AAA ATPase Vps4 [13].

To generate EVs, ESCRT’s subcomplexes perform a sequence of tasks that lead first to the production of ILVs and then to the generation of exo- or ectosomal EVs [39]. Briefly, ESCRT-0 sequesters ubiquitinated protein cargo, ESCRT-I, -II, and -III induce ILV budding, while the Vps4 enzymatic subcomplex regulates membrane scission [31,4042]. Each complex comprises specific subunits and domains (e.g., ubiquitin-binding), adaptors, and affiliated proteins (e.g., Tsg101 and Alix in certain exosomes) [43]. These subunits determine their respective unit’s particular roles in the process, the nature of the protein cargo loaded [44], and, eventually, the EV cargo’s destination [37].

Studies into the evolution of the ESCRT system identified numerous homologs of the ESCRT families I, II, III, and associated proteins in a wide variety of protozoa, including Plasmodium [22,45], Trypanosoma [23,45,46], Leishmania [45], Giardia [45], and Entamoeba [4751]. It was found that the ESCRT proteins are conserved, suggesting a potentially fully functional system in many of these organisms. This insight highlights the need for further studies concentrating on the specific functions and roles of the ESCRT proteins in parasitic protozoa.

II. EV functions and biogenesis in Apicomplexa: Plasmodium falciparum and Toxoplasma gondii

Consisting of some 6,000 species [52], apicomplexan parasites are among the most prevalent and morbidity-causing pathogens worldwide, responsible for severe diseases in millions of humans and animals each year [52]. Some of its most known genera that infect humans are Plasmodium (haemosporidans), Toxoplasma (eucoccidians), Babesia (piroplasms), and Cryptosporidium (cryptosporidians). Being intracellular obligate parasites, apicomplexans constantly face the hostile environment of the host, which poses unique challenges for signaling and communication both, among the parasite population and within the host [7,5355].

One of the methods by which apicomplexans manipulate host cells and evade the immune system is the secretion of EVs with different cargo components, which are eventually taken up by various target cells [7,56]. Apicomplexa-derived EVs are involved in a multitude of processes, including in transferring virulence factors [18,20,5759], easing parasite invasion into host cells [58], and modifying the host immune response [18,20,5962]. Nonetheless, EV biogenesis in apicomplexan parasites has been scarcely studied, and the ESCRT system has been implicated in EV biogenesis only in P. falciparum [22], the causative agent of the most deadly form of malaria disease [1].

Studies have identified a wide range of active cargo components, such as genomic DNA [19,20], RNA [18,57,59,61], host and parasitic proteins [58,63], glycans [64], and lipids [65,66], in P. falciparum-derived EVs. Moreover, the cargo composition in the EVs changes with the parasite’s developmental stage, suggesting active cargo sorting into the EVs [20]. P. falciparum-derived EVs have been shown to advance several pathogenesis-promoting processes in the human host. First, they allow the parasites to communicate among themselves via the exchange of active cargo, even when enclosed inside P. falciparum-infected human red blood cells (RBCs) [19,61]. In particular, it was demonstrated that P. falciparum EVs help promote this parasite’s sexual stage development (gametocytogenesis), an essential process for transmission [19,61,67]. Second, parasites use them to manipulate the host’s immune response [18,20,59,61]. Third, they aid in promoting vascular changes and endothelial cell activation [68] and permeability [57], factors that contribute to the establishment of cerebral malaria, the most severe neurological complication of P. falciparum malaria. Lastly, P. falciparum-derived EVs prime naïve RBCs for parasite invasion [58]. This feat is achieved by the transfer of assembled and functional 20S proteasome complexes [58], which, upon uptake by naïve human RBCs, alter their cytoskeletal integrity [58]. Similarly, Plasmodium vivax, the most widely distributed human malaria parasite, was also shown to produce EVs [69] and manipulate the host’s NF-κB signaling to promote cytoadherence [62].

Toxoplasma, the causative agent of toxoplasmosis [70,71], was also shown to secrete EVs. In particular, EVs secreted from T. gondii-infected human foreskin fibroblasts [72] or dendritic cells [73] exhibit a unique profile of protein and nucleic acids that starkly differs from that of EVs derived from uninfected cells [72,73]. T. gondii-derived EVs have been reported to modulate the murine macrophage immune response by the induction of IL-10, TNF-α, and iNOS, probably by delivering miRNA to these host cells [60,74]. In addition, the secretion of the inflammatory cytokines IL-12, IFN-γ, and TNF-α, mediated through the JNK pathway, was demonstrated in murine macrophages stimulated with T. gondii EVs [75]. Interestingly, BALB/c mice inoculated with T.gondii-derived EVs showed humoral and cellular immune responses as well as a prolonged survival time [74,76], protecting the mice against acute parasite infection. These results may suggest that EVs could serve as vaccine candidates against toxoplasmosis [76].

While accumulating evidence points to the contribution of parasite-derived EVs to malaria and toxoplasmosis pathogenesis, less is known about the underlying mechanisms of EV biogenesis in these organisms and in Apicomplexa in general. A study of the phylogenetics of the ESCRT system revealed a lack of the full ESCRT machinery in Plasmodium, particularly of the ESCRT-0, -I, and -II subcomplexes [45]. Yet, in silico predictions demonstrated that P. falciparum possesses at least two putative proteins from the ESCRT-III complex: Vps2 and Vps32/Snf7 [45,77]. Furthermore, an accessory protein to the ESCRT-III complex, the Vps4 ATPase homologue (PfVps4), was found in the cytoplasm of the parasite during its trophozoite blood stage, detected by anti-Vps4 antibodies [78]. When transfected into COS cells, this homologue retained its function in MVB formation, hinting toward the existence of functional ESCRT machinery that mediates the production of MVBs in P. falciparum [78].

A key study improved our understanding of the mechanisms of P. falciparum EV release by demonstrating the activation of a functional ESCRT-III machinery by an “alternative recruitment pathway,” seemingly independent from other ESCRT subcomplexes [22]. Namely, it was found that two ESCRT-III proteins encoded in the P. falciparum genome [45], PfVps32 and PfVps60, as well as PfBro1 (a parasitic homologue of the human Alix protein) were all present in EVs derived from P. falciparum-infected human RBCs but not in EVs derived from naïve RBCs [22]. Although all three proteins localized to the cytoplasm of the parasite, PfVps32 and PfBro1 were also exported to the RBC cytoplasm, where it was suggested that they participate in microvesicle (ectosome) biogenesis and shedding [22] (Fig 1, Apicomplexa). In addition, PfVps60 KO parasites secreted less EVs compared to their WT counterparts, lending further support to PfVps60’s participation in EV biogenesis during P. falciparum infection [22]. The same study used the giant unilamellar vesicle (GUV) membrane model and P. falciparum-ESCRT-III-purified recombinant proteins to further demonstrate that the PfBro1 protein can trigger the formation of buds by recruiting PfVps32 and PfVps60 proteins to the GUV membrane and activating them [22], as occurs with other ESCRT-III homologues [79,80] during MVB generation (Fig 1, Apicomplexa). Thus, using the purified proteins allowed the recreation of the two mechanisms of EV production in P. falciparum: MVB generation, as well as membrane shedding in GUVs that mimic the composition of the erythrocyte plasma membrane.

thumbnail
Fig 1. Overview of EV biogenesis and ESCRT protein involvement in human parasitic protozoa.

The schematic diagram represents the current knowledge about EV biogenesis mechanisms in the most studied parasitic protozoa, organized by taxonomic and clinical proximity: apicomplexans, kinetoplastids, metamonads, and amoebae. The EVs’ colors reflect their biogenetic origins: MVB-derived exosomes (MVB-EV, green), plasma membrane–derived ectosomes (PM-EV, blue), nanotube-derived EVs (Nanotube-EV, purple), reservosome-derived EVs (Reservosome-EV, red), Maurer’s cleft-derived EVs (MC-EV, brown), or EVs of unknown biogenetic origin (unknown-EV, yellow). The ? symbol indicates those parasites in which EV secretion is not yet proven, but suggested (Naegleria fowleri), or those demonstrated to secrete EVs but their biogenetic origin remains uncertain (T. gondii and Acanthamoeba). The specific ESCRT proteins that have been experimentally demonstrated to participate in each biogenetic route are indicated. Green letters represent involvement in the MVB-EV pathway, and blue letters represent involvement in the PM-EV pathway, two pathways with proven ESCRT involvement. PfPTP2, a non-ESCRT protein found to be related to EV secretion in P. falciparum is also presented in brown letters. Although N. fowleri is not taxonomically related to the other amoebae belonging to Amoebozoa, it was grouped with the others for the purpose of simplicity. EV, extracellular vesicle; FP, flagellar pocket; MC, Maurer’s clefts; MVB, multivesicular body; PM, plasma membrane. Created with BioRender.com and licensed for publication (agreement number: GK24X52HZC).

https://doi.org/10.1371/journal.ppat.1011140.g001

Maurer’s clefts, an elusive membranal system that appears in P. falciparum-infected RBCs, are also thought to be involved in active EV secretion. This system is critical for the parasite’s ability to actively export PfEMP1, the major virulence factor of malaria, and other parasitic proteins to the RBC membrane [81]. It has been shown that the deletion of PfPTP2, a protein localized to Maurer’s clefts that is involved in transporting PfEMP1 to the RBC membrane [82], leads to a significant reduction in EV secretion from infected RBCs [19] (Fig 1, Apicomplexa).

T. gondii, too, employs the ESCRT machinery to promote its pathogenesis. It was found that during infection, this parasite recruits three components of the host’s ESCRT machinery [83] to the protein-laden membrane (PVM) that envelopes its parasitophorous vacuole [27]. Two host proteins related to the ESCRT-III machinery, CHMP4B and its regulator, CC2D1A, were highly enriched in the PVM [83]. These findings suggest that host ESCRT machinery promotes or maintains Toxoplasma’s nanotubular intravacuolar network [83]. Another study showed that T. gondii rhoptry neck proteins cooperate to actively recruit different host proteins during invasion, including ESCRT-I components Alix and TSG101 [84]. Parasite mutants that cannot recruit these host proteins showed insufficient invasion into host cells and reduced virulence in mice [84]. In addition, the T. gondii effector protein TgGRA14 was shown to recruit the host ESCRT machinery to the PV membrane of the parasite and use it for vesicular trafficking and uptake of host cytosolic proteins [85]. Thus, it is evident that T. gondii is capable of exploiting host ESCRT machinery for resource procurement and vesicular trafficking. These results may also suggest that the parasite uses the host ESCRT machinery for EV biogenesis (Fig 1, Apicomplexa), in line with reports on other intracellular parasites [27]. Further research is needed, however, to uncover the exact mechanism of EV biogenesis in T. gondii and the players involved.

III. EV functions and biogenesis in Kinetoplastida: Trypanosoma brucei, Trypanosoma cruzi, and Leishmania

Kinetoplastid parasites belonging to the Trypanosomatidae family are responsible for a wide variety of human diseases, including leishmaniasis (caused by numerous Leishmania species), American Trypanosomiasis or Chagas disease (caused by T. cruzi), and human African Trypanosomiasis or “sleeping sickness” (caused by T. brucei) [86]. These three parasitic diseases are considered neglected tropical diseases by the World Health Organization [87] and mainly affect developing countries with impoverished populations [87].

EV secretion is one of the strategies kinetoplastid parasites use to sustain their complex life cycles in the human host [88,89]. These EVs aid trypanosomatids invade into host cells [15,90], communicate with other cells [15,23], advance the course of the disease [9193], regulate the immune response [92,94,95], and survive in their vectors and hosts [88,89,96]. EVs even possibly play a central role in protein secretion [97100], as proteins secreted by trypanosomatids, in general, lack a signal peptide; for example, in Leishmania, only 5% to 9% of secreted proteins contain a signal peptide [100102]. Secretome analyses in Trypanosoma, L. major, and L. donovani revealed that most of the protein secretome in these organisms is released via EVs [97100].

Most of the current knowledge on trypanosomatid EV biogenesis stems from T. brucei studies. T. brucei-derived EVs have been implicated in multiple biological functions relevant to pathogenesis, including antigen and virulence factor transference (mainly of the variant surface glycoproteins) [103], survival of the extracellular parasites in blood [98], host erythrocyte lysis [103], immune response manipulation [104], and central nervous system inflammation [105]. This wide range of cellular targets points towards different EV subpopulations with different possible biogenetic origins. One subpopulation of T. brucei EVs is generated in the flagellar pocket [98,103] (Fig 1, Kinetoplastida), a main cellular compartment unique to trypanosomatids with central functions in vesicular trafficking, endocytosis, and exocytosis [106]. This EV subgroup range in size from 50 to 100 nm and originate from nanotubes [103], which are highly dynamic filamentous membrane projections of up to 20 μm that bud from the flagellar pocket and vesiculate to form EVs [98,103]. These nanotube-derived EVs were first identified in blood-stage trypomastigotes [103] and later on also in vector-residing procyclic trypomastigotes [23].

T. brucei is also able to generate exosomes of 40 to 100 nm via MVBs when the RNA trans-splicing pathway is disrupted [23], as well as during heat shock [23]. Despite appearing to be secreted from the flagellar pocket compartment, these exosomes differ morphologically from previously characterized flagellar pocket nanotube-derived EVs [98,103]. MVB-derived EVs were shown to regulate social motility in T. brucei procyclic parasites, an important process during vector invasion and proliferation [23,107].

T. cruzi, too, employs EVs for a range of functions, including host cell invasion [15,90,108,109], pathogenesis and disease progression [91,95], vector colonization and metacyclogenesis [15,110], and immune system evasion and manipulation [111113]. T. cruzi-derived EVs are considered regulators of both the acute and chronic forms of Chagas disease [114], achieved through the modulation of inflammation and remote signaling between extracellular trypomastigotes and intracellular amastigotes in diverse host cell niches [114]. EVs have been successfully isolated from most stages of the T. cruzi life cycle [115], including the vector-invasive epimastigote [110,116], axenic amastigotes [117], trypomastigotes [90,91,116] (bloodstream, tissue culture-derived, and metacyclic), and T. cruzi-infected host cells [118], and their cargo has been compared across stages [116] and strains [111].

Proteomic analysis of the T. cruzi secretome revealed that epimastigotes and metacyclic trypomastigotes both secrete two subpopulations of EVs: larger ectosomes that bud directly from the cell membrane surrounding the flagellum and smaller exosomes, possibly MVB-derived [119], which are also released through the flagellar pocket [119] (Fig 1, Kinetoplastida). These data suggest that, similar to the case of T. brucei [23], there may be at least two EV biogenesis pathways in play in T. cruzi: plasma membrane–derived ectosomes and MVB-derived exosomes [119], both in the flagellar pocket.

Just like in T. brucei and T. cruzi, the EVs secreted by Leishmania play essential roles for its pathogenesis, including immune cell regulation [92,120,121], invasion and intracellular proliferation [122], cutaneous lesion formation [123], immune cell recruitment [124], and disease progression [125,126]. EVs have been isolated from most stages of the life cycle of the Leishmania parasite, including axenic amastigotes [16], intracellular amastigotes in infected macrophages [9294], procyclic and metacyclic promastigotes [120,123,127], indicating that both its intracellular and extracellular forms produce EVs. It was further determined that EVs that originate from amastigotes and promastigotes have different sizes [120]. The proteomic profile of procyclic and metacyclic promastigotes is also unique [127], suggesting these stages might have specific biogenetic pathways that generate EVs of diverse cargos and sizes.

Unlike Trypanosoma, the flagellar pocket in Leishmania has not been demonstrated to be directly involved in EV secretion, although the protein secretion endosomal systems are also located in the flagellar pocket [128,129]. Moreover, it was found that the LRV1 virus, which infects L. guyanensis, is released within the parasite’s EVs [130]. The virus also exploits the parasite’s biogenetic machinery in order to be loaded onto EVs [130]. LRV1 virions were enriched both in MVBs and the flagellar pocket of the parasite [130], suggesting that the flagellar pocket may be involved in EV biogenesis also in Leishmania [130] (Fig 1, Kinetoplastida). Whether the flagellar pocket and MVBs are responsible for EV release in conditions other than LRV1 infection and whether the ESCRT system is associated with these organelles in the context of EV biogenesis remain elusive questions.

Little is currently known about the EV biogenesis pathways in kinetoplastid parasites [88,126]. Yet, at least in the case of in T. cruzi, multiple stress conditions, such as nutrient starvation [15], low temperature and pH [131], nitrosative stress [131], and challenges with host cells [95], trigger EV release. In Leishmania, changes in extracellular temperature and pH can trigger EV secretion too [100]. This points toward EV release as being a tightly regulated process in these parasites, with different biogenetic pathways possibly activated by different stressors.

A phylogenetic study of the ESCRT system identified several homologues of ESCRT-I, -II, and -III family members in T. brucei, L. major, and T. cruzi [45], indicating that this system is conserved in Kinetoplastida [45]. The finding of protein homologues of most of the ESCRT families in kinetoplastids suggests that the ESCRT complex is fully functional in these organisms and can be active in both extracellular and intracellular stages of infection, potentially freeing them from total dependence on the host-derived ESCRT complex [27]. Kinetoplastid ESCRT protein members were shown to be involved in canonical functions such as endocytosis [46], ubiquitin-tagged protein degradation [45,132], surface protein recycling, and vesicular traffic to the endosome [45,46,133], and were suggested to be involved also in EV biogenesis [23,88,126].

Four ESCRT proteins have been fully characterized in T. brucei (TbVps23, TbVps28, TbVps4, and TbVps24) and demonstrated to be important players in endocytosis of ubiquitinated proteins [45], lysosomal degradation, and recycling of the Invariant Surface Glycoprotein 65 (ISG65) [46,133], all of which occur within the flagellar pocket. A link between MVBs and the flagellar pocket in T. brucei arises from the finding that the flagellar pocket is involved in the rescue of variant and invariant surface glycoproteins internalized in MVBs [132,134] and targeted to the lysosome by recycling back to the membrane [132,134]. This evidence may suggest an intimate connection between ESCRT proteins in the T. brucei endosomal compartment, MVBs, and the flagellar pocket. Nevertheless, the ESCRT machinery has yet to be directly associated with flagellar pocket EV biogenesis.

Only the ESCRT-II component Vps36 has been shown to be critical for the secretion of MVB-derived EVs [23] in T. brucei. Specifically, silencing Vps36 hindered the production of EVs derived from MVBs but did not stop the secretion of nanotube-derived EVs [23]. Thus, the biogenesis of nanotube-derived EVs in the T. brucei flagellar pocket may be independent from the ESCRT complex, or at least not involve Vps36 [23], while MVB-derived EV biogenesis and secretion does rely on Vps36 [23] (Fig 1, Kinetoplastida). It further stands to reason that some of the yet understudied ESCRT candidates may be involved in flagellar pocket EV biogenesis through similar mechanisms. This initial evidence links ESCRT proteins to EV biogenesis in trypanosomatids and opens the door to future explorations of the specific roles and involvement of ESCRT proteins in T. brucei.

In T. cruzi, the EV biogenetic pathways remain almost completely unknown, and no ESCRT proteins have been characterized yet, despite their predicted existence [45]. Similarly, only a few studies have investigated the biogenesis of EVs in Leishmania. Analogues of the ESCRT complex families I, II, and III were found in Leishmania [45], but their roles in EV biogenesis remain unexplored. EV proteomic studies have found Rab GTPases [100], Alix [100], and other ESCRT homologues to be present within Leishmania EVs [99101,123]. TEM analysis has identified MVBs, suggested to be active in EV secretion, in Leishmania promastigotes within the vector sandfly [123]. Collectively, these data suggest that MVBs and the ESCRT system might be active in EV biogenesis, at least in the extracellular stages of this parasite. Interestingly, human ESCRT member proteins have been identified in EVs secreted by Leishmania-infected macrophages [92]. Thus, it has been suggested that intracellular Leishmania amastigotes may also be able to “hijack,” at least partially, the host cell’s ESCRT system and vesicle secretion mechanisms to release their own EVs [27,100]. Therefore, Leishmania could export virulence factors into the host’s cytosol or to the parasitophorous vacuole and “hijack” the existing EV protein sorting and secretion machinery of its host cells.

Importantly, both T. cruzi’s and T. brucei’s EV production has been related to small RNA pathways. For example, both tsRNA [15] and SL RNAs [23] are recruited to MVBs and endosomal compartments and sorted into EVs during stress. The sorting of specific tRNA- and rRNA-derived small RNA into EVs was also demonstrated in Leishmania [16]. In T. cruzi, nutrient deprivation causes tsRNA to be recruited to endosomal compartments identified as reservosomes [15] and then released within EVs of 20 to 200 nm in size [15]. Reservosomes may represent a distinct EV biogenetic pathway [15,135] (Fig 1, Kinetoplastida), as they are enriched in proteins that are also abundant in EVs, such as cruzipain and Rab homologues [89,136]. EVs released during nutritional stress of T. cruzi epimastigotes were shown to contain unique small RNAs derived from rRNA, tRNA, sno/snRNAs, and protein coding sequences [135]. Collectively, these data suggest that specialized EV-RNA sorting pathways may exist in these parasites as part of their EV biogenetic machinery. However, further research is needed to establish EV biogenetic pathways.

IV. EV functions and biogenesis in Metamonada flagellates: Trichomonas vaginalis and Giardia intestinalis

The large group of Metamonad protozoans include the retortamonads, diplomonads, and, possibly, the parabasalids and oxymonads as well [137], as the composition of this group is not entirely settled and taxonomic classification has been revised several times. Among its members are several human pathogens, such as G. intestinalis and T. vaginalis [138]. T. vaginalis is the causative agent of trichomoniasis, the most common pathogenic protozoan infection worldwide [139], with an estimated 160 million new cases of infection each year [139]. As to G. intestinalis (syn. Giardia lamblia), it is probably the most notorious of all metamonads [140], causing giardiasis, a gastrointestinal disease of humans and animals [140], with approximately 280 million new human cases of infection annually [138].

Metamonads utilize EVs (both exosomes and ectosomes) as a mechanism for intercellular communication [138,141,142], for survival and persistence within their hosts [143], and for host manipulation [138]. Among the wide range of bioactive molecules transferred within the various EV types [143] are virulence and differentiation factors [138]. The exosomes and microvesicles [143] that T. vaginalis secretes, for example, have been implicated in the modulation of parasite adherence [144,145] and the delivery of virulence factors to host cells [144]. They were also shown to modulate its host’s immune response by manipulating the expression of cytokines IL6 and IL8 in host vaginal epithelial cells [144] and modulating macrophage activity (by increasing the release of NO and inducing the secretion of anti-inflammatory cytokine IL10) [146].

Giardia parasites contain several acidified peripheral vacuoles (APVs), found adjacent to the plasma membrane, which act partially as endosomes and lysosomes [147,148]. Using high-resolution electron microscopy, it was found that ILVs are present inside some of the APVs, suggesting that those organelles could also act as MVBs [148]. Two distinct populations of EVs [24,149], each with a distinct proteomic profile and size, were described: exosome-like vesicles (ElVs) and microvesicles [24,149]. Both subtypes are internalized by mammalian cells [149], and one of them is able to mildly activate immature dendritic cells [150]. In addition, Giardia EVs are also involved in disrupting host intestinal epithelial junctions and in inhibiting the growth of commensal bacteria [151], which may account for the prevalence of post-infectious syndromes following disease eradication [151,152].

Several studies provide evidence for the involvement of ESCRT machinery in EV biogenesis in both T. vaginalis and G. intestinalis (and also cargo sorting in the case of T. vaginalis; [145]). Proteomic analysis of isolated exosomes and ectosomes harvested from T. vaginalis identified ESCRT-III machinery member VPS32 [143,144]. Indeed, VPS32 plays a key role in EV biogenesis and cargo sorting in T. vaginalis [145] (Fig 1, Metamonada). By using transgenic parasites expressing a standard T. vaginalis expression vector, VPS32 was found to localize to ILVs inside the MVBs as well as to ectosomes that protrude from the cell surface [145] (Fig 1, Metamonada). VPS32 is also present in EVs being endocytosed to the cell or exocytosed out of the cell [145]. Transgenic parasites overexpressing VPS32 adhered more strongly to host prostate cells compared to control parasites, highlighting this protein’s important role in mediating parasite–host interactions [145]. Two proteins identified in the proteomics data are Rab5 and Rab7, both of which have been implicated in exosome release in other cell types [153,154].

Giardia possess a relatively reduced portfolio of ESCRT machinery, containing only orthologs for Vps22 and Vps25 (ESCRT-II proteins) [155], Vps2 and Vps24 (ESCRT-III proteins) [45,155], Vps46 and AAA-ATPase Vps4 in its genome [45,155]. Transgenic parasites expressing deficient GlVps4a protein demonstrate a reduction in ILV formation on the APV and decreased ElV release compared to WT parasites [24] (Fig 1, Metamonada). Moreover, overexpression of GlVps4a led to more ILVs, but the amount of produced ElVs was similar to that of the WT strain, suggesting the involvement of another mechanism in EV release, independent of GlVps4a [24]. GIRab11, too, is involved in EV production in G. intestinalis [24]. Down-regulation of GIRab11 was shown to inhibit ElV formation in Rab11-deficient transgenic cells, while its overexpression raised the number of APVs containing ILVs, but not ElV release, suggesting that it participates in the biogenesis of ILVs [24]. Lastly, adding exogenous ceramide caused an increase in ILV formation inside APVs, and its localization in the endoplasmic reticulum, APV, and ILV membranes [24], similar to a known ESCRT-independent ceramide pathway involved in ILV and exosome formation [156]. In conclusion, ESCRT, Rab, and ceramide play a role in EV biogenesis in G. intestinalis.

V. EV functions and biogenesis in amoebae

Amoebae encompass a heterogeneous group of protozoa that move, at least in one phase of their life cycle, through cytoplasmic projections [157]. These organisms were grouped under the classical, yet obsolete, taxonomic group Sarcodina [157]. The biology of amoebae is quite variable, ranging from free-living organisms to facultative and obligatory parasites [157,158]. Thus, modern taxonomic approaches have reclassified many of its members to diverse taxons [158]. Five amoeba parasite species encompass most of the clinical cases in immunocompetent humans: Entamoeba histolytica (causes intestinal amoebiasis) [159], Naegleria fowleri (causes primary amoebic meningoencephalitis) [5], Balamuthia mandrillaris (causes granulomatous amoebic encephalitis), Acanthamoeba spp. (causes amoebic keratitis), and Sappinia diploidea (causes nongranulomatous amoebic encephalitis) [5].

The most studied amoeba-derived EVs are those produced by Acanthamoeba castellanii [160,161], and their immunomodulatory effects were explored in macrophages [162]. EV production by N. fowleri has not been demonstrated yet, even though it was shown to produce membrane vesiculation as a complement resistance mechanism [163] and to feature contact-dependent release of electrodense vesicles [164]. Moreover, several pathogenicity factors of N. fowleri are stored in intracellular vesicles, such as naegleriapore A and B [165], or appear in membrane-derived vesicles, such as the virulence factor CD59-like protein [166]. These data suggest the involvement of vesicular trafficking and, possibly, EV secretion, in the pathogenesis of N. fowleri, a parasite well-known to possess multiple pathogenic tools such as amoebostomes, secreted cytolytic enzymes, and immune protein deactivators [167]. EVs secreted by Entamoeba histolytica were characterized by proteomics [168], and specific small RNA populations were identified [168]. These EVs were demonstrated to regulate parasite–parasite communication during encystation [168]: While EVs derived from cysts promoted encystation of active trophozoites, trophozoite-derived EVs prevented encystation [168].

EV biogenesis remains mostly an open question in amoebae (Fig 1, Amoebae). ESCRT I, II, III, and ESCRT-associated homologues were identified in Naegleria gruberi [45], a nonpathogenic amoeba closely related to N. fowleri. Additionally, the amoebic homologue of Alix, an associated ESCRT protein, was found to be up-regulated in pathogenic strains of N. fowleri [169]. Members of ESCRT families 0, I, II, III, and accessory proteins were demonstrated to be up-regulated during erythrophagocytosis by E. histolytica [51]. Follow-up studies characterized the roles of the following members of the ESCRT system in E. histolytica: EhVps4 [51] (ESCRT accessory protein), EhADH112 [50] (homologue of human Alix), EhVps20, EhVps24, EhVps32, EhVps2 [48,50] (ESCRT-III members), and EhVps23 [47] (ESCRT-I member). These proteins were shown to be fundamental in canonical ESCRT activities, such as phagocytosis [47,49], MVB formation [50], and ubiquitin-mediated vesicular trafficking [4749]. Importantly, while EV biogenesis was not directly assessed, ESCRT-I member EhVps23 was observed within MVBs as well as in secreted exosomes [49,168], suggesting its connection to exosome biogenesis in E. histolytica (Fig 1, Amoebae). Interestingly, trophozoites overexpressing EhVps23 presented increased growth, phagocytosis, migration, and in vivo hepatic amoebic abscess formation [49], suggesting that this protein serves as an important pathogenesis factor [49]. Collectively, these data point to the involvement of ESCRT proteins in E. histolytica vesicular trafficking and, potentially, EV secretion, but further studies are needed to determine the involvement of the ESCRT system.

VI. Concluding remarks

The role of EVs in human parasitic protozoan biology and pathogenesis has been demonstrated by a variety of studies for apicomplexans, trypanosomatids, metamonads, and amoebae. In this review, we postulate that the ESCRT protein system, which is widely conserved throughout the evolution of protozoans [45], is an intimately involved player in EV biogenesis in these parasitic pathogens. ESCRT homologues have been identified and studied in all of the clades of human parasitic pathogens, with several studies directly characterizing the involvement of ESCRT protein members in EV biogenesis, specifically in Plasmodium falciparum [22], Trypanosoma brucei [23], Trichomonas vaginalis [145], Giardia intestinalis [24], and Entamoeba histolytica [49]. In addition, host- and parasite-derived ESCRT proteins are commonly found in EV proteomic profiles, and functional MVBs with ILVs have been observed multiple times in diverse parasites. Alternative (and possibly complimentary) EV biogenetic routes independent from the ESCRT system may be active and central in protozoan parasites, as was indeed shown for P. falciparum [19], Trypanosoma [15,23], and Giardia [24]. Some intracellular parasites may even be able to “hijack” the host’s ESCRT system for their own EV secretion [27], as viruses do [34,36].

The wide diversity in EV biogenesis throughout protozoan parasites is coherent with the different EV subpopulations recently identified in several parasites [23,63]. There is also great diversity in the size and protein cargo among the different life cycle stages and environmental stress conditions. Namely, parasites may use diverse EV biogenesis pathways as a robust response to environmental cues, including parasite–parasite and host–parasite interactions.

Two important obstacles that research into parasitic protozoa EV biogenesis faces are (1) the huge biological divergence of parasitic protozoa, which hinders the ability to find protein homologues throughout the widely diverse parasitic clades, and (2) the limited availability of experimental research models and genetic tools for many parasites, especially for intracellular stages, which makes the study of parasitic EVs methodologically challenging. Additionally, most mechanistic studies in the field have been performed with EVs isolated from in vitro parasite cultures, since their isolation and characterization from clinical in vivo samples remains arduous. Future studies that adapt more powerful bioinformatic tools for phylogenetic analyses of protein and gene homology, as well as the development and application of reproducible gene editing techniques to protozoans and improved EV isolation methods from clinical samples, will undoubtedly advance the field toward the full elucidation of the biological roles of understudied proteins in EV biology. Some current open questions are suggested in the Fig 2. Open questions in EV biogenesis of parasitic protozoa box.

thumbnail
Fig 2. Open questions in EV biogenesis of parasitic protozoa.

ESCRT, Endosomal Sorting Complex Required for Transport; EV, extracellular vesicle; MVB, multivesicular body.

https://doi.org/10.1371/journal.ppat.1011140.g002

References

  1. 1. Cowman AF, Berry D, Baum J. The cellular and molecular basis for malaria parasite invasion of the human red blood cell. J Cell Biol [Internet]. 2012 Sep 17;198(6):961–71. Available from: https://rupress.org/jcb/article/198/6/961/36943/The-cellular-and-molecular-basis-for-malaria. pmid:22986493
  2. 2. Rodrigues JCF, Godinho JLP, De Souza W. Biology of Human Pathogenic Trypanosomatids: Epidemiology, Lifecycle and Ultrastructure. Subcell Biochem. 2014:1–42. Available from: http://link.springer.com/10.1007/978-94-007-7305-9_1. pmid:24264239
  3. 3. Maudlin I. African trypanosomiasis. Ann Trop Med Parasitol [Internet]. 2006 Dec 18;100(8):679–701. Available from: http://www.tandfonline.com/doi/full/10.1179/136485906X112211. pmid:17227648
  4. 4. Kissinger P. Trichomonas vaginalis: a review of epidemiologic, clinical and treatment issues. BMC Infect Dis [Internet]. 2015 Dec 5;15(1):307. Available from: http://bmcinfectdis.biomedcentral.com/articles/10.1186/s12879-015-1055-0. pmid:26242185
  5. 5. Visvesvara GS, Moura H, Schuster FL. Pathogenic and opportunistic free-living amoebae: Acanthamoeba spp., Balamuthia mandrillaris, Naegleria fowleri, and Sappinia diploidea. FEMS Immunol Med Microbiol [Internet]. 2007 Jun;50(1):1–26. Available from: https://academic.oup.com/femspd/article-lookup/doi/10.1111/j.1574-695X.2007.00232.x. pmid:17428307
  6. 6. Szempruch AJ, Dennison L, Kieft R, Harrington JM, Hajduk SL. Sending a message: extracellular vesicles of pathogenic protozoan parasites. Nat Rev Microbiol [Internet]. 2016 Nov 12;14(11):669–75. Available from: http://www.nature.com/articles/nrmicro.2016.110. pmid:27615028
  7. 7. Ofir-Birin Y, Regev-Rudzki N. Extracellular vesicles in parasite survival. Science (80-) [Internet]. 2019 Feb 22;363(6429):817–8. Available from: https://www.science.org/doi/10.1126/science.aau4666. pmid:30792291
  8. 8. Yáñez-Mó M, Siljander PR-M, Andreu Z, Zavec AB, Borràs FE, Buzas EI, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles [Internet]. 2015;4:27066. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25979354. pmid:25979354
  9. 9. van Niel G D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol [Internet]. 2018 Apr 17;19(4):213–28. Available from: http://www.nature.com/articles/nrm.2017.125. pmid:29339798
  10. 10. Gill S, Catchpole R, Forterre P. Extracellular membrane vesicles in the three domains of life and beyond. FEMS Microbiol Rev [Internet]. 2019 May 1;43(3):273–303. Available from: https://academic.oup.com/femsre/article/43/3/273/5195520. pmid:30476045
  11. 11. Harding C V., Heuser JE, Stahl PD. Exosomes: Looking back three decades and into the future. J Cell Biol [Internet]. 2013 Feb 18;200(4):367–71. Available from: https://rupress.org/jcb/article/200/4/367/37222/Exosomes-Looking-back-three-decades-and-into-the. pmid:23420870
  12. 12. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci [Internet]. 2018 Jan 21;75(2):193–208. Available from: http://link.springer.com/10.1007/s00018-017-2595-9. pmid:28733901
  13. 13. Juan T, Fürthauer M. Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin Cell Dev Biol [Internet]. 2018 Feb;74:66–77. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1084952117302525. pmid:28807885
  14. 14. Bayer-Santos E, Lima FM, Ruiz JC, Almeida IC, da Silveira JF. Characterization of the small RNA content of Trypanosoma cruzi extracellular vesicles. Mol Biochem Parasitol [Internet]. 2014 Feb;193(2):71–4. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0166685114000206. pmid:24583081
  15. 15. Garcia-Silva MR, Cura Das Neves RF, Cabrera-Cabrera F, Sanguinetti J, Medeiros LC, Robello C, et al. Extracellular vesicles shed by Trypanosoma cruzi are linked to small RNA pathways, life cycle regulation, and susceptibility to infection of mammalian cells. Parasitol Res. 2014 Jan 17;113(1):285–304. pmid:24241124
  16. 16. Lambertz U, Oviedo Ovando ME, Vasconcelos EJ, Unrau PJ, Myler PJ, Reiner NE. Small RNAs derived from tRNAs and rRNAs are highly enriched in exosomes from both old and new world Leishmania providing evidence for conserved exosomal RNA Packaging. BMC Genomics [Internet]. 2015 Dec 5;16(1):151. Available from: https://bmcgenomics.biomedcentral.com/articles/10.1186/s12864-015-1260-7. pmid:25764986
  17. 17. Siles-Lucas M, Morchon R, Simon F, Manzano-Roman R. Exosome-transported microRNAs of helminth origin: new tools for allergic and autoimmune diseases therapy? Parasite Immunol [Internet]. 2015 Apr;37(4):208–14. Available from: https://onlinelibrary.wiley.com/doi/10.1111/pim.12182. pmid:25712154
  18. 18. Ofir-Birin Y, Ben Ami Pilo H, Cruz Camacho A, Rudik A, Rivkin A, Revach O-Y, et al. Malaria parasites both repress host CXCL10 and use it as a cue for growth acceleration. Nat Commun [Internet]. 2021 Dec 11;12(1):4851. Available from: https://www.nature.com/articles/s41467-021-24997-7. pmid:34381047
  19. 19. Regev-Rudzki N, Wilson DW, Carvalho TG, Sisquella X, Coleman BM, Rug M, et al. Cell-Cell Communication between Malaria-Infected Red Blood Cells via Exosome-like Vesicles. Cell [Internet]. 2013 May;153(5):1120–33. Available from: pmid:23683579
  20. 20. Sisquella X, Ofir-Birin Y, Pimentel MA, Cheng L, Abou Karam P, Sampaio NG, et al. Malaria parasite DNA-harbouring vesicles activate cytosolic immune sensors. Nat Commun [Internet]. 2017;8(1):1985. Available from: pmid:29215015
  21. 21. Douanne N, Dong G, Amin A, Bernardo L, Blanchette M, Langlais D, et al. Leishmania parasites exchange drug-resistance genes through extracellular vesicles. Cell Rep [Internet]. 2022 Jul;40(3):111121. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2211124722009275. pmid:35858561
  22. 22. Avalos-Padilla Y, Georgiev VN, Lantero E, Pujals S, Verhoef R, N. Borgheti-Cardoso L, et al. The ESCRT-III machinery participates in the production of extracellular vesicles and protein export during Plasmodium falciparum infection. Coppens I, editor. PLoS Pathog [Internet]. 2021 Apr 2;17(4):e1009455. Available from: https://dx.plos.org/10.1371/journal.ppat.1009455. pmid:33798247
  23. 23. Eliaz D, Kannan S, Shaked H, Arvatz G, Tkacz ID, Binder L, et al. Exosome secretion affects social motility in Trypanosoma brucei. PLoS Pathog. 2017 Mar 3;13(3):e1006245. pmid:28257521
  24. 24. Moyano S, Musso J, Feliziani C, Zamponi N, Frontera LS, Ropolo AS, et al. Exosome Biogenesis in the Protozoa Parasite Giardia lamblia: A Model of Reduced Interorganellar Crosstalk. Cells [Internet]. 2019 Dec 9;8(12):1600. Available from: https://www.mdpi.com/2073-4409/8/12/1600. pmid:31835439
  25. 25. van Dongen HM, Masoumi N, Witwer KW, Pegtel DM. Extracellular Vesicles Exploit Viral Entry Routes for Cargo Delivery. Microbiol Mol Biol Rev [Internet]. 2016 Jun;80(2):369–86. Available from: https://journals.asm.org/doi/10.1128/MMBR.00063-15. pmid:26935137
  26. 26. Alenquer M, Amorim M. Exosome Biogenesis, Regulation, and Function in Viral Infection. Viruses [Internet]. 2015 Sep 17;7(9):5066–83. Available from: http://www.mdpi.com/1999-4915/7/9/2862.
  27. 27. Gioseffi A, Edelmann MJ, Kima PE. Intravacuolar Pathogens Hijack Host Extracellular Vesicle Biogenesis to Secrete Virulence Factors. Front Immunol. 2021;12:662944. pmid:33959131
  28. 28. Vietri M, Radulovic M, Stenmark H. The many functions of ESCRTs. Nat Rev Mol Cell Biol [Internet]. 2020 Jan 8;21(1):25–42. Available from: http://www.nature.com/articles/s41580-019-0177-4. pmid:31705132
  29. 29. Campsteijn C, Vietri M, Stenmark H. Novel ESCRT functions in cell biology: spiraling out of control? Curr Opin Cell Biol [Internet]. 2016 Aug;41:1–8. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0955067416300382. pmid:27031044
  30. 30. Alfred V, Vaccari T. When membranes need an ESCRT: endosomal sorting and membrane remodelling in health and disease. Swiss Med Wkly [Internet]. 2016 Sep 15. Available from: http://doi.emh.ch/smw.2016.14347. pmid:27631343
  31. 31. Raiborg C, Stenmark H. The ESCRT machinery in endosomal sorting of ubiquitylated membrane proteins. Nature [Internet]. 2009 Mar 26;458(7237):445–52. Available from: https://www.nature.com/articles/nature07961. pmid:19325624
  32. 32. Jimenez AJ, Maiuri P, Lafaurie-Janvore J, Divoux S, Piel M, Perez F. ESCRT Machinery Is Required for Plasma Membrane Repair. Science (80-) [Internet]. 2014 Feb 28;343(6174). Available from: https://www.science.org/doi/10.1126/science.1247136. pmid:24482116
  33. 33. Scheffer LL, Sreetama SC, Sharma N, Medikayala S, Brown KJ, Defour A, et al. Mechanism of Ca2+-triggered ESCRT assembly and regulation of cell membrane repair. Nat Commun [Internet]. 2014 Dec 23;5(1):5646. Available from: http://www.nature.com/articles/ncomms6646.
  34. 34. Carlton JG, Martin-Serrano J. Parallels Between Cytokinesis and Retroviral Budding: A Role for the ESCRT Machinery. Science (80-) [Internet]. 2007 Jun 29;316(5833):1908–12. Available from: https://www.science.org/doi/10.1126/science.1143422.
  35. 35. Carlton JG, Agromayor M, Martin-Serrano J. Differential requirements for Alix and ESCRT-III in cytokinesis and HIV-1 release. Proc Natl Acad Sci [Internet]. 2008 Jul 29;105(30):10541–6. Available from: https://pnas.org/doi/full/10.1073/pnas.0802008105. pmid:18641129
  36. 36. Votteler J, Sundquist WI. Virus Budding and the ESCRT Pathway. Cell Host Microbe [Internet]. 2013 Sep;14(3):232–41. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1931312813002941. pmid:24034610
  37. 37. Hurley JH. ESCRT s are everywhere. EMBO J [Internet]. 2015 Oct 26;34(19):2398–407. Available from: https://onlinelibrary.wiley.com/doi/10.15252/embj.201592484. pmid:26311197
  38. 38. Hurley JH. ESCRT complexes and the biogenesis of multivesicular bodies. Curr Opin Cell Biol [Internet]. 2008 Feb;20(1):4–11. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0955067407001913. pmid:18222686
  39. 39. Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol [Internet]. 2015 Jun;25(6):364–72. Available from: https://linkinghub.elsevier.com/retrieve/pii/S096289241500015X. pmid:25683921
  40. 40. Babst M, Katzmann DJ, Snyder WB, Wendland B, Emr SD. Endosome-Associated Complex, ESCRT-II, Recruits Transport Machinery for Protein Sorting at the Multivesicular Body. Dev Cell [Internet]. 2002 Aug;3(2):283–9. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1534580702002198. pmid:12194858
  41. 41. Henne WM, Buchkovich NJ, Emr SD. The ESCRT Pathway. Dev Cell [Internet]. 2011 Jul;21(1):77–91. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1534580711002073. pmid:21763610
  42. 42. Henne WM, Stenmark H, Emr SD. Molecular Mechanisms of the Membrane Sculpting ESCRT Pathway. Cold Spring Harb Perspect Biol [Internet]. 2013 Sep 1;5(9):a016766–a016766. Available from: http://cshperspectives.cshlp.org/lookup/doi/10.1101/cshperspect.a016766. pmid:24003212
  43. 43. Dores MR, Chen B, Lin H, Soh UJK, Paing MM, Montagne WA, et al. ALIX binds a YPX3L motif of the GPCR PAR1 and mediates ubiquitin-independent ESCRT-III/MVB sorting. J Cell Biol [Internet]. 2012 Apr 30;197(3):407–19. Available from: https://rupress.org/jcb/article/197/3/407/36930/ALIX-binds-a-YPX3L-motif-of-the-GPCR-PAR1-and. pmid:22547407
  44. 44. Mir B, Goettsch C. Extracellular Vesicles as Delivery Vehicles of Specific Cellular Cargo. Cells [Internet]. 2020 Jul 2;9(7):1601. Available from: https://www.mdpi.com/2073-4409/9/7/1601. pmid:32630649
  45. 45. Leung KF, Dacks JB, Field MC. Evolution of the multivesicular body ESCRT machinery; retention across the eukaryotic lineage. Traffic. 2008 Oct;9(10):1698–716. pmid:18637903
  46. 46. Silverman JS, Muratore KA, Bangs JD. Characterization of the late endosomal ESCRT machinery in trypanosoma brucei. Traffic. 2013 Oct;14(10):1078–90. pmid:23905922
  47. 47. Galindo A, Javier-Reyna R, García-Rivera G, Bañuelos C, Montaño S, Ortega-Lopez J, et al. EhVps23: A Component of ESCRT-I That Participates in Vesicular Trafficking and Phagocytosis of Entamoeba histolytica. Front Cell Infect Microbiol [Internet]. 2021 Oct 29;11. Available from: https://www.frontiersin.org/articles/10.3389/fcimb.2021.770759/full. pmid:34778112
  48. 48. Avalos-Padilla Y, Knorr RL, Javier-Reyna R, García-Rivera G, Lipowsky R, Dimova R, et al. The Conserved ESCRT-III Machinery Participates in the Phagocytosis of Entamoeba histolytica. Front Cell Infect Microbiol [Internet]. 2018 Mar 1;8. Available from: http://journal.frontiersin.org/article/10.3389/fcimb.2018.00053/full. pmid:29546036
  49. 49. Galindo A, Javier-Reyna R, García-Rivera G, Bañuelos C, Chávez-Munguía B, Salazar-Villatoro L, et al. EhVps23, an ESCRT-I Member, Is a Key Factor in Secretion, Motility, Phagocytosis and Tissue Invasion by Entamoeba histolytica. Front Cell Infect Microbiol [Internet]. 2022 Mar 14;12. Available from: https://www.frontiersin.org/articles/10.3389/fcimb.2022.835654/full. pmid:35360117
  50. 50. Bañuelos C, García-Rivera G, López-Reyes I, Mendoza L, González-Robles A, Herranz S, et al. EhADH112 Is a Bro1 Domain-Containing Protein Involved in the Entamoeba histolytica Multivesicular Bodies Pathway. J Biomed Biotechnol [Internet]. 2012;2012:1–15. Available from: http://www.hindawi.com/journals/bmri/2012/657942/.
  51. 51. López-Reyes I, García-Rivera G, Bañuelos C, Herranz S, Vincent O, López-Camarillo C, et al. Detection of the Endosomal Sorting Complex Required for Transport in Entamoeba histolytica and Characterization of the EhVps4 Protein. J Biomed Biotechnol [Internet]. 2010;2010:1–15. Available from: http://www.hindawi.com/journals/bmri/2010/890674/. pmid:20508821
  52. 52. Rojas-Pirela M, Medina L, Rojas MV, Liempi AI, Castillo C, Pérez-Pérez E, et al. Congenital Transmission of Apicomplexan Parasites: A Review. Front Microbiol [Internet]. 2021 Sep 29;12. Available from: https://www.frontiersin.org/articles/10.3389/fmicb.2021.751648/full. pmid:34659187
  53. 53. Kolářová I, Valigurová A. Hide-and-Seek: A Game Played between Parasitic Protists and Their Hosts. Microorganisms [Internet]. 2021 Nov 25;9(12):2434. Available from: https://www.mdpi.com/2076-2607/9/12/2434. pmid:34946036
  54. 54. Seeber F, Steinfelder S. Recent advances in understanding apicomplexan parasites. F1000Res [Internet]. 2016 Jun 14;5:1369. Available from: https://f1000research.com/articles/5-1369/v1. pmid:27347391
  55. 55. Marcilla A, Martin-Jaular L, Trelis M, de Menezes-Neto A, Osuna A, Bernal D, et al. Extracellular vesicles in parasitic diseases. J Extracell Vesicles [Internet]. 2014 Jan 22;3(1):25040. Available from: https://www.tandfonline.com/doi/full/10.3402/jev.v3.25040. pmid:25536932
  56. 56. Babatunde KA, Yesodha Subramanian B, Ahouidi AD, Martinez Murillo P, Walch M, Mantel P-Y. Role of Extracellular Vesicles in Cellular Cross Talk in Malaria. Front Immunol [Internet]. 2020 Jan 31;11. Available from: https://www.frontiersin.org/article/10.3389/fimmu.2020.00022/full. pmid:32082312
  57. 57. Mantel P-Y, Hjelmqvist D, Walch M, Kharoubi-Hess S, Nilsson S, Ravel D, et al. Infected erythrocyte-derived extracellular vesicles alter vascular function via regulatory Ago2-miRNA complexes in malaria. Nat Commun [Internet]. 2016 Nov 10;7(1):12727. Available from: http://www.nature.com/articles/ncomms12727. pmid:27721445
  58. 58. Dekel E, Yaffe D, Rosenhek-Goldian I, Ben-Nissan G, Ofir-Birin Y, Morandi MI, et al. 20S proteasomes secreted by the malaria parasite promote its growth. Nat Commun [Internet]. 2021 Dec 19;12(1):1172. Available from: http://www.nature.com/articles/s41467-021-21344-8. pmid:33608523
  59. 59. Ye W, Chew M, Hou J, Lai F, Leopold SJ, Loo HL, et al. Microvesicles from malaria-infected red blood cells activate natural killer cells via MDA5 pathway. Goodier M, editor. PLoS Pathog [Internet]. 2018 Oct 4;14(10):e1007298. Available from: https://dx.plos.org/10.1371/journal.ppat.1007298. pmid:30286211
  60. 60. Silva VO, Maia MM, Torrecilhas AC, Taniwaki NN, Namiyama GM, Oliveira KC, et al. Extracellular vesicles isolated from Toxoplasma gondii induce host immune response. Parasite Immunol [Internet]. 2018 Sep;40(9):e12571. Available from: https://onlinelibrary.wiley.com/doi/10.1111/pim.12571. pmid:29974519
  61. 61. Mantel P-Y, Hoang AN, Goldowitz I, Potashnikova D, Hamza B, Vorobjev I, et al. Malaria-infected erythrocyte-derived microvesicles mediate cellular communication within the parasite population and with the host immune system. Cell Host Microbe. 2013 May;13(5):521–34. pmid:23684304
  62. 62. Toda H, Diaz-Varela M, Segui-Barber J, Roobsoong W, Baro B, Garcia-Silva S, et al. Plasma-derived extracellular vesicles from Plasmodium vivax patients signal spleen fibroblasts via NF-kB facilitating parasite cytoadherence. Nat Commun [Internet]. 2020 Dec 2;11(1):2761. Available from: http://www.nature.com/articles/s41467-020-16337-y. pmid:32487994
  63. 63. Abou Karam P, Rosenhek-Goldian I, Ziv T, Ben Ami Pilo H, Azuri I, Rivkin A, et al. Malaria parasites release vesicle subpopulations with signatures of different destinations. EMBO Rep [Internet]. 2022 Jun. Available from: https://onlinelibrary.wiley.com/doi/10.15252/embr.202254755. pmid:35642585
  64. 64. Ben Ami Pilo H, Khan Khilji S, Lühle J, Biskup K, Levy Gal B, Rosenhek Goldian I, et al. Sialylated N -glycans mediate monocyte uptake of extracellular vesicles secreted from Plasmodium falciparum -infected red blood cells. J Extracell Biol [Internet]. 2022 Feb 21;1(2). Available from: https://onlinelibrary.wiley.com/doi/10.1002/jex2.33.
  65. 65. Gulati S, Ekland EH, Ruggles K V, Chan RB, Jayabalasingham B, Zhou B, et al. Profiling the Essential Nature of Lipid Metabolism in Asexual Blood and Gametocyte Stages of Plasmodium falciparum. Cell Host Microbe [Internet]. 2015 Sep 9;18(3):371–81. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26355219. pmid:26355219
  66. 66. Borgheti-Cardoso LN, Kooijmans SAA, Chamorro LG, Biosca A, Lantero E, Ramírez M, et al. Extracellular vesicles derived from Plasmodium-infected and non-infected red blood cells as targeted drug delivery vehicles. Int J Pharm [Internet]. 2020 Sep 25;587:119627. Available from: http://www.ncbi.nlm.nih.gov/pubmed/32653596. pmid:32653596
  67. 67. Neveu G, Richard C, Dupuy F, Behera P, Volpe F, Subramani PA, et al. Plasmodium falciparum sexual parasites develop in human erythroblasts and affect erythropoiesis. Blood. 2020 Sep;136(12):1381–93. pmid:32589714
  68. 68. Debs S, Cohen A, Hosseini-Beheshti E, Chimini G, Hunt NH, Grau GER. Interplay of extracellular vesicles and other players in cerebral malaria pathogenesis. Biochim Biophys Acta Gen Subj [Internet]. 2019 Feb;1863(2):325–31. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0304416518303337. pmid:30339917
  69. 69. Campos FM, Franklin BS, Teixeira-Carvalho A, Filho AL, de Paula SC, Fontes CJ, et al. Augmented plasma microparticles during acute Plasmodium vivax infection. Malar J [Internet]. 2010 Dec 16;9(1):327. Available from: https://malariajournal.biomedcentral.com/articles/10.1186/1475-2875-9-327. pmid:21080932
  70. 70. Attias M, Teixeira DE, Benchimol M, Vommaro RC, Crepaldi PH, De Souza W. The life-cycle of Toxoplasma gondii reviewed using animations. Parasit Vectors [Internet]. 2020 Dec 23;13(1):588. Available from: https://parasitesandvectors.biomedcentral.com/articles/10.1186/s13071-020-04445-z. pmid:33228743
  71. 71. Matta SK, Rinkenberger N, Dunay IR, Sibley LD. Toxoplasma gondii infection and its implications within the central nervous system. Nat Rev Microbiol [Internet]. 2021 Jul 24;19(7):467–80. Available from: http://www.nature.com/articles/s41579-021-00518-7. pmid:33627834
  72. 72. Wowk PF, Zardo ML, Miot HT, Goldenberg S, Carvalho PC, Mörking PA. Proteomic profiling of extracellular vesicles secreted from Toxoplasma gondii. Proteomics [Internet]. 2017 Aug;17(15–16):1600477. Available from: https://onlinelibrary.wiley.com/doi/10.1002/pmic.201600477. pmid:28643940
  73. 73. Li D-L, Zou W-H, Deng S-Q, Peng H-J. Analysis of the Differential Exosomal miRNAs of DC2.4 Dendritic Cells Induced by Toxoplasma gondii Infection. Int J Mol Sci [Internet]. 2019 Nov 5;20(21):5506. Available from: https://www.mdpi.com/1422-0067/20/21/5506. pmid:31694199
  74. 74. Ramírez-Flores C, Mondragón R. Elucidating the Role of Extracellular Vesicles Released by Toxoplasma gondii: A Review. J Data Mining Genomics Proteomics. 2021 Feb 13;1.
  75. 75. Li Y, Xiu F, Mou Z, Xue Z, Du H, Zhou C, et al. Exosomes derived from Toxoplasma gondii stimulate an inflammatory response through JNK signaling pathway. Nanomedicine [Internet]. 2018 May;13(10):1157–68. Available from: https://www.futuremedicine.com/doi/10.2217/nnm-2018-0035. pmid:29542367
  76. 76. Li Y, Liu Y, Xiu F, Wang J, Cong H, He S, et al. Characterization of exosomes derived from Toxoplasma gondii and their functions in modulating immune responses. Int J Nanomedicine [Internet]. 2018 Jan;13:467–77. Available from: https://www.dovepress.com/characterization-of-exosomes-derived-from-toxoplasma-gondii-and-their—peer-reviewed-article-IJN. pmid:29403276
  77. 77. Jimenez-Ruiz E, Morlon-Guyot J, Daher W, Meissner M. Vacuolar protein sorting mechanisms in apicomplexan parasites. Mol Biochem Parasitol [Internet]. 2016 Sep;209(1–2):18–25. Available from: https://linkinghub.elsevier.com/retrieve/pii/S016668511630007X. pmid:26844642
  78. 78. Yang M, Coppens I, Wormsley S, Baevova P, Hoppe HC, Joiner KA. The Plasmodium falciparum Vps4 homolog mediates multivesicular body formation. J Cell Sci [Internet]. 2004 Aug 1;117(17):3831–8. Available from: https://journals.biologists.com/jcs/article/117/17/3831/27845/The-Plasmodium-falciparum-Vps4-homolog-mediates. pmid:15252121
  79. 79. Wollert T, Hurley JH. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature [Internet]. 2010 Apr 21;464(7290):864–9. Available from: http://www.nature.com/articles/nature08849. pmid:20305637
  80. 80. Booth A, Marklew CJ, Ciani B, Beales PA. In Vitro Membrane Remodeling by ESCRT is Regulated by Negative Feedback from Membrane Tension. iScience [Internet]. 2019 May;15:173–84. Available from: https://linkinghub.elsevier.com/retrieve/pii/S258900421930118X.
  81. 81. Mundwiler-Pachlatko E, Beck H-P. Maurer’s clefts, the enigma of Plasmodium falciparum. Proc Natl Acad Sci [Internet]. 2013;110(50):19987–19994. Available from: https://www.pnas.org/doi/abs/10.1073/pnas.1309247110.
  82. 82. Maier AG, Rug M, O’Neill MT, Brown M, Chakravorty S, Szestak T, et al. Exported Proteins Required for Virulence and Rigidity of Plasmodium falciparum-Infected Human Erythrocytes. Cell [Internet]. 2008 Jul;134(1):48–61. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0092867408006910. pmid:18614010
  83. 83. Cygan AM, Jean Beltran PM, Mendoza AG, Branon TC, Ting AY, Carr SA, et al. Proximity-Labeling Reveals Novel Host and Parasite Proteins at the Toxoplasma Parasitophorous Vacuole Membrane. Blader IJ, Coyne CB, editors. MBio [Internet]. 2021 Dec 21;12(6). Available from: https://journals.asm.org/doi/10.1128/mBio.00260-21. pmid:34749525
  84. 84. Guérin A, Corrales RM, Parker ML, Lamarque MH, Jacot D, El Hajj H, et al. Efficient invasion by Toxoplasma depends on the subversion of host protein networks. Nat Microbiol [Internet]. 2017 Oct 28;2(10):1358–66. Available from: http://www.nature.com/articles/s41564-017-0018-1. pmid:28848228
  85. 85. Rivera-Cuevas Y, Mayoral J, Di Cristina M, Lawrence A-LE, Olafsson EB, Patel RK, et al. Toxoplasma gondii exploits the host ESCRT machinery for parasite uptake of host cytosolic proteins. Olias P, editor. PLoS Pathog [Internet]. 2021 Dec 13;17(12):e1010138. Available from: https://dx.plos.org/10.1371/journal.ppat.1010138. pmid:34898650
  86. 86. Nussbaum K, Honek J C.v.C, Cadmus C, Efferth T. Trypanosomatid Parasites Causing Neglected Diseases. Curr Med Chem. 2010 May 1;17(15):1594–617. pmid:20166934
  87. 87. Bodimeade C, Marks M, Mabey D. Neglected tropical diseases: Elimination and eradication. Clin Med J R Coll Physicians London. 2019 Mar 14;19(2):157–60.
  88. 88. Rossi IV, Ferreira Nunes MA, Vargas-Otalora S, da Silva Ferreira TC, Cortez M, Ramirez MI. Extracellular Vesicles during TriTryps infection: Complexity and future challenges. Mol Immunol [Internet]. 2021 Apr;132:172–83. Available from: https://www.sciencedirect.com/science/article/pii/S0161589021000080. pmid:33601226
  89. 89. Torrecilhas AC, Soares RP, Schenkman S, Fernández-Prada C, Olivier M. Extracellular Vesicles in Trypanosomatids: Host Cell Communication. Front Cell Infect Microbiol. 2020;10:602502. pmid:33381465
  90. 90. Neves RFC, Fernandes ACS, Meyer-Fernandes JR, Souto-Padrón T. Trypanosoma cruzi-secreted vesicles have acid and alkaline phosphatase activities capable of increasing parasite adhesion and infection. Parasitol Res. 2014 Aug 7;113(8):2961–72. pmid:24906990
  91. 91. Trocoli Torrecilhas AC, Tonelli RR, Pavanelli WR, da Silva JS, Schumacher RI, de Souza W, et al. Trypanosoma cruzi: parasite shed vesicles increase heart parasitism and generate an intense inflammatory response. Microbes Infect. 2009 Jan;11(1):29–39. pmid:19028594
  92. 92. Hassani K, Olivier M. Immunomodulatory Impact of Leishmania-Induced Macrophage Exosomes: A Comparative Proteomic and Functional Analysis. Milon G, editor. PLoS Negl Trop Dis [Internet]. 2013 May 2;7(5):e2185. Available from: https://dx.plos.org/10.1371/journal.pntd.0002185. pmid:23658846
  93. 93. Gioseffi A, Hamerly T, Van K, Zhang N, Dinglasan RR, Yates PA, et al. Leishmania -infected macrophages release extracellular vesicles that can promote lesion development. Life Sci Alliance [Internet]. 2020 Dec 29;3(12):e202000742. Available from: https://www.life-science-alliance.org/lookup/doi/10.26508/lsa.202000742. pmid:33122174
  94. 94. Soto-Serna LE, Diupotex M, Zamora-Chimal J, Ruiz-Remigio A, Delgado-Domínguez J, Cervantes-Sarabia RB, et al. Leishmania mexicana: Novel Insights of Immune Modulation through Amastigote Exosomes. J Immunol Res [Internet]. 2020;2020:8894549. Available from: http://www.ncbi.nlm.nih.gov/pubmed/33344659. pmid:33344659
  95. 95. Wyllie MP, Ramirez MI. Microvesicles released during the interaction between Trypanosoma cruzi TcI and TcII strains and host blood cells inhibit complement system and increase the infectivity of metacyclic forms of host cells in a strain-independent process. Pathog Dis. 2017 Sep 29;75(7).
  96. 96. de Souza W, Barrias ES. Membrane-bound extracellular vesicles secreted by parasitic protozoa: cellular structures involved in the communication between cells. Parasitol Res. 2020 Jul 12;119(7):2005–23. pmid:32394001
  97. 97. Ribeiro KS, Vasconcellos CI, Soares RP, Mendes MT, Ellis CC, Aguilera-Flores M, et al. Proteomic analysis reveals different composition of extracellular vesicles released by two Trypanosoma cruzi strains associated with their distinct interaction with host cells. J Extracell Vesicles. 2018 Dec 1;7(1):1463779. pmid:29696081
  98. 98. Geiger A, Hirtz C, Bécue T, Bellard E, Centeno D, Gargani D, et al. Exocytosis and protein secretion in Trypanosoma. BMC Microbiol. 2010;10(1):20. pmid:20102621
  99. 99. Silverman JM, Chan SK, Robinson DP, Dwyer DM, Nandan D, Foster LJ, et al. Proteomic analysis of the secretome of Leishmania donovani. Genome Biol [Internet]. 2008;9(2):R35. Available from: http://genomebiology.biomedcentral.com/articles/10.1186/gb-2008-9-2-r35. pmid:18282296
  100. 100. Silverman JM, Clos J, De’Oliveira CC, Shirvani O, Fang Y, Wang C, et al. An exosome-based secretion pathway is responsible for protein export from Leishmania and communication with macrophages. J Cell Sci [Internet]. 2010 Mar 15;123(6):842–52. Available from: https://journals.biologists.com/jcs/article/123/6/842/31455/An-exosome-based-secretion-pathway-is-responsible. pmid:20159964
  101. 101. Corrales RM, Sereno D, Mathieu-Daudé F. Deciphering the Leishmania exoproteome: What we know and what we can learn. FEMS Immunology and Medical Microbiology. 2010. p. 27–38. pmid:19807787
  102. 102. Douanne N, Dong G, Douanne M, Olivier M, Fernandez-Pradaid C. Unravelling the proteomic signature of extracellular vesicles released by drug-resistant leishmania infantum parasites. PLoS Negl Trop Dis. 2020 Jul 6;14(7):1–32.
  103. 103. Szempruch AJ, Sykes SE, Kieft R, Dennison L, Becker AC, Gartrell A, et al. Extracellular Vesicles from Trypanosoma brucei Mediate Virulence Factor Transfer and Cause Host Anemia. Cell. 2016 Jan;164(1–2):246–57. pmid:26771494
  104. 104. Stijlemans B, Caljon G, Van Den Abbeele J, Van Ginderachter JA, Magez S, De Trez C. Immune evasion strategies of Trypanosoma brucei within the mammalian host: Progression to pathogenicity. Front Immunol. 2016;7:233. pmid:27446070
  105. 105. Dozio V, Lejon V, Mumba Ngoyi D, Büscher P, Sanchez JC, Tiberti N. Cerebrospinal Fluid-Derived Microvesicles From Sleeping Sickness Patients Alter Protein Expression in Human Astrocytes. Front Cell Infect Microbiol. 2019;20:9. pmid:31824868
  106. 106. Field MC, Carrington M. The trypanosome flagellar pocket. Nat Rev Microbiol. 2009;7:775–786. pmid:19806154
  107. 107. Saada EA, DeMarco SF, Shimogawa MM, Hill KL. “With a Little Help from My Friends”—Social Motility in Trypanosoma brucei. PLoS Pathogens. 2015;11:e1005272. pmid:26679190
  108. 108. Ramirez MI, Deolindo P, de Messias-Reason IJ, Arigi EA, Choi H, Almeida IC, et al. Dynamic flux of microvesicles modulate parasite–host cell interaction of Trypanosoma cruzi in eukaryotic cells. Cell Microbiol. 2017 Apr;19(4):e12672. pmid:27665486
  109. 109. Moreira LR, Serrano FR, Osuna A. Extracellular vesicles of trypanosoma cruzi tissue-culture cell-derived trypomastigotes: Induction of physiological changes in non-parasitized culture cells. PLoS Negl Trop Dis. 2019 Feb 21;13(2):e0007163. pmid:30789912
  110. 110. Paranaiba LF, Guarneri AA, Torrecilhas AC, Melo MN, Soares RP. Extracellular vesicles isolated from Trypanosoma cruzi affect early parasite migration in the gut of rhodnius prolixus but not in Triatoma infestans. Mem Inst Oswaldo Cruz. 2019;114(11). pmid:31851215
  111. 111. Nogueira PM, Ribeiro K, Silveira ACO, Campos JH, Martins-Filho OA, Bela SR, et al. Vesicles from different Trypanosoma cruzi strains trigger differential innate and chronic immune responses. J Extracell Vesicles. 2015 Jan 1;4(1):28734. pmid:26613751
  112. 112. De Pablos LM, Díaz Lozano IM, Jercic MI, Quinzada M, Giménez MJ, Calabuig E, et al. The C-terminal region of Trypanosoma cruzi MASPs is antigenic and secreted via exovesicles. Sci Rep. 2016 Jun 8;6(1):27293. pmid:27270330
  113. 113. Díaz Lozano IM, De Pablos LM, Longhi SA, Zago MP, Schijman AG, Osuna A. Immune complexes in chronic Chagas disease patients are formed by exovesicles from Trypanosoma cruzi carrying the conserved MASP N-terminal region. Sci Rep. 2017 Apr 15;7(1):44451. pmid:28294160
  114. 114. Torró LM d. P, Moreira LR, Osuna A. Extracellular vesicles in chagas disease: A new passenger for an old disease. Front Microbiol. 2018;9:1190. pmid:29910793
  115. 115. Dantas-Pereira L, Menna-Barreto R, Lannes-Vieira J. Extracellular Vesicles: Potential Role in Remote Signaling and Inflammation in Trypanosoma cruzi-Triggered Disease. Front Cell Dev Biol. 2021;9:798054. pmid:34988085
  116. 116. Moreira LR, Prescilla-Ledezma A, Cornet-Gomez A, Linares F, Jódar-Reyes AB, Fernandez J, et al. Biophysical and biochemical comparison of extracellular vesicles produced by infective and non-infective stages of trypanosoma cruzi. Int J Mol Sci. 2021 May 13;22(10):5183. pmid:34068436
  117. 117. Bonfim-Melo A, Ferreira ER, Florentino PTV, Mortara RA. Amastigote synapse: The tricks of Trypanosoma cruzi extracellular amastigotes. Front Microbiol. 2018;9:1341. pmid:30013522
  118. 118. Cronemberger-Andrade A, Xander P, Soares RP, Pessoa NL, Campos MA, Ellis CC, et al. Trypanosoma cruzi-Infected Human Macrophages Shed Proinflammatory Extracellular Vesicles That Enhance Host-Cell Invasion via Toll-Like Receptor 2. Front Cell Infect Microbiol. 2020 Mar 20;10. pmid:32266161
  119. 119. Bayer-Santos E, Aguilar-Bonavides C, Rodrigues SP, Cordero EM, Marques AF, Varela-Ramirez A, et al. Proteomic analysis of trypanosoma cruzi secretome: Characterization of two populations of extracellular vesicles and soluble proteins. J Proteome Res. 2013 Feb 1;12(2):883–97. pmid:23214914
  120. 120. Castelli G, Bruno F, Saieva L, Alessandro R, Galluzzi L, Diotallevi A, et al. Exosome secretion by Leishmania infantum modulate the chemotactic behavior and cytokinic expression creating an environment permissive for early infection. Exp Parasitol [Internet]. 2019 Mar;198:39–45. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0014489418303345. pmid:30716304
  121. 121. Nogueira PM, de Menezes-Neto A, Borges VM, Descoteaux A, Torrecilhas AC, Xander P, et al. Immunomodulatory Properties of Leishmania Extracellular Vesicles During Host-Parasite Interaction: Differential Activation of TLRs and NF-κB Translocation by Dermotropic and Viscerotropic Species. Front Cell Infect Microbiol [Internet]. 2020 Jul 29;10. Available from: https://www.frontiersin.org/article/10.3389/fcimb.2020.00380/full.
  122. 122. Colineau L, Clos J, Moon K-M, Foster LJ, Reiner NE. Leishmania donovani chaperonin 10 regulates parasite internalization and intracellular survival in human macrophages. Med Microbiol Immunol [Internet]. 2017 Jun 11;206(3):235–57. Available from: http://link.springer.com/10.1007/s00430-017-0500-7. pmid:28283754
  123. 123. Atayde VD, Aslan H, Townsend S, Hassani K, Kamhawi S, Olivier M. Exosome Secretion by the Parasitic Protozoan Leishmania within the Sand Fly Midgut. Cell Rep [Internet]. 2015;13(5):957–967. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2211124715010839. pmid:26565909
  124. 124. Hassani K, Shio MT, Martel C, Faubert D, Olivier M. Absence of Metalloprotease GP63 Alters the Protein Content of Leishmania Exosomes. Langsley G, editor. PLoS ONE [Internet]. 2014 Apr 15;9(4):e95007. Available from: https://dx.plos.org/10.1371/journal.pone.0095007. pmid:24736445
  125. 125. Ghosh J, Bose M, Roy S, Bhattacharyya SN. Leishmania donovani Targets Dicer1 to Downregulate miR-122, Lower Serum Cholesterol, and Facilitate Murine Liver Infection. Cell Host Microbe [Internet]. 2013 Mar;13(3):277–88. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1931312813000711. pmid:23498953
  126. 126. Dong G, Filho AL, Olivier M. Modulation of host-pathogen communication by extracellular vesicles (EVs) of the protozoan parasite Leishmania. Front Cell Infect Microbiol. 2019;9:100. pmid:31032233
  127. 127. Forrest DM, Batista M, Marchini FK, Tempone AJ, Traub-Csekö YM. Proteomic analysis of exosomes derived from procyclic and metacyclic-like cultured Leishmania infantum chagasi. J Proteomics [Internet]. 2020 Sep;227:103902. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1874391920302700. pmid:32673755
  128. 128. Atayde VD, Hassani K, da Silva Lira Filho A, Borges AR, Adhikari A, Martel C, et al. Leishmania exosomes and other virulence factors: Impact on innate immune response and macrophage functions. Cell Immunol [Internet]. 2016 Nov;309:7–18. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0008874916300624. pmid:27499212
  129. 129. Field MC, Natesan SKA, Gabernet-Castello C, Lila Koumandou V. Intracellular Trafficking in the Trypanosomatids. Traffic [Internet]. 2007 Jun;8(6):629–39. Available from: https://onlinelibrary.wiley.com/doi/10.1111/j.1600-0854.2007.00558.x. pmid:17461800
  130. 130. Atayde VD, da Silva Lira Filho A, Chaparro V, Zimmermann A, Martel C, Jaramillo M, et al. Exploitation of the Leishmania exosomal pathway by Leishmania RNA virus 1. Nat Microbiol [Internet]. 2019 Apr 28;4(4):714–23. Available from: http://www.nature.com/articles/s41564-018-0352-y. pmid:30692670
  131. 131. Vasconcelos CI, Cronemberger-Andrade A, Souza-Melo N, Maricato JT, Xander P, Batista WL, et al. Stress Induces Release of Extracellular Vesicles by Trypanosoma cruzi Trypomastigotes. J Immunol Res. 2021 Sep 23;2021:1–12. pmid:34604391
  132. 132. Leung KF, Riley FS, Carrington M, Field MC. Ubiquitylation and developmental regulation of invariant surface protein expression in trypanosomes. Eukaryot Cell. 2011 Jul;10(7):916–31. pmid:21571921
  133. 133. Umaer K, Bangs JD. Late ESCRT machinery mediates the recycling and Rescue of Invariant Surface Glycoprotein 65 in Trypanosoma brucei. Cell Microbiol. 2020 Nov 13;22(11). pmid:32618070
  134. 134. Engstler M, Thilo L, Weise F, Grünfelder CG, Schwarz H, Boshart M, et al. Kinetics of endocytosis and recycling of the GPI-anchored variant surface glycoprotein in Trypanosoma brucei. J Cell Sci. 2004 Mar 1;117(7):1105–15. pmid:14996937
  135. 135. Fernandez-Calero T, Garcia-Silva R, Pena A, Robello C, Persson H, Rovira C, et al. Profiling of small RNA cargo of extracellular vesicles shed by Trypanosoma cruzi reveals a specific extracellular signature. Mol Biochem Parasitol. 2015 Jan;199(1–2):19–28. pmid:25795082
  136. 136. Sant’Anna C, Nakayasu ES, Pereira MG, Lourenço D, De Souza W, Almeida IC, et al. Subcellular proteomics of Trypanosoma cruzi reservosomes. Proteomics. 2009 Apr;9(7):1782–1794. pmid:19288526
  137. 137. Cavalier-Smith T. The excavate protozoan phyla Metamonada Grasse emend. (Anaeromonadea, Parabasalia, Carpediemonas, Eopharyngia) and Loukozoa emend. (Jakobea, Malawimonas): their evolutionary affinities and new higher taxa. Int J Syst Evol Microbiol [Internet]. 2003 Nov 1;53(6):1741–58. Available from: https://www.microbiologyresearch.org/content/journal/ijsem/10.1099/ijs.0.02548-0.
  138. 138. Nievas YR, Lizarraga A, Salas N, Cóceres VM, Miguel N. Extracellular vesicles released by anaerobic protozoan parasites: Current situation. Cell Microbiol [Internet]. 2020 Nov 10;22(11). Available from: https://onlinelibrary.wiley.com/doi/10.1111/cmi.13257. pmid:32858768
  139. 139. WHO. Report on global sexually transmitted infection surveillance [Internet]. Licence: C. Geneva: World Health Organization; 2018. Available from: https://www.who.int/publications/i/item/9789241565691.
  140. 140. Koehler A V., Jex AR, Haydon SR, Stevens MA, Gasser RB. Giardia/giardiasis—A perspective on diagnostic and analytical tools. Biotechnol Adv [Internet]. 2014 Mar;32(2):280–9. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0734975013001869. pmid:24189092
  141. 141. Sabatke B, Gavinho B, Coceres V, de Miguel N, Ramirez MI. Unveiling the role of EVs in anaerobic parasitic protozoa. Mol Immunol [Internet]. 2021 May;133:34–43. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0161589021000547. pmid:33621941
  142. 142. Riestra AM, de Miguel N, Dessi D, Simoes-Barbosa A, Mercer FK. Trichomonas vaginalis: Lifestyle, Cellular Biology, and Molecular Mechanisms of Pathogenesis. In: de Souza W, editor. Lifecycles of Pathogenic Protists in Humans. Microbiology Monographs, vol 35. Cham: Springer. 2022.
  143. 143. Nievas YR, Coceres VM, Midlej V, de Souza W, Benchimol M, Pereira-Neves A, et al. Membrane-shed vesicles from the parasite Trichomonas vaginalis: characterization and their association with cell interaction. Cell Mol Life Sci [Internet]. 2018 Jun 8;75(12):2211–26. Available from: http://link.springer.com/10.1007/s00018-017-2726-3. pmid:29222644
  144. 144. Twu O, de Miguel N, Lustig G, Stevens GC, Vashisht AA, Wohlschlegel JA, et al. Trichomonas vaginalis Exosomes Deliver Cargo to Host Cells and Mediate Host∶Parasite Interactions. Petri WA, editor. PLoS Pathog [Internet]. 2013 Jul 11;9(7):e1003482. Available from: https://dx.plos.org/10.1371/journal.ppat.1003482.
  145. 145. Salas N, Coceres VM, Melo T dos S, Pereira-Neves A, Maguire VG, Rodriguez TM, et al. VPS32, a member of the ESCRT complex, modulates adherence to host cells in the parasite Trichomonas vaginalis by affecting biogenesis and cargo sorting of released extracellular vesicles. Cell Mol Life Sci [Internet]. 2022 Jan 24;79(1):11. Available from: https://link.springer.com/10.1007/s00018-021-04083-3.
  146. 146. Olmos-Ortiz LM, Barajas-Mendiola MA, Barrios-Rodiles M, Castellano LE, Arias-Negrete S, Avila EE, et al. Trichomonas vaginalis exosome-like vesicles modify the cytokine profile and reduce inflammation in parasite-infected mice. Parasite Immunol [Internet]. 2017 Jun;39(6):e12426. Available from: https://onlinelibrary.wiley.com/doi/10.1111/pim.12426. pmid:28345149
  147. 147. Adam RD. Biology of Giardia lamblia. Clin Microbiol Rev [Internet]. 2001 Jul;14(3):447–75. Available from: https://journals.asm.org/doi/10.1128/CMR.14.3.447-475.2001. pmid:11432808
  148. 148. Midlej V, de Souza W, Benchimol M. The peripheral vesicles gather multivesicular bodies with different behavior during the Giardia intestinalis life cycle. J Struct Biol [Internet]. 2019 Sep;207(3):301–11. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1047847719301388. pmid:31276754
  149. 149. Gavinho B, Sabatke B, Feijoli V, Rossi IV, da Silva JM, Evans-Osses I, et al. Peptidylarginine Deiminase Inhibition Abolishes the Production of Large Extracellular Vesicles From Giardia intestinalis, Affecting Host-Pathogen Interactions by Hindering Adhesion to Host Cells. Front Cell Infect Microbiol [Internet]. 2020 Sep 23;10. Available from: https://www.frontiersin.org/article/10.3389/fcimb.2020.00417/full. pmid:33072615
  150. 150. Evans-Osses I, Mojoli A, Monguió-Tortajada M, Marcilla A, Aran V, Amorim M, et al. Microvesicles released from Giardia intestinalis disturb host-pathogen response in vitro. Eur J Cell Biol [Internet]. 2017 Mar;96(2):131–42. Available from: http://www.ncbi.nlm.nih.gov/pubmed/28236495. pmid:28236495
  151. 151. Siddiq A, Allain T, Dong G, Olivier M, Buret A. Giardia extracellular vesicles disrupt intestinal epithelial junctions and inhibit the growth of commensal bacteria while increasing their swimming motility. FASEB J [Internet]. 2020 Apr 15;34(S1):1–1. Available from: https://onlinelibrary.wiley.com/doi/10.1096/fasebj.2020.34.s1.00515.
  152. 152. Fink MY, Singer SM. The Intersection of Immune Responses, Microbiota, and Pathogenesis in Giardiasis. Trends Parasitol [Internet]. 2017 Nov;33(11):901–13. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1471492217301897. pmid:28830665
  153. 153. Abels ER, Breakefield XO. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell Mol Neurobiol [Internet]. 2016 Apr 6;36(3):301–12. Available from: http://link.springer.com/10.1007/s10571-016-0366-z. pmid:27053351
  154. 154. Wang G, Hu H-B, Chang Y, Huang Y, Song Z-Q, Zhou S-B, et al. Rab7 regulates primary cilia disassembly through cilia excision. J Cell Biol [Internet]. 2019 Dec 2;218(12):4030–41. Available from: https://rupress.org/jcb/article/218/12/4030/132511/Rab7-regulates-primary-cilia-disassembly-through. pmid:31619485
  155. 155. Saha N, Dutta S, Datta SP, Sarkar S. The minimal ESCRT machinery of Giardia lamblia has altered inter-subunit interactions within the ESCRT-II and ESCRT-III complexes. Eur J Cell Biol [Internet]. 2018 Jan;97(1):44–62. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0171933517301486. pmid:29224850
  156. 156. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, et al. Ceramide Triggers Budding of Exosome Vesicles into Multivesicular Endosomes. Science (80-) [Internet]. 2008 Feb 29;319(5867):1244–7. Available from: https://www.science.org/doi/10.1126/science.1153124. pmid:18309083
  157. 157. Pawlowski J, Burki F. Untangling the phylogeny of amoeboid protists. J Eukaryot Microbiol [Internet]. 2009 Jan;56(1):16–25. Available from: https://onlinelibrary.wiley.com/doi/10.1111/j.1550-7408.2008.00379.x. pmid:19335771
  158. 158. Adl SM, Simpson AGB, Farmer MA, Andersen RA, Anderson OR, Barta JR, et al. The new higher level classification of eukaryotes with emphasis on the taxonomy of protists. J Eukaryot Microbiol [Internet]. 2005 Oct;52(5):399–451. Available from: https://onlinelibrary.wiley.com/doi/10.1111/j.1550-7408.2005.00053.x. pmid:16248873
  159. 159. Carrero JC, Reyes-López M, Serrano-Luna J, Shibayama M, Unzueta J, León-Sicairos N, et al. Intestinal amoebiasis: 160 years of its first detection and still remains as a health problem in developing countries. Int J Med Microbiol [Internet]. 2020 Jan;310(1):151358. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1438422119303467. pmid:31587966
  160. 160. Gonçalves D, Ferreira M, Guimarães A. Extracellular Vesicles from the Protozoa Acanthamoeba castellanii: Their Role in Pathogenesis, Environmental Adaptation and Potential Applications. Bioengineering [Internet]. 2019 Feb 1;6(1):13. Available from: http://www.mdpi.com/2306-5354/6/1/13. pmid:30717103
  161. 161. Gonçalves D de S, Ferreira M da S, Liedke SC, Gomes KX, de Oliveira GA, Leão PEL, et al. Extracellular vesicles and vesicle-free secretome of the protozoa Acanthamoeba castellanii under homeostasis and nutritional stress and their damaging potential to host cells. Virulence [Internet]. 2018 Dec 31;9(1):818–36. Available from: https://www.tandfonline.com/doi/full/10.1080/21505594.2018.1451184. pmid:29560793
  162. 162. Costa AO, Chagas IAR, Menezes-Neto A, Rêgo FD, Nogueira PM, Torrecilhas AC, et al. Distinct immunomodulatory properties of extracellular vesicles released by different strains of Acanthamoeba. Cell Biol Int [Internet]. 2021 May 4;45(5):1060–71. Available from: https://onlinelibrary.wiley.com/doi/10.1002/cbin.11551. pmid:33448518
  163. 163. Toney DM, Marciano-Cabral F. Membrane vesiculation of Naegleria fowleri amoebae as a mechanism for resisting complement damage. J Immunol [Internet]. 1994 Mar 15;152(6):2952–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8144894. pmid:8144894
  164. 164. Chávez-Munguía B, Salazar Villatoro L, Omaña-Molina M, Rodríguez-Monroy MA, Segovia-Gamboa N, Martínez-Palomo A. Naegleria fowleri: Contact-dependent secretion of electrondense granules (EDG). Exp Parasitol [Internet]. 2014 Jul;142:1–6. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0014489414000873. pmid:24721258
  165. 165. Herbst R, Ott C, Jacobs T, Marti T, Marciano-Cabral F, Leippe M. Pore-forming polypeptides of the pathogenic protozoon Naegleria fowleri. J Biol Chem. 2002 Jun;277(25):22353–60. pmid:11948186
  166. 166. Fritzinger AE, Toney DM, MacLean RC, Marciano-Cabral F. Identification of a Naegleria fowleri membrane protein reactive with anti-human CD59 antibody. Infect Immun. 2006 Feb;74(2):1189–95. pmid:16428768
  167. 167. Jahangeer M, Mahmood Z, Munir N, Waraich U, Tahir IM, Akram M, et al. Naegleria fowleri : Sources of infection, pathophysiology, diagnosis, and management; a review. Clin Exp Pharmacol Physiol [Internet]. 2020 Feb 15;47(2):199–212. Available from: https://onlinelibrary.wiley.com/doi/10.1111/1440-1681.13192. pmid:31612525
  168. 168. Sharma M, Morgado P, Zhang H, Ehrenkaufer G, Manna D, Singh U. Characterization of Extracellular Vesicles from Entamoeba histolytica Identifies Roles in Intercellular Communication That Regulates Parasite Growth and Development. Saeij JPJ, editor. Infect Immun [Internet]. 2020 Sep 18;88(10). Available from: https://journals.asm.org/doi/10.1128/IAI.00349-20. pmid:32719158
  169. 169. Zysset-Burri DC, Müller N, Beuret C, Heller M, Schürch N, Gottstein B, et al. Genome-wide identification of pathogenicity factors of the free-living amoeba Naegleria fowleri. BMC Genomics [Internet]. 2014 Dec 19;15(1):496. Available from: https://bmcgenomics.biomedcentral.com/articles/10.1186/1471-2164-15-496. pmid:24950717