Acessibilidade / Reportar erro

Overview of extracellular vesicles in pathogens with special focus on human extracellular protozoan parasites

Abstract

Extracellular vesicles (EVs) are lipid-bilayered membrane-delimited particles secreted by almost any cell type, involved in different functions according to the cell of origin and its state. From these, cell to cell communication, pathogen-host interactions and modulation of the immune response have been widely studied. Moreover, these vesicles could be employed for diagnostic and therapeutic purposes, including infections produced by pathogens of diverse types; regarding parasites, the secretion, characterisation, and roles of EVs have been studied in particular cases. Moreover, the heterogeneity of EVs presents challenges at every stage of studies, which motivates research in this area. In this review, we summarise some aspects related to the secretion and roles of EVs from several groups of pathogens, with special focus on the most recent research regarding EVs secreted by extracellular protozoan parasites.

Key words:
extracellular vesicles; protozoa; parasites; infectious diseases; arthropods; helminths


Research in cell biology has been developing for years to generate structural and functional descriptions of fundamental mechanisms for life, accompanied by molecular biology techniques. The application of this knowledge in the biomedical field has been renewed with the establishment of an investigation line around extracellular vesicles (EVs). In general, EVs in mammals are described as vesicular bodies that are released from the cell through, at least, two pathways: fusion of multivesicular endosomes (MVE) with the plasma membrane (exosomes; diameter: 50 - ~ 200 nm) or direct budding from the plasma membrane (ectosomes; diameter up to 1000 nm);11. Heijnen H, Schiel AE, Fijnheer R, Geuze HJ, Sixma JJ. Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and a-granules. Blood. 1999; 94(11): 3791-9.,22. Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015; 25(6): 364-72. doi: 10.1016/j.tcb.2015.01.004.
https://doi.org/10.1016/j.tcb.2015.01.00...
however, it is impossible to claim that fractions contain only one type of vesicle.33. Mathieu M, Névo N, Jouve M, Valenzuela JI, Maurin M, Verweij FJ, et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nat Commun. 2021; 12: 4389. doi: 10.1038/s41467-021-24384-2.
https://doi.org/10.1038/s41467-021-24384...
The most studied machinery for EVs generation is the endosomal sorting complex required for transport (ESCRT) subcomplexes system, a highly conserved assembly in mammals; homolog proteins and processes have been described also in protozoa.44. Cruz Camacho A, Alfandari D, Kozela E, Regev-Rudzki N. Biogenesis of extracellular vesicles in protozoan parasites: the ESCRT complex in the trafficking fast lane? PLoS Pathog. 2023; 19(2): e1011140. doi: 10.1371/journal.ppat.1011140.
https://doi.org/10.1371/journal.ppat.101...
The biogenesis process will influence the vesicle’s content (or cargo) which, in turn, also depends on the cell of origin and the cellular microenvironment and state. In general terms, proteins, lipids, DNA, and different types of RNAs are part of the EVs cargoes.55. Abels ER, Breakefield XO. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 2016; 36(3): 301-12. doi: 10.1007/s10571-016-0366-z.
https://doi.org/10.1007/s10571-016-0366-...

The composition of EVs and, fundamentally, their proteome, is defined for the first instance by those proteins involved in their formation machinery, such as Tsg101 and Alix; in addition, proteins of the tetraspanin family (CD63, CD9, CD81) are frequently found in these vesicles and in apoptotic bodies, also considered EVs (but not addressed in this work). Other structural components are cytoskeletal proteins and glycoproteins.66. Théry C, Boussac M, Véron P, Ricciardi-Castagnoli P, Raposo G, Garin J, et al. Proteomic analysis of dendritic cell-derived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J Immunol. 2001; 166: 7309-18. doi: 10.4049/jimmunol.166.12.7309.
https://doi.org/10.4049/jimmunol.166.12....
,77. Palmisano G, Jensen SS, Le Bihan MC, Lainé J, McGuire JN, Pociot F, et al. Characterization of membrane-shed microvesicles from cytokine-stimulated ß-cells using proteomics strategies. Mol Cell Proteomics. 2012; 11(8): 230-43. doi: 10.1074/mcp.M111.012732.
https://doi.org/10.1074/mcp.M111.012732...
However, it is important to highlight that this general composition could vary depending on the cell of origin, so that characterisation analyses to evaluate the composition of EVs secreted by a specific type of cell or organism should be performed since there is a lack of universal identification methods for EVs.88. Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles. 2024; 13: e12404. doi: 10.1002/jev2.12404.
https://doi.org/10.1002/jev2.12404...

The biological role of EVs has been understood mainly as the effects they exert on acceptor cells: (i) as part of intercellular communication, through the transport of active molecules, (ii) and results on immune response and tissue repair/regeneration processes.99. Doyle LM, Wang MZ. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 2019; 8: 727. doi: 10.3390/cells8070727.
https://doi.org/10.3390/cells8070727...
Likewise, one of the major areas of global interest for the study of EVs is tumour biology, where cancer cell derived EVs have been found to fulfil a range of functions, from supporting tumour progression, angiogenesis and development of the tumour microenvironment to, conversely, anti-tumour effects from EVs of cancerous and non-cancerous origin mediated by the immune response, reflecting their heterogeneity and the need for further research.1010. Chulpanova DS, Kitaeva KV, James V, Rizvanov AA, Solovyeva VV. Therapeutic prospects of extracellular vesicles in cancer treatment. Front Immunol. 2018; 9: 1534. doi: 10.3389/fimmu.2018.01534.
https://doi.org/10.3389/fimmu.2018.01534...
,1111. Kok VC, Yu CC. Cancer-derived exosomes: their role in cancer biology and biomarker development. Int J Nanomedicine. 2020; 15: 8019-36. doi: 10.2147/IJN.S272378.
https://doi.org/10.2147/IJN.S272378...
However, in view of the description and characterisation of EVs in these pathologic conditions and various biological fluids, the idea of developing a “liquid biopsy”’ has been supported thanks to exosomal markers, and it would allow not only detection but also prognosis.1212. Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016; 126(4): 1208-15. doi: 10.1172/JCI81135.
https://doi.org/10.1172/JCI81135...
Thus, EVs have become interesting candidates: (i) for the identification of biomarkers associated with different pathological processes, (ii) as delivery platforms with EVs subjected to bioengineering procedures and iii) cell-free candidate vaccines.1313. Kodam SP, Ullah M. Diagnostic and therapeutic potential of extracellular vesicles. Technol Cancer Res Treat. 2021; 20: 1-10. doi: 10.1177/15330338211041203.
https://doi.org/10.1177/1533033821104120...

Although the clinical utility of EVs in understanding and approaching malignancies such as cancer has been of great interest, their role in infectious processes is another line to be exploited as they are involved as messengers of the immune response and inflammatory processes, with cytokine stimulation, antigen presentation through major histocompatibility complex (MHC)-I and MHC-II, and activation of T and B cells,1414. Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep. 2015; 16(1): 24-43. doi: 10.15252/embr.201439363.
https://doi.org/10.15252/embr.201439363...
as well as their ability to act as key molecule carriers and potential cellular intercommunicators.1515. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013; 200(4): 373-83. doi: 10.1083/jcb.201211138.
https://doi.org/10.1083/jcb.201211138...
The isolation of EVs has been carried out from multiple pathogenic microorganisms (PEVs) and their composition and activity is varied from case to case, even within the same taxonomic genus, although the animal model, the experimental design and the cell type assayed should be associated with this variation.1414. Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep. 2015; 16(1): 24-43. doi: 10.15252/embr.201439363.
https://doi.org/10.15252/embr.201439363...
In this sense, a variety of reviews addressing EVs from pathogens have been published.1616. Bielska E, May RC. Extracellular vesicles of human pathogenic fungi. Curr Opin Microbiol. 2019; 52: 90-9. doi: 10.1016/j.mib.2019.05.007.
https://doi.org/10.1016/j.mib.2019.05.00...
,1717. Kim JH, Lee J, Park J, Gho YS. Gram-negative and Gram-positive bacterial extracellular vesicles. Semin Cell Dev Biol. 2015; 40: 97-104. doi: 10.1016/j.semcdb.2015.02.006.
https://doi.org/10.1016/j.semcdb.2015.02...
,1818. Urbanelli L, Buratta S, Tancini B, Sagini K, Delo F, Porcellati S, et al. The role of extracellular vesicles in viral infection and transmission. Vaccines. 2019; 7: 102. doi: 10.3390/vaccines7030102.
https://doi.org/10.3390/vaccines7030102...
,1919. Carrera-Bravo C, Koh EY, Tan KSW. The roles of parasite-derived extracellular vesicles in disease and host-parasite communication. Parasitol Int. 2021; 83: 102373. doi: 10.1016/j.parint.2021.102373.
https://doi.org/10.1016/j.parint.2021.10...

Microbial pathogenesis is also a studied process in which PEVs participate by harbouring genes and virulence factors, toxins and molecules for coordination and communication between pathogens.2020. Ofir-Birin Y, Heidenreich M, Regev-Rudzki N. Pathogen-derived extracellular vesicles coordinate social behaviour and host manipulation. Semin Cell Dev Biol. 2017; 67: 83-90. doi: 10.1016/j.semcdb.2017.03.004.
https://doi.org/10.1016/j.semcdb.2017.03...
PEVs also have a role in the pathogen-host interplay, by manipulating or interfering with the immune response or cellular specific cascades due to RNA signalling molecules, protein ligands or pathogen-associated molecular patterns (PAMPS).2121. Duncan L, Yoshioka M, Chandad F, Grenier D. Loss of lipopolysaccharide receptor CD14 from the surface of human macrophage-like cells mediated by Porphyromonas gingivalis outer membrane vesicles. Microb Pathog. 2004; 36: 319-25. doi: 10.1016/j.micpath.2004.02.004.
https://doi.org/10.1016/j.micpath.2004.0...
,2222. Bhatnagar S, Shinagawa K, Castellino FJ, Schorey JS. Exosomes released from macrophages infected with intracellular pathogens stimulate a proinflammatory response in vitro and in vivo. Blood. 2007; 110(9): 3234-44. doi: 10.1182/blood-2007-03-079152.
https://doi.org/10.1182/blood-2007-03-07...
,2323. 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. 2014; 193(2): 71-4. doi: 10.1016/j.molbiopara.2014.02.004.
https://doi.org/10.1016/j.molbiopara.201...
On the other hand, many of these organisms (bacteria, fungi, parasites, viruses) have also completely intracellular stages, phases, or life cycles in which they can hijack the endosomal machinery of the invaded cells to modify or alter EVs trafficking, induced, in this case, by the pathogen.2424. Cipriano MJ, Hajduk SL. Drivers of persistent infection: pathogen-induced extracellular vesicles. Essays Biochem. 2018; 62: 135-47. doi: 10.1042/EBC20170083.
https://doi.org/10.1042/EBC20170083...

The group of medically important parasites comprises agents, mainly protozoans, with complex life cycles, involving invasive and non-invasive evolutionary stages. The role of extracellular vesicles in influencing parasitosis could have several edges: from functions of adaptation to the host environment and effects on the pathogen’s infectivity, to involvement in invasion signalling and immune modulation.2525. 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. 2014; 3: 25040. doi: 10.3402/jev.v3.25040.
https://doi.org/10.3402/jev.v3.25040...
In this review, a brief overview of the main findings in the EVs field, in relation to parasites (with special attention to extracellular protozoa) is presented.

EVs in microbiology

During infections, EVs have not fully deciphered or described roles, and the extent to which they contribute to pathogen establishment is a topic to be exploited in different areas of microbiology and cell physiology. In microbiology, the secretion of EVs from some types of viruses, bacteria, fungi, and parasites has been described and extensively studied in some cases. As it has been reported elsewhere and it’s not the main objective of the review, only a brief description of highlights regarding EVs of non-parasitic origin will be presented.

EVs can be aroused from virus-infected cells. In these cases, EVs can also carry viral elements, such as proteins or receptors that make the acceptor cell more susceptible to infection, as described for human immunodeficiency virus (HIV),2626. Arenaccio C, Anticoli S, Manfredi F, Chiozzini C, Olivetta E, Federico M. Latent HIV-1 is activated by exosomes from cells infected with either replication-competent or defective HIV-1. Retrovirology. 2015; 12: 87. doi: 10.1186/s12977-015-0216-y.
https://doi.org/10.1186/s12977-015-0216-...
,2727. Mack M, Kleinschmidt A, Bruhl H, Klier C, Nelson P, Cihak J, et al. Transfer of the chemokine receptor CCR5 between cells by membrane-derived microparticles: a mechanism for cellular human immunodeficiency virus 1 infection. Nat Med. 2000; 6(7): 769-75. and similar to the transference of CD9 and ACE2 receptors that has been recently proposed for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19).2828. Hassanpour M, Rezaie J, Nouri M, Panahi Y. The role of extracellular vesicles in COVID-19 virus infection. Infect Genet Evol. 2020; 85: 104422. doi: 10.1016/j.meegid.2020.104422.
https://doi.org/10.1016/j.meegid.2020.10...
In addition, viruses such as hepatitis C can use EVs as machinery to infect cells via viral RNA and achieve replication without relying on the virion or viral receptors.2929. Bukong TN, Momen-Heravi F, Kodys K, Bala S, Szabo G. Exosomes from Hepatitis C infected patients transmit HCV infection and contain replication competent viral RNA in complex with Ago2-miR122-HSP90. PLoS Pathog. 2014; 10(10): e1004424. doi: 10.1371/journal.ppat.1004424.
https://doi.org/10.1371/journal.ppat.100...
,3030. Longatti A, Boyd B, Chisari FV. Virion-independent transfer of replication-competent Hepatitis C virus RNA between permissive cells. J Virol. 2015; 89(5): 2956-61. doi: 10.1128/jvi.02721-14.
https://doi.org/10.1128/jvi.02721-14...
In this sense, it could be suggested a dual functionality, as they also represent a means to trigger antiviral responses by the activation of adaptive immunity via viral antigens and molecular effectors. However, a major methodological limitation is the complexity involved in separating viral particles from the EVs to be assayed.1818. Urbanelli L, Buratta S, Tancini B, Sagini K, Delo F, Porcellati S, et al. The role of extracellular vesicles in viral infection and transmission. Vaccines. 2019; 7: 102. doi: 10.3390/vaccines7030102.
https://doi.org/10.3390/vaccines7030102...

EVs of prokaryotic origin have been found not only in in vitro cultures, but also in in vivo cultures and even from environmental samples.3131. Kim YS, Choi EJ, Lee WH, Choi SJ, Roh TY, Park J, et al. Extracellular vesicles, especially derived from Gram-negative bacteria, in indoor dust induce neutrophilic pulmonary inflammation associated with both Th1 and Th17 cell responses. Clin Exp Allergy. 2013; 43: 443-54. doi: 10.1111/cea.12085.
https://doi.org/10.1111/cea.12085...
,3232. Yang D, Chen X, Wang J, Lou Q, Lou Y, Li L, et al. Dysregulated lung commensal bacteria drive interleukin-17B production to promote pulmonary fibrosis through their outer membrane vesicles. Immunity. 2019; 50(3): 692-706. doi: 10.1016/j.immuni.2019.02.001.
https://doi.org/10.1016/j.immuni.2019.02...
These are essentially the same bilayered particle as in eukaryotic cells, as they correspond to membranous “capsules” released by a cell into the extracellular space, but there are fundamental structural differences given by the conformation of the cell envelope. For instance, in gram-negative bacteria, EVs are rich in lipopolysaccharides, although there are generally cargoes common to all bacteria such as proteins involved in metabolic pathways and genetic material.1717. Kim JH, Lee J, Park J, Gho YS. Gram-negative and Gram-positive bacterial extracellular vesicles. Semin Cell Dev Biol. 2015; 40: 97-104. doi: 10.1016/j.semcdb.2015.02.006.
https://doi.org/10.1016/j.semcdb.2015.02...
,3333. Briaud P, Carroll RK. Extracellular vesicle biogenesis and functions in gram-positive bacteria. Infect Immun. 2020; 8(12): e00433-20. doi: 10.1128/IAI.00433-20.
https://doi.org/10.1128/IAI.00433-20...
In bacteria, the formation and secretion of the so-called outer membrane vesicles (OMVs) is given globally by gene regulation3434. Kulp AJ, Sun B, Ai T, Manning AJ, Orench-Rivera N, Schmid AK, et al. Genome-wide assessment of outer membrane vesicle production in Escherichia coli. PLoS One. 2015; 10(9): e0139200. doi: 10.1371/journal.pone.0139200.
https://doi.org/10.1371/journal.pone.013...
,3535. Nevermann J, Silva A, Otero C, Oyarzún DP, Barrera B, Gil F, et al. Identification of genes involved in biogenesis of outer membrane vesicles (OMVs) in Salmonella enterica Serovar Typhi. Front Microbiol. 2019; 10: 104. doi: 10.3389/fmicb.2019.00104.
https://doi.org/10.3389/fmicb.2019.00104...
and different approaches such as: (i) a particular distribution of phospholipids in a membrane region, or (ii) the accumulation of molecules in the periplasmic space and the consequent turgor pressure and interaction between negative charges, which promote plasma membrane’s curvature.3636. Roier S, Zingl FG, Cakar F, Durakovic S, Kohl P, Eichmann TO, et al. A novel mechanism for the biogenesis of outer membrane vesicles in Gram-negative bacteria. Nat Commun. 2016; 7: 10515. doi: 10.1038/ncomms10515.
https://doi.org/10.1038/ncomms10515...
,3737. Schlatterer K, Beck C, Hanzelmann D, Lebtig M, Fehrenbacher B, Schaller M, et al. The mechanism behind bacterial lipoprotein release: Phenol-soluble modulins mediate toll-like receptor 2 activation via extracellular vesicle release from Staphylococcus aureus. mBio. 2018; 9(6): e01851-18. doi: 10.1128/mbio.01851-18.
https://doi.org/10.1128/mbio.01851-18...
Likewise, the release could be related to compromises in membrane stability, where there are lytic effectors leading to disruption of the peptidoglycan wall.3838. Turnbull L, Toyofuku M, Hynen AL, Kurosawa M, Pessi G, Petty NK, et al. Explosive cell lysis as a mechanism for the biogenesis of bacterial membrane vesicles and biofilms. Nat Commun. 2016; 7: 11220. doi: 10.1038/ncomms11220.
https://doi.org/10.1038/ncomms11220...
,3939. Wang X, Thompson CD, Weidenmaier C, Lee JC. Release of Staphylococcus aureus extracellular vesicles and their application as a vaccine platform. Nat Commun. 2018; 9: 1379. doi: 10.1038/s41467-018-03847-z.
https://doi.org/10.1038/s41467-018-03847...

Regarding their physiology and clinical relevance, OMVs have been involved not only in bacterial communication processes, horizontal gene transfer and influence on the microenvironment,1717. Kim JH, Lee J, Park J, Gho YS. Gram-negative and Gram-positive bacterial extracellular vesicles. Semin Cell Dev Biol. 2015; 40: 97-104. doi: 10.1016/j.semcdb.2015.02.006.
https://doi.org/10.1016/j.semcdb.2015.02...
but also their cargo has been employed as an arsenal to interact with the host; in fact, TLRs have been involved in interactions of EVs with mammalian target cells4040. Schaar V, De Vries SPW, Perez Vidakovics MLA, Bootsma HJ, Larsson L, Hermans PWM, et al. Multicomponent Moraxella catarrhalis outer membrane vesicles induce an inflammatory response and are internalized by human epithelial cells. Cell Microbiol. 2011; 13(3): 432-49. doi: 10.1111/j.1462-5822.2010.01546.x.
https://doi.org/10.1111/j.1462-5822.2010...
,4141. Van Bergenhenegouwen J, Kraneveld AD, Rutten L, Kettelarij N, Garssen J, Vos AP. Extracellular vesicles modulate host-microbe responses by altering TLR2 activity and phagocytosis. PLoS One. 2014; 9(2): e89121. doi: 10.1371/journal.pone.0089121.
https://doi.org/10.1371/journal.pone.008...
and their content may include virulence factors with cytotoxic and antibiotic resistance effect.4242. Ciofu O, Beveridge TJ, Kadurugamuwa J, Walther-Rasmussen J, Høiby N. Chromosomal ß-lactamase is packaged into membrane vesicles and secreted from Pseudomonas aeruginosa. J Antimicrob Chemother. 2000; 45: 9-13.,4343. Rivera J, Cordero RJB, Nakouzi AS, Frases S, Nicola A, Casadevall A. Bacillus anthracis produces membrane-derived vesicles containing biologically active toxins. Proc Natl Acad Sci USA. 2010; 107(44): 19002-7. doi: 10.1073/pnas.1008843107.
https://doi.org/10.1073/pnas.1008843107...
,4444. Vanaja SK, Russo AJ, Behl B, Banerjee I, Yankova M, Deshmukh SD, et al. Bacterial outer membrane vesicles mediate cytosolic localization of LPS and Caspase-11 activation. Cell. 2016; 165: 1106-19. doi: 10.1016/j.cell.2016.04.015.
https://doi.org/10.1016/j.cell.2016.04.0...
On the other hand, regulation of signalling pathways leading to immunomodulation has been determined in bacteria of the oral and intestinal microbiota.4545. Ñahui Palomino RA, Vanpouille C, Costantini PE, Margolis L. Microbiota-host communications: bacterial extracellular vesicles as a common language. PLoS Pathog. 2021; 17(5): e1009508. doi: 10.1371/journal.ppat.1009508.
https://doi.org/10.1371/journal.ppat.100...
A recent study with cervicovaginal pathobionts and commensal bacteria EVs indicate differential cargo and viability/cytoadherence effects when evaluating them onto a culture model with ectocervical cells and Trichomonas vaginalis, showing a possible role in host-pathogen interaction.4646. Artuyants A, Campos TL, Rai AK, Johnson PJ, Dauros-Singorenko P, Phillips A, et al. Extracellular vesicles produced by the protozoan parasite Trichomonas vaginalis contain a preferential cargo of tRNA-derived small RNAs. Int J Parasitol. 2020; 50: 1145-55. doi: 10.1016/j.ijpara.2020.07.003.
https://doi.org/10.1016/j.ijpara.2020.07...

The study of EVs in this type of pathogens has been virtuous: Escherichia coli, Moraxella catarrhalis or Pseudomonas spp. have been subjects of research, as well as the gram-positive Bacillus subtilis and Staphylococcus aureus.4747. Brown L, Wolf JM, Prados-Rosales R, Casadevall A. Through the wall: extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi. Nat Rev Microbiol. 2015; 13(10): 620-30. doi: 10.1038/nrmicro3480.
https://doi.org/10.1038/nrmicro3480...
Actually, licensed OMVs based vaccines against meningococcal infections have been developed and, in this sense, advanced discovery is currently exploring on enteric pathogens.4848. Lieberman LA. Outer membrane vesicles: a bacterial-derived vaccination system. Front Microbiol. 2022; 13: 1029146. doi: 10.3389/fmicb.2022.1029146.
https://doi.org/10.3389/fmicb.2022.10291...
Besides, and thanks to electron microscopy, EVs have also been described in mycobacteria and fungi of medical importance.4747. Brown L, Wolf JM, Prados-Rosales R, Casadevall A. Through the wall: extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi. Nat Rev Microbiol. 2015; 13(10): 620-30. doi: 10.1038/nrmicro3480.
https://doi.org/10.1038/nrmicro3480...
Particularly, in the latter, it should be noted that their secretion has been described in yeasts such as Cryptococcus neoformans,4949. Rodrigues ML, Nimrichter L, Oliveira DL, Frases S, Miranda K, Zaragoza O, et al. Vesicular polysaccharide export in Cryptococcus neoformans is a eukaryotic solution to the problem of fungal trans-cell wall transport. Eukaryot Cell. 2007; 6(1): 48-59. doi: 10.1128/EC.00318-06.
https://doi.org/10.1128/EC.00318-06...
as well as in filamentous fungi such as Sporothrix brasiliensis.5050. Ikeda MAK, De Almeida JRF, Jannuzzi GP, Cronemberger-Andrade A, Torrecilhas ACT, Moretti NS, et al. 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. doi: 10.3389/fmicb.2018.02286.
https://doi.org/10.3389/fmicb.2018.02286...

EVs in fungi, as eukaryotic organisms, share release mechanisms with those described in mammals, as they appear to be linked to the endocytic secretory pathways associated with the ESCRT and Golgi reassembly and stacking proteins (GRASP) machinery, and the ER-GA-exocyst-PM axis.5151. Liebana-Jordan M, Brotons B, Falcon-Perez JM, Gonzalez E. Extracellular vesicles in the fungi kingdom. Int J Mol Sci. 2021; 22: 7221. doi: 10.3390/ijms22137221.
https://doi.org/10.3390/ijms22137221...
Likewise, their size ranges from 20 - 50 nm to 1000 nm, according to different reports and under different methodologies such as dynamic light scattering (DLS), electron microscopy (EM) and nanoparticle tracking analysis (NTA).1616. Bielska E, May RC. Extracellular vesicles of human pathogenic fungi. Curr Opin Microbiol. 2019; 52: 90-9. doi: 10.1016/j.mib.2019.05.007.
https://doi.org/10.1016/j.mib.2019.05.00...
Other vesicles studied in this group have been the periplasmic vesicles, which are those inside the fungal cell wall, between the cell membrane and the inner face of the chitin barrier,5151. Liebana-Jordan M, Brotons B, Falcon-Perez JM, Gonzalez E. Extracellular vesicles in the fungi kingdom. Int J Mol Sci. 2021; 22: 7221. doi: 10.3390/ijms22137221.
https://doi.org/10.3390/ijms22137221...
but also EVs from protoplasmic models have helped to understand fungal vesicles roles.5252. Rizzo J, Chaze T, Miranda K, Roberson RW, Gorgette O, Nimrichter L, et al. Characterization of extracellular vesicles produced by Aspergillus fumigatus protoplasts. mSphere. 2020; 5(4): e00476-20. doi: 10.1128/msphere.00476-20.
https://doi.org/10.1128/msphere.00476-20...
Finally, in addition to pathogenic related functions, cell wall-remodelling enzymes for easier vesicle passage and immunogenic protein content have been found in EVs of Histoplasma caspsulatum;5353. Albuquerque PC, Nakayasu ES, Rodrigues ML, Frases S, Casadevall A, Zancope-Oliveira RM, et al. Vesicular transport in Histoplasma capsulatum: an effective mechanism for trans-cell wall transfer of proteins and lipids in ascomycetes. Cell Microbiol. 2008; 10(8): 1695-710. doi: 10.1111/j.1462-5822.2008.01160.x.
https://doi.org/10.1111/j.1462-5822.2008...
also, fungal EVs are suspected to be involved in modulation of immune effectors and in cryptococcosis and sporotrichosis outcome due to virulence enhancement.5454. Piffer AC, Kuczera D, Rodrigues ML, Nimrichter L. The paradoxical and still obscure properties of fungal extracellular vesicles. Mol Immunol. 2021; 135: 137-46. doi: 10.1016/j.molimm.2021.04.009.
https://doi.org/10.1016/j.molimm.2021.04...

Important findings in EVs for clinical parasitology

The release of EVs in the context of a parasitic infection becomes complex as one has those produced by the host and by the parasite.1414. Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep. 2015; 16(1): 24-43. doi: 10.15252/embr.201439363.
https://doi.org/10.15252/embr.201439363...
In fact, it has been proposed that the fusion between the EVs of protozoan parasites and those of the host cell could have effects on any of the involved; that’s why the understanding the participation of EVs in host-parasite interaction and cell communication would probably redefine the concepts of parasitism.5555. Evans-Osses I, Reichembach LH, Ramirez MI. Exosomes or microvesicles? Two kinds of extracellular vesicles with different routes to modify protozoan-host cell interaction. Parasitol Res. 2015; 114: 3567-75. doi: 10.1007/s00436-015-4659-9.
https://doi.org/10.1007/s00436-015-4659-...
Furthermore, the dissemination of genetic elements of the parasite through EVs supports their possible involvement in co-adaptative and co-evolutionary processes of gene regulation and synchronisation with the host metabolism.5656. Barteneva NS, Maltsev N, Vorobjev IA. Microvesicles and intercellular communication in the context of parasitism. Front Cell Infect Microbiol. 2013; 3: 49. doi: 10.3389/fcimb.2013.00049.
https://doi.org/10.3389/fcimb.2013.00049...
The role as strong parasite-parasite communication messengers and further effects on this regard, as motility/migration signals in African trypanosomes has been demonstrated,5757. 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; 13(3): e1006245. doi: 10.1371/journal.ppat.1006245.
https://doi.org/10.1371/journal.ppat.100...
cargo manipulation and functional small RNA roles in PEVs are advancing areas in this research field.5858. Sharma M, Lozano-Amado D, Chowdhury D, Singh U. Extracellular Vesicles and Their impact on the biology of protozoan parasites. Trop Med Infect Dis. 2023: 8: 448. doi: 10.3390/tropicalmed8090448.
https://doi.org/10.3390/tropicalmed80904...

The proteome and transcriptome of parasitic EVs reveals the presence of molecules associated not only to immunomodulation, but also to reproduction and survival, so that any subsequent discovery in relation to their functions would give rise to new ways of understanding pathogenesis, how parasite-host communication occurs, and the study of new drug targets.5959. Nawaz M, Malik MI, Hameed M, Zhou J. Research progress on the composition and function of parasite-derived exosomes. Acta Trop. 2019; 196: 30-6. doi: 10.1016/j.actatropica.2019.05.004.
https://doi.org/10.1016/j.actatropica.20...
Even diagnostic applications are already on the horizon, as proposed by Wang et al.,6060. Wang Y, Yuan W, Kimber M, Lu M, Dong L. Rapid differentiation of host and parasitic exosome vesicles using microfluidic photonic crystal biosensor. ACS Sens. 2018; 3: 1616-21. doi: 10.1021/acssensors.8b00360.
https://doi.org/10.1021/acssensors.8b003...
who worked in the development of a biosensor to discriminate between EVs from Ascaris suum and those from mice macrophages, where the differential binding of these EVs through a specific marker (CD63), absent in the parasite EVs, causes a shift in the wavelength resonance;6060. Wang Y, Yuan W, Kimber M, Lu M, Dong L. Rapid differentiation of host and parasitic exosome vesicles using microfluidic photonic crystal biosensor. ACS Sens. 2018; 3: 1616-21. doi: 10.1021/acssensors.8b00360.
https://doi.org/10.1021/acssensors.8b003...
even though, each potential diagnostic tool should be carefully validated due to, for example, the possibility of finding other tetraspanins in parasite-derived EVs.6161. Kifle DW, Chaiyadet S, Waardenberg AJ, Wise I, Cooper M, Becker L, et al. Uptake of Schistosoma mansoni extracellular vesicles by human endothelial and monocytic cell lines and impact on vascular endothelial cell gene expression. Int J Parasitol. 2020; 50: 685-96. doi: 10.1016/j.ijpara.2020.05.005.
https://doi.org/10.1016/j.ijpara.2020.05...
,6262. Chaiyadet S, Sotillo J, Krueajampa W, Thongsen S, Smout M, Brindley PJ, et al. Silencing of Opisthorchis viverrini tetraspanin gene expression results in reduced secretion of extracellular vesicles. Front Cell Infect Microbiol. 2022; 12: 827521. doi: 10.3389/fcimb.2022.827521.
https://doi.org/10.3389/fcimb.2022.82752...
For this reason, the proper characterisation of the content of EVs is relevant.

During the different forms of parasitism, PEVs derived from extracellular parasites could be found, but also those secreted from intracellular infected cells and parasitic antigen stimulated cells.6363. Wu Z, Wang L, Li J, Wang L, Wu Z, Sun X. Extracellular vesicle-mediated communication within host-parasite interactions. Front Immunol. 2019; 9: 3066. doi: 10.3389/fimmu.2018.03066.
https://doi.org/10.3389/fimmu.2018.03066...

Macro-extracellular parasites: arthropods and helminths

Before delving into protozoan parasites, it is worthwhile to review what has been identified in other groups of classical parasitology, such as arthropods that act as biological vectors and helminths of medical importance, with specific cases of their EVs pivot findings.

EVs of some arthropods have been implicated in the dissemination/infection process of the microorganisms they transmit,6464. Sultana H, Neelakanta G. Arthropod exosomes as bubbles with message(s) to transmit vector-borne diseases. Curr Opin Insect Sci. 2020; 40: 39-47. doi: 10.1016/j.cois.2020.05.017.
https://doi.org/10.1016/j.cois.2020.05.0...
but also as part of the vector-host-pathogen triad.6565. Hackenberg M, Kotsyfakis M. Exosome-mediated pathogen transmission by arthropod vectors. Trends Parasitol. 2018; 34(7): 549-52. doi: 10.1016/j.pt.2018.04.001.
https://doi.org/10.1016/j.pt.2018.04.001...
In this sense, these vesicles have been described as possible mediators in the transmission of flavivirus proteins and RNA, as demonstrated by Zhou et al.6666. Zhou W, Woodson M, Neupane B, Bai F, Sherman MB, Choi KH, et al. Exosomes serve as novel modes of tick-borne flavivirus transmission from arthropod to human cells and facilitates dissemination of viral RNA and proteins to the vertebrate neuronal cells. PLoS Pathog. 2018; 14(1): e1006764. doi: 10.1371/journal.ppat.1006764.
https://doi.org/10.1371/journal.ppat.100...
in their in vitro model with an Ixodes scapularis cell line infected with langat virus (LGTV) and human keratinocytes/endothelial cells. The same was demonstrated in cell lines derived from Aedes aegypti and Ae. albopictus mosquitoes with dengue virus type 2 (DENV2) viral particles.6767. Vora A, Zhou W, Londono-Renteria B, Woodson M, Sherman MB, Colpitts TM, et al. Arthropod EVs mediate dengue virus transmission through interaction with a tetraspanin domain containing glycoprotein Tsp29Fb. Proc Natl Acad Sci USA. 2018; 115(28): E6604-13. doi: 10.1073/pnas.1720125115.
https://doi.org/10.1073/pnas.1720125115...
Besides, viral-like particles have been observed in extracellular vesicles derived from DENV infected C6/36 cells.6868. Reyes-Ruiz JM, Osuna-Ramos JF, De Jesús-González LA, Hurtado-Monzón AM, Farfan-Morales CN, Cervantes-Salazar M, et al. Isolation and characterization of exosomes released from mosquito cells infected with dengue virus. Virus Res. 2019; 266: 1-14. doi: 10.1016/j.virusres.2019.03.015.
https://doi.org/10.1016/j.virusres.2019....

In addition, Oliva Chávez et al.6969. Oliva Chávez AS, Wang X, Marnin L, Archer NK, Hammond HL, Carroll EEMC, et al. Tick extracellular vesicles enable arthropod feeding and promote distinct outcomes of bacterial infection. Nat Commun. 2021; 12: 3696. doi: 10.1038/s41467-021-23900-8.
https://doi.org/10.1038/s41467-021-23900...
demonstrated an impaired feeding ability of I. scapularis by silencing genes of soluble NSF (N-ethylmaleimide-sensitive fusion protein) receptor (SNARE) molecules (vamp33 and synaptobrevin 2) related to the release of EVs, in parallel to an increase of γδ-T cells at the site of the bite. In turn, these EVs present in tick saliva might play roles not only in tick-borne pathogens transmission dynamics, as it has been seen for protozoan parasites and bacteria, but also in feeding-facilitating immunomodulatory responses at the ectoparasite-host skin interplay.7070. Butler LR, Gonzalez J, Pedra JHF, Chavez ASO. Tick extracellular vesicles in host skin immunity and pathogen transmission. Trends Parasitol. 2023; 39(10): 873-85. doi: 10.1016/j.pt.2023.07.009.
https://doi.org/10.1016/j.pt.2023.07.009...
Moreover, the vector as an arthropod host could be affected by microbial EVs, as the case of the regulation of the innate immune response of Ae. aegypti by EVs of microfilariae.7171. Loghry HJ, Kwon H, Smith RC, Sondjaja NA, Minkler SJ, Young S, et al. Extracellular vesicles secreted by Brugia malayi microfilariae modulate the melanization pathway in the mosquito host. Sci Rep. 2023; 13: 8778. doi: 10.1038/s41598-023-35940-9.
https://doi.org/10.1038/s41598-023-35940...
Of course, some non-vector free living arthropods like dust mites have been implicated in other types of human damage such as allergic processes and, for instance, Dermatophagoides farinae EVs were shown to be immunoreactive against specific serum IgE and to induce airway inflammation in mice.7272. Yang T, Xu Z, Yu J, Liu J, Wang W, Hong S. Exosomes derived from Dermatophagoides farinae induce allergic airway inflammation. Microbiol Spectr. 2023; 11(4): 1-19. doi: 10.1128/spectrum.05054-22.
https://doi.org/10.1128/spectrum.05054-2...

In the helminths group, EVs, as part of the excretory-secretory products, have been studied from different perspectives, such as in Trichuris muris, for pathogenesis understanding purposes using organoids,7373. Duque-Correa MA, Schreiber F, Rodgers FH, Goulding D, Forrest S, White R, et al. Development of caecaloids to study host-pathogen interactions: new insights into immunoregulatory functions of Trichuris muris extracellular vesicles in the caecum. Int J Parasitol. 2020; 50: 707-18. doi: 10.1016/j.ijpara.2020.06.001.
https://doi.org/10.1016/j.ijpara.2020.06...
or in Fasciola hepatica and Brugia malayi, where proteomics and inmunological-based visualisation techniques have been instrumentalised to elucidate the biogenic pathways and cellular origin of vesicles.7474. Harischandra H, Yuan W, Loghry HJ, Zamanian M, Kimber MJ. Profiling extracellular vesicle release by the filarial nematode Brugia malayi reveals sex-specific differences in cargo and a sensitivity to ivermectin. PLoS Negl Trop Dis. 2018; 12(4): e0006438. doi: 10.1371/journal.pntd.0006438.
https://doi.org/10.1371/journal.pntd.000...
,7575. de la Torre-Escudero E, Gerlach JQ, Bennett APS, Cwiklinski K, Jewhurst HL, Huson KM, et al. Surface molecules of extracellular vesicles secreted by the helminth pathogen Fasciola hepatica direct their internalisation by host cells. PLoS Negl Trop Dis. 2019; 13(1): e0007087. doi: 10.1371/journal.pntd.0007087.
https://doi.org/10.1371/journal.pntd.000...
The participation of carbohydrates in lectin-EVs binding patterns and macrophage internalisation has also been evidenced,7575. de la Torre-Escudero E, Gerlach JQ, Bennett APS, Cwiklinski K, Jewhurst HL, Huson KM, et al. Surface molecules of extracellular vesicles secreted by the helminth pathogen Fasciola hepatica direct their internalisation by host cells. PLoS Negl Trop Dis. 2019; 13(1): e0007087. doi: 10.1371/journal.pntd.0007087.
https://doi.org/10.1371/journal.pntd.000...
as well as the description of virulence factors in EVs from Paragonimus kellicotti lung cyst fluid7676. Di Maggio LS, Fischer K, Yates D, Curtis KC, Rosa BA, Martin J, et al. The proteome of extracellular vesicles of the lung fluke Paragonimus kellicotti produced in vitro and in the lung cyst. Sci Rep. 2023; 13: 13726. doi: 10.1038/s41598-023-39966-x.
https://doi.org/10.1038/s41598-023-39966...
and Echinococcus multilocularis protoescoleces.7777. Liu C, Cao J, Zhang H, Field MC, Yin J. Extracellular vesicles secreted by Echinococcus multilocularis: important players in angiogenesis promotion. Microbes Infect. 2023; 25: 105147. doi: 10.1016/j.micinf.2023.105147.
https://doi.org/10.1016/j.micinf.2023.10...
Moreover, varied functional-immune assays have revealed the immunomodulatory capacity of Trichinella spiralis EVs,7878. Kosanovic M, Cvetkovic J, Gruden-Movsesijan A, Vasilev S, Svetlana M, Ilic N, et al. Trichinella spiralis muscle larvae release extracellular vesicles with immunomodulatory properties. Parasite Immunol. 2019; 41: e12665. doi: 10.1111/pim.12665.
https://doi.org/10.1111/pim.12665...
the phenotypic modification of dendritic cells and the reduction of macrophages migratory capacity induced by EVs from the trematode F. hepatica7979. Murphy A, Cwiklinski K, Lalor R, O'connell B, Robinson MW, Gerlach J, et al. Fasciola hepatica extracellular vesicles isolated from excretory-secretory products using a gravity flow method modulate dendritic cell phenotype and activity. PLoS Negl Trop Dis. 2020; 14(9): e0008626. doi: 10.1371/journal.pntd.0008626.
https://doi.org/10.1371/journal.pntd.000...
,8080. Sánchez-López CM, González-Arce A, Soler C, Ramírez-Toledo V, Trelis M, Bernal D, et al. Extracellular vesicles from the trematodes Fasciola hepatica and Dicrocoelium dendriticum trigger different responses in human THP-1 macrophages. J Extracell Vesicles. 2023; 12(4): e12317. doi: 10.1002/jev2.12317.
https://doi.org/10.1002/jev2.12317...
and the expression of miRNAs associated with the mTOR signalling pathway as part of the cargo in EVs from filarial nematodes,8181. Ricciardi A, Bennuru S, Tariq S, Kaur S, Wu W, Elkahloun AG, et al. Extracellular vesicles released from the filarial parasite Brugia malayi downregulate the host mTOR pathway. PLoS Negl Trop Dis. 2021; 15(1): e0008884. doi: 10.1371/journal.pntd.0008884.
https://doi.org/10.1371/journal.pntd.000...
which all supports a potential for downregulation. This type of active biomolecules (miRNAs) derived from EVs are part of the developing diagnostic arsenal, as they could be identified from biological samples and have been achieved from serum of Schistosoma spp. infected patients.8282. Meningher T, Lerman G, Regev-Rudzki N, Gold D, Ben-Dov IZ, Sidi Y, et al. Schistosomal microRNAs isolated from extracellular vesicles in sera of infected patients: a new tool for diagnosis and follow-up of human schistosomiasis. J Infect Dis. 2017; 215: 378-86. doi: 10.1093/infdis/jiw539.
https://doi.org/10.1093/infdis/jiw539...
Computational prediction of miARN found in EVs from different nematodes support their immunological relevance as immune networks genes are targeted by these molecules.8383. Eichenberger RM, Talukder MH, Field MA, Wangchuk P, Giacomin P, Loukas A, et al. Characterization of Trichuris muris secreted proteins and extracellular vesicles provides new insights into host-parasite communication. J Extracell Vesicles. 2018; 7: 1428004. doi: 10.1080/20013078.2018.1428004.
https://doi.org/10.1080/20013078.2018.14...
,8484. Eichenberger RM, Ryan S, Jones L, Buitrago G, Polster R, de Oca MM, et al. Hookworm secreted extracellular vesicles interact with host cells and prevent inducible colitis in mice. Front Immunol. 2018; 9: 850. doi: 10.3389/fimmu.2018.00850.
https://doi.org/10.3389/fimmu.2018.00850...
,8585. Hansen EP, Fromm B, Andersen SD, Marcilla A, Andersen KL, Borup A, et al. Exploration of extracellular vesicles from Ascaris suum provides evidence of parasite-host cross talk. J Extracell Vesicles. 2019; 8: 1578116. doi: 10.1080/20013078.2019.1578116.
https://doi.org/10.1080/20013078.2019.15...
For helminthiasis, as in other microorganisms, the production of vaccines based on EVs and their antigens is still an interesting proposal that gives new routes for resolving doubts about antigen expression control and its variability, the response that can be induced or the adjuvants to be used.8686. Drurey C, Coakley G, Maizels RM. Extracellular vesicles: new targets for vaccines against helminth parasites. Int J Parasitol. 2020; 50: 623-33. doi: 10.1016/j.ijpara.2020.04.011.
https://doi.org/10.1016/j.ijpara.2020.04...
In mice immunised with F. gigantica exosome-like particles, burden reduction after metacercariae infection and immunoglobulin production has been pointed out.8787. Sheng ZA, Wu CL, Wang DY, Zhong SH, Yang X, Rao GS, et al. Proteomic analysis of exosome-like vesicles from Fasciola gigantica adult worm provides support for new vaccine targets against fascioliasis. Parasit Vectors. 2023; 16: 62. doi: 10.1186/s13071-023-05659-7.
https://doi.org/10.1186/s13071-023-05659...
On the other hand, immunogenic antigens as part of the cargo of EVs of helminths might be an interesting subject for immunodiagnostic advances research.8888. Gonzáles WHR, Coelho GR, Pimenta DC, de Paula FM, Gryschek RCB. Proteomic analysis of the excretory-secretory products from Strongyloides venezuelensis infective larvae: new insights for the immunodiagnosis of human strongyloidiasis. Parasitol Res. 2022; 121(11): 3155-70. doi: 10.1007/s00436-022-07636-y.
https://doi.org/10.1007/s00436-022-07636...
,8989. Li C, Li C, Xu F, Wang H, Jin X, Zhang Y, et al. Identification of antigens in the Trichinella spiralis extracellular vesicles for serological detection of early stage infection in swine. Parasit Vectors. 2023; 16: 387. doi: 10.1186/s13071-023-06013-7.
https://doi.org/10.1186/s13071-023-06013...

Protozoan parasites

Many protozoan parasites successfully exert intracellular parasitism and have adapted to their human hosts in such a way that they are even able to modulate, at a certain stage, part of the interaction with the vascular endothelium and its microenvironment through EVs, facilitating the establishment of infections such as it occurs in malaria.9090. Babatunde KA, Subramanian BY, Ahouidi AD, Murillo PM, Walch M, Mantel PY. Role of extracellular vesicles in cellular cross talk in malaria. Front Immunol. 2020; 11: 22. doi: 10.3389/fimmu.2020.00022.
https://doi.org/10.3389/fimmu.2020.00022...
,9191. Varikuti S, Jha BK, Holcomb EA, McDaniel JC, Karpurapu M, Srivastava N, et al. The role of vascular endothelium and exosomes in human protozoan parasitic diseases. Vessel Plus. 2020; 4: 28. doi: 10.20517/2574-1209.2020.27.
https://doi.org/10.20517/2574-1209.2020....
Likewise, infected host cells can induce pro- and anti-invasion responses through their EVs.9292. de Souza W, Barrias ES. Membrane-bound extracellular vesicles secreted by parasitic protozoa: cellular structures involved in the communication between cells. Parasitol Res. 2020; 119: 2005-23. doi: 10.1007/s00436-020-06691-7.
https://doi.org/10.1007/s00436-020-06691...
However, the first contact between the parasite (sometimes coming from a vector) and the host tissues necessarily occurs in invasive forms that, to continue the life cycle, eventually reappear at certain times or under specific circumstances.9393. Kato K, Sugi T, Iwanaga T. Roles of Apicomplexan protein kinases at each life cycle stage. Parasitol Int. 2012; 61: 224-34. doi: 10.1016/j.parint.2011.12.002.
https://doi.org/10.1016/j.parint.2011.12...
,9494. Rossi IV, Nunes MAF, Vargas-Otalora S, Ferreira TCS, Cortez M, Ramirez MI. Extracellular vesicles during TriTryps infection: complexity and future challenges. Mol Immunol. 2021; 132: 172-83. doi: 10.1016/j.molimm.2021.01.008.
https://doi.org/10.1016/j.molimm.2021.01...
In the framework of experimentation with protozoa and their EVs, these vesicles can sometimes be studied in axenic culture.9494. Rossi IV, Nunes MAF, Vargas-Otalora S, Ferreira TCS, Cortez M, Ramirez MI. Extracellular vesicles during TriTryps infection: complexity and future challenges. Mol Immunol. 2021; 132: 172-83. doi: 10.1016/j.molimm.2021.01.008.
https://doi.org/10.1016/j.molimm.2021.01...

Intracellular protozoan parasites

In apicomplexan-related infections like malaria, provoked by species of the obligate intracellular parasite Plasmodium, the study of EVs obtained from its invasive stage is scarce, since the growth of the parasite requires the use of cell culture. In this sense, several works have focused on the study of infected-red cell derived EVs;9595. Abdi A, Yu L, Goulding D, Rono MK, Bejon P, Choudhary J, et al. Proteomic analysis of extracellular vesicles from a Plasmodium falciparum Kenyan clinical isolate defines a core parasite secretome. Wellcome Open Res. 2017; 2: 50. doi: 10.12688/wellcomeopenres.11910.1.
https://doi.org/10.12688/wellcomeopenres...
,9696. Vimonpatranon S, Roytrakul S, Phaonakrop N, Lekmanee K, Atipimonpat A, Srimark N, et al. Extracellular vesicles derived from early and late stage Plasmodium falciparum-infected red blood cells contain invasion-associated proteins. J Clin Med. 2022; 11: 4250. doi: 10.3390/jcm11144250.
https://doi.org/10.3390/jcm11144250...
however, it would be worth exploring the role of EVs from their sporozoites and merozoites in the mechanisms of invasion of hepatocytes and red blood cells.

EVs secreted by tachyzoites of Toxoplasma gondii, another parasite, have also been characterised using transmission electron microscopy (TEM) and NTA, and purified by gel exclusion chromatography.9797. 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. 2018; 13(10): 1157-68. doi: 10.2217/nnm-2018-0035.
https://doi.org/10.2217/nnm-2018-0035...
,9898. Quiarim TM, Maia MM, da Cruz AB, Taniwaki NN, Namiyama GM, Pereira-Chioccola VL. Characterization of extracellular vesicles isolated from types I, II and III strains of Toxoplasma gondii. Acta Trop. 2021; 219: 105915. doi: 10.1016/j.actatropica.2021.105915.
https://doi.org/10.1016/j.actatropica.20...
Furthermore, they have been related to: (i) the in vitro stimulation of a proinflammatory profile in macrophages,9797. 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. 2018; 13(10): 1157-68. doi: 10.2217/nnm-2018-0035.
https://doi.org/10.2217/nnm-2018-0035...
(ii) the expression of different miRNAs as possible cargoes,9898. Quiarim TM, Maia MM, da Cruz AB, Taniwaki NN, Namiyama GM, Pereira-Chioccola VL. Characterization of extracellular vesicles isolated from types I, II and III strains of Toxoplasma gondii. Acta Trop. 2021; 219: 105915. doi: 10.1016/j.actatropica.2021.105915.
https://doi.org/10.1016/j.actatropica.20...
,9999. 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. 2018; 40(9): e12571. doi: 10.1111/pim.12571.
https://doi.org/10.1111/pim.12571...
(iii) the promotion of host immune evasion,100100. Gómez-Chávez F, Murrieta-Coxca JM, Caballero-Ortega H, Morales-Prieto DM, Markert UR. Host-pathogen interactions mediated by extracellular vesicles in Toxoplasma gondii infection during pregnancy. J Reprod Immunol. 2023; 158: 103957. doi: 10.1016/j.jri.2023.103957.
https://doi.org/10.1016/j.jri.2023.10395...
and (iv) enhanced virulence (in terms of parasitaemia) in mice, five days post infection (p.i.) (through co-inoculation of EVs and tachyzoites).9898. Quiarim TM, Maia MM, da Cruz AB, Taniwaki NN, Namiyama GM, Pereira-Chioccola VL. Characterization of extracellular vesicles isolated from types I, II and III strains of Toxoplasma gondii. Acta Trop. 2021; 219: 105915. doi: 10.1016/j.actatropica.2021.105915.
https://doi.org/10.1016/j.actatropica.20...
Immunisation with tachyzoite-released EVs showed to trigger humoral immune responses, increasing the survival rate of mice challenged with a lethal dose of parasites. Finally, immunohistochemistry showed high expression of tumour necrosis factor (TNF-α) in spleen cells, along with IL-10 and interferon (IFN-γ) in spleen and brain cells.101101. Maia MM, da Cruz AB, Taniwaki NN, Namiyama GM, Gava R, Gomes AHS, et al. Immunization with extracellular vesicles excreted by Toxoplasma gondii confers protection in murine infection, activating cellular and humoral responses. Int J Parasitol. 2021; 51: 559-69. doi: 10.1016/j.ijpara.2020.11.010.
https://doi.org/10.1016/j.ijpara.2020.11...

On the other hand, the trypanosomatid protozoan parasites Trypanosoma cruzi and Leishmania sp., which cause American trypanosomiasis (Chagas disease) and leishmaniasis, share the characteristic of being transmitted to humans mainly by arthropod vectors: triatomine bugs and sandflies, respectively. The effect of EVs in the interaction of these parasites with their vectors during the extrinsic cycle (stage in which they also manifest themselves extracellularly) has been catalogued as negative for early migration of T. cruzi in the digestive tract of Rhodnius prolixus pre-fed with epimastigote-derived EVs; although with no effect on the amount of metacyclic trypomastigotes (the infective form for humans) at 28 days p.i., nor in Triatoma infestans in general.102102. 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: e190217. doi: 10.1590/0074-02760190217.
https://doi.org/10.1590/0074-02760190217...
The secretion of parasite EVs occurs not only in the arthropod midgut, but also at the vector-host interface, as it has been demonstrated with Leishmania-derived EVs present in the inoculum at the site of the bite.103103. 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. 2015; 13(5): 957-67. doi: 10.1016/j.celrep.2015.09.058.
https://doi.org/10.1016/j.celrep.2015.09...

The first encounter of Leishmania sp. with host cells occurs at the dermal level.104104. Serafim TD, Coutinho-Abreu IV, Dey R, Kissinger R, Valenzuela JG, Oliveira F, et al. Leishmaniasis: the act of transmission. Trends Parasitol. 2021; 37(11): 976-87. doi: 10.1016/j.pt.2021.07.003.
https://doi.org/10.1016/j.pt.2021.07.003...
Mice footpad co-injection of EVs and metacyclic promastigotes of L. major causes exacerbated swelling and increased parasite load, with a rise in the expression of proinflammatory cytokines such as IL-17a.103103. 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. 2015; 13(5): 957-67. doi: 10.1016/j.celrep.2015.09.058.
https://doi.org/10.1016/j.celrep.2015.09...
,105105. Barbosa FMC, Dupin TV, Toledo MS, Reis NFC, Ribeiro K, Cronemberger-Andrade A, et al. Extracellular vesicles released by Leishmania (Leishmania) amazonensis promote disease progression and induce the production of different cytokines in macrophages and B-1 Cells. Front Microbiol. 2018; 9: 3056. doi: 10.3389/fmicb.2018.03056.
https://doi.org/10.3389/fmicb.2018.03056...
In counterbalance, the production of IL-6 and IL-10, along with the de-stimulation of TNF-α, has also been observed in monocytes and macrophages in the presence of Leishmania-derived EVs,105105. Barbosa FMC, Dupin TV, Toledo MS, Reis NFC, Ribeiro K, Cronemberger-Andrade A, et al. Extracellular vesicles released by Leishmania (Leishmania) amazonensis promote disease progression and induce the production of different cytokines in macrophages and B-1 Cells. Front Microbiol. 2018; 9: 3056. doi: 10.3389/fmicb.2018.03056.
https://doi.org/10.3389/fmicb.2018.03056...
,106106. Silverman JM, Clos J, Horakova E, Wang AY, Wiesgigl M, Kelly I, et al. Leishmania exosomes modulate innate and adaptive immune responses through effects on monocytes and dendritic cells. J Immunol. 2010; 185: 5011-22. doi: 10.4049/jimmunol.1000541.
https://doi.org/10.4049/jimmunol.1000541...
associated with an immunosuppressive effect and benefiting parasite’s survival.107107. Dong G, Wagner V, Minguez-Menendez A, Fernandez-Prada C, Olivier M. Extracellular vesicles and leishmaniasis: current knowledge and promising avenues for future development. Mol Immunol. 2021; 135: 73-83. doi: 10.1016/j.molimm.2021.04.003.
https://doi.org/10.1016/j.molimm.2021.04...
Indeed, the presence of GP63 in Leishmania EVs represents an anti-inflammatory regulation mechanism.108108. Hassani K, Shio MT, Martel C, Faubert D, Olivier M. Absence of metalloprotease GP63 alters the protein content of Leishmania exosomes. PLoS One. 2014; 9(4): e95007. doi: 10.1371/journal.pone.0095007.
https://doi.org/10.1371/journal.pone.009...
Besides, an important enrichment of RNA cargo has been found in 120 nm EVs of axenic cultures of Leishmania.109109. Lambertz U, Ovando MEO, Vasconcelos EJR, 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. 2015; 16: 151. doi: 10.1186/s12864-015-1260-7.
https://doi.org/10.1186/s12864-015-1260-...

Back to the case of T. cruzi, the causative agent of Chagas disease, in the context of the acute phase of the infection, there are several interesting findings: EVs produced during early parasite-host contact promote parasite infectivity in Vero cells110110. 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; 19: e12672. doi: 10.1111/cmi.12672.
https://doi.org/10.1111/cmi.12672...
,111111. 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; 13(2): e0007163. doi: 10.1371/journal.pntd.0007163.
https://doi.org/10.1371/journal.pntd.000...
and their injection in mice prior to trypomastigote inoculation leads to more inflammation, higher parasitism and formation of amastigotes nests, with CD4+ lymphocytes infiltration in the heart.112112. Torrecilhas ACT, 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; 11: 29-39. doi: 10.1016/j.micinf.2008.10.003.
https://doi.org/10.1016/j.micinf.2008.10...
It has also been proven that T. cruzi EVs can inhibit complement lytic activity,113113. Lozano IMD, 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; 7: 44451. doi: 10.1038/srep44451.
https://doi.org/10.1038/srep44451...
which is a form of initial immune evasion.

More recent studies on T. cruzi trypomastigote-derived EVs reveal an increase in Ca2+ mobilisation and permeabilisation in Vero cells treated with these vesicles,111111. 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; 13(2): e0007163. doi: 10.1371/journal.pntd.0007163.
https://doi.org/10.1371/journal.pntd.000...
as well as the induction of a proinflammatory profile of cytokines (TNF-α and IL-6) in macrophages and muscle cells.114114. Choudhuri S, Garg NJ. PARP1-cGAS-NF-?B pathway of proinflammatory macrophage activation by extracellular vesicles released during Trypanosoma cruzi infection and Chagas disease. PLoS Pathog. 2020; 16(4): e1008474. doi: 10.1371/journal.ppat.1008474.
https://doi.org/10.1371/journal.ppat.100...
EVs in T. cruzi may diverge in structure and composition, depending on the stage of the parasite (trypomastigote vs. epimastigote):115115. 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; 22: 5183. doi: 10.3390/ijms22105183.
https://doi.org/10.3390/ijms22105183...
average sizes of 183 nm and 259 nm were determined in epimastigote-derived EVs, resulting larger than trypomastigote-derived EVs (60 nm and 143 nm). Moreover, significant differences were found in the exoproteome, particularly in one of the most important virulence factors: proteins of the trans-sialidase family, with greater presence and diversity in trypomastigote-derived EVs.115115. 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; 22: 5183. doi: 10.3390/ijms22105183.
https://doi.org/10.3390/ijms22105183...

Even though the specific generation pathways of EVs in trypanosomatids are unknown,9494. Rossi IV, Nunes MAF, Vargas-Otalora S, Ferreira TCS, Cortez M, Ramirez MI. Extracellular vesicles during TriTryps infection: complexity and future challenges. Mol Immunol. 2021; 132: 172-83. doi: 10.1016/j.molimm.2021.01.008.
https://doi.org/10.1016/j.molimm.2021.01...
there is some evidence that there could be ESCRT independent mechanisms, as nanotube derived EVs in T. brucei, ESCRT dependent multivesicular bodies (MVBs) in Leishmania, or new biogenesis pathways like reservosomes EVs in T. cruzi.44. Cruz Camacho A, Alfandari D, Kozela E, Regev-Rudzki N. Biogenesis of extracellular vesicles in protozoan parasites: the ESCRT complex in the trafficking fast lane? PLoS Pathog. 2023; 19(2): e1011140. doi: 10.1371/journal.ppat.1011140.
https://doi.org/10.1371/journal.ppat.101...
Cargoes of trypanosomatid-derived EVs are the reflection of well-known glycoproteins and soluble proteins from the parasite, which eventually interact with TLRs.116116. 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. doi: 10.3389/fcimb.2020.602502.
https://doi.org/10.3389/fcimb.2020.60250...
Besides, an interesting approach for implication in virulence might be related to the capacity of EVs of inducing/transferring resistant phenotypes or improving parasite fitness.117117. 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. 2022; 40: 111121. doi: 10.1016/j.celrep.2022.111121.
https://doi.org/10.1016/j.celrep.2022.11...
,118118. Rossi IV, Nunes MAF, Sabatke B, Ribas HT, Winnischofer SMB, Ramos ASP, et al. An induced population of Trypanosoma cruzi epimastigotes more resistant to complement lysis promotes a phenotype with greater differentiation, invasiveness, and release of extracellular vesicles. Front Cell Infect Microbiol. 2022; 12: 1046681. doi: 10.3389/fcimb.2022.1046681.
https://doi.org/10.3389/fcimb.2022.10466...

Other trypanosomatid, already mentioned, but eminently extracellular along its life cycle is T. brucei, the causative agent of African sleeping sickness, whose EVs covered with variant surface glycoproteins (VSGs) have been involved in pathogenesis due to their fusogenic capacity with erythrocytes.119119. 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; 164(0): 246-57. doi: 10.1016/j.cell.2015.11.051.
https://doi.org/10.1016/j.cell.2015.11.0...
Furthermore, upregulation triggering effects on CD4+ and CD8+ T cells and stimulation of MHC expression in macrophages have also been observed.120120. Dias-Guerreiro T, Palma-Marques J, Mourata-Gonçalves P, Alexandre-Pires G, Valério-Bolas A, Gabriel A, et al. African trypanosomiasis: extracellular vesicles shed by Trypanosoma brucei brucei manipulate host mononuclear cells. Biomedicines. 2021; 9: 1056. doi: 10.3390/biomedicines9081056.
https://doi.org/10.3390/biomedicines9081...

Extracellular protozoan parasites

There is another group of unicellular eukaryotes that exert parasitism extracellularly through vegetative forms, the trophozoites, and lack intracellular evolutionary forms; the mechanisms of pathogenesis in these cases involve some effectors other than the intracellular arsenal. However, EVs have come to light in recent research as molecular mediators of these pathogens. In Figure, a depiction of the principal role of their EVs over several scenarios is shown. A size comparison of EVs obtained from extracellular protozoa, by several techniques such as NTA, TEM, and DLS, is presented in Table. This table also summarises isolation methodologies employed by different groups that investigate EVs from these parasites; most of these methods are recommended and implemented by other protozoan EVs researchers.121121. Fernandez-Becerra C, Xander P, Alfandari D, Dong G, Aparici-Herraiz I, Rosenhek-Goldian I, et al. Guidelines for the purification and characterization of extracellular vesicles of parasites. J Extracell Biol. 2023; 2: e117. doi: 10.1002/jex2.117.
https://doi.org/10.1002/jex2.117...


Explored general roles of extracellular vesicles (EVs) derived from extracellular protozoa in pathogenesis, parasite-parasite communication, and its relationship with immune effectors. (A) Entamoeba histolytica EVs are possible involved in en/excystment processes and have effects over neutrophils; (B) Giardia duodenalis EVs provoke alteration of Caco-2 cells tight junctions and enterobacteria, promote adhesion of the parasite and induce a proinflammatory outcome; (C) free living amoeba (i.e., Acanthamoeba sp., Naegleria fowleri) derived-EVs are uptaken by glial cells and other mammalian cells and are also associated with a proinflammatory chemokine/cytokine production; (D) Trichomonas vaginalis induce the production of nitric oxide (NO) in macrophages and stimulate adhesion of the parasites to ectocervical cells. Figure created with BioRender.com.

Giardia duodenalis - G. duodenalis (syn. G. intestinalis) is an intestinal parasite that adheres, as its trophozoite form, to the epithelium of the small intestine, with effects on enterocytes that induce malabsorptive diarrhoea.122122. Buret AG. Pathophysiology of enteric infections with Giardia duodenalis. Parasite. 2008; 15: 261-5. doi: 10.1051/parasite/2008153261.
https://doi.org/10.1051/parasite/2008153...
Its cystic form protects it from environmental adversities and differentiation involves the transport of components of the extracellular cystic wall by dense granule-like vesicles, called encystation specific vesicles, whose origin is associated with the ER.123123. Lanfredi-Rangel A, Attias M, Reiner DS, Gillin FD, De Souza W. Fine structure of the biogenesis of Giardia lamblia encystation secretory vesicles. J Struct Biol. 2003; 143: 153-63. doi: 10.1016/S1047-8477(03)00123-0.
https://doi.org/10.1016/S1047-8477(03)00...
It has been proposed that, during the release of their content, remnants of the plasma membrane catalogued as “empty vesicles or membrane ghosts” are formed and remain attached to flagella or suspended in the extracellular milieu.124124. Benchimol M. The release of secretory vesicle in encysting Giardia lamblia. FEMS Microbiol Lett. 2004; 235: 81-7. doi: 10.1016/j.femsle.2004.04.014.
https://doi.org/10.1016/j.femsle.2004.04...

During the last decade, EVs have been described as part of the secretome of G. duodenalis in axenic cultures,125125. Ma'ayeh SY, Liu J, Peirasmaki D, Hörnaeus K, Lind SB, Grabherr M, et al. Characterization of the Giardia intestinalis secretome during interaction with human intestinal epithelial cells: the impact on host cells. PLoS Negl Trop Dis. 2017; 11(12): e0006120. doi: 10.1371/journal.pntd.0006120.
https://doi.org/10.1371/journal.pntd.000...
with average size of 201,6 nm;126126. 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. 2017; 96: 131-42. doi: 10.1016/j.ejcb.2017.01.005.
https://doi.org/10.1016/j.ejcb.2017.01.0...
now, it is possible to focus on specific size subpopulations. For instance, there is a modified differential centrifugation protocol that enriches populations > 100 nm.127127. Sana A, Rossi IV, Sabatke B, Bonato LB, Medeiros LCS, Ramirez MI. An improved method to enrich large extracellular vesicles derived from Giardia intestinalis through differential centrifugation. Life. 2023; 13: 1799. doi: 10.3390/life13091799.
https://doi.org/10.3390/life13091799...
Actually, lipid profiles vary between small and large EVs128128. Faria CP, Ferreira B, Lourenço A, Guerra I, Melo T, Domingues P, et al. Lipidome of extracellular vesicles from Giardia lamblia. PLoS One. 2023; 18(9): e0291292. doi: 10.1371/journal.pone.0291292.
https://doi.org/10.1371/journal.pone.029...
which can help to understand to role of some lipid species in EVs release, as it has been proposed,126126. 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. 2017; 96: 131-42. doi: 10.1016/j.ejcb.2017.01.005.
https://doi.org/10.1016/j.ejcb.2017.01.0...
adhesion, encystation and signalling.128128. Faria CP, Ferreira B, Lourenço A, Guerra I, Melo T, Domingues P, et al. Lipidome of extracellular vesicles from Giardia lamblia. PLoS One. 2023; 18(9): e0291292. doi: 10.1371/journal.pone.0291292.
https://doi.org/10.1371/journal.pone.029...
Besides, their involvement in pathogenic processes has begun to be elucidated: there is increased trophozoite adhesion to Caco-2 cells in the presence of G. duodenalis-derived EVs, they contribute to the maturation of dendritic cells,126126. 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. 2017; 96: 131-42. doi: 10.1016/j.ejcb.2017.01.005.
https://doi.org/10.1016/j.ejcb.2017.01.0...
alter tight junctions given by ZO-1 and Claudin-4,129129. Siddiq A, Allain T, Dong G, Olivier M, Buret A. Giardia extracellular vesicles disrupt intestinal epithelial and inhibit the growth of commensal bacterial while increasing their swimming motility. FASEB J. 2020; 34(S1): 1-1. doi: 10.1096/fasebj.2020.34.s1.00515.
https://doi.org/10.1096/fasebj.2020.34.s...
and there are virulence factors such as antigenic variable surface proteins (VSPs) and giardin in cyst-derived EVs126126. 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. 2017; 96: 131-42. doi: 10.1016/j.ejcb.2017.01.005.
https://doi.org/10.1016/j.ejcb.2017.01.0...
,130130. Zhao P, Cao L, Wang X, Dong J, Zhang N, Li X, et al. Extracellular vesicles secreted by Giardia duodenalis regulate host cell innate immunity via TLR2 and NLRP3 inflammasome signaling pathways. PLoS Negl Trop Dis. 2021; 15(4): e0009304. doi: 10.1371/journal.pntd.0009304.
https://doi.org/10.1371/journal.pntd.000...
and trophozoites.131131. Gavinho B, Sabatke B, Feijoli V, Rossi IV, da Silva JM, Evans-Osses I, et al. Peptidyl arginine 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. 2020; 10: 417. doi: 10.3389/fcimb.2020.00417.
https://doi.org/10.3389/fcimb.2020.00417...
In general, proinflammatory effects and raised immunogenicity are driven by EVs secreted by G. duodenalis.132132. Ferreira B, Lourenço A, Sousa MC. Protozoa-derived extracellular vesicles on intercellular communication with special emphasis on Giardia lamblia. Microorganisms. 2022; 10: 2422. doi: 10.3390/microorganisms10122422.
https://doi.org/10.3390/microorganisms10...

In addition, G. duodenalis has also an internal membranous system: peripheral vesicles (PVs), which have been linked to part of the ESCRT machinery,133133. 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. 2018; 97: 44-62. doi: 10.1016/j.ejcb.2017.11.004.
https://doi.org/10.1016/j.ejcb.2017.11.0...
highlighting the possibility that it operates at this level as part of a secretory pathway. PVs can act as microvesicular bodies with intraluminal vesicles (ILVs), so could be linked to the origin of EVs.134134. 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. 2019; 207: 301-11. doi: 10.1016/j.jsb.2019.07.002.
https://doi.org/10.1016/j.jsb.2019.07.00...
It has been proposed that these occurs in both vegetative and resistance forms, adding a potential link to differentiation between these phases.134134. 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. 2019; 207: 301-11. doi: 10.1016/j.jsb.2019.07.002.
https://doi.org/10.1016/j.jsb.2019.07.00...
Indeed, another author highlights an EVs release that depends on ESCRT-associated molecules.135135. 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. 2019; 8: 1600. doi: 10.3390/cells8121600.
https://doi.org/10.3390/cells8121600...

Among other pathophysiological roles associated with EVs of G. duodenalis, a subpopulation of 187,6 nm was able to restore parasite adhesion capacity after the treatment with Cl-amidine, an inhibitor of peptidyl arginine deiminase in Caco-2 cells.131131. Gavinho B, Sabatke B, Feijoli V, Rossi IV, da Silva JM, Evans-Osses I, et al. Peptidyl arginine 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. 2020; 10: 417. doi: 10.3389/fcimb.2020.00417.
https://doi.org/10.3389/fcimb.2020.00417...
Also, pretreatment of murine macrophages with G. duodenalis-derived EVs generated increases in cytokines such as IL-6 and TNF-α, as with it happened with trophozoites.136136. Zhao P, Cao L, Wang X, Li J, Dong J, Zhang N, et al. Giardia duodenalis extracellular vesicles regulate the proinflammatory immune response in mouse macrophages in vitro via the MAPK, AKT and NF-?B pathways. Parasit Vectors. 2021; 14: 358. doi: 10.1186/s13071-021-04865-5.
https://doi.org/10.1186/s13071-021-04865...
In addition, G. duodenalis EVs induced phosphorylation and activation of p38, ERK and AKT signalling pathways, the NF-κB pathway136136. Zhao P, Cao L, Wang X, Li J, Dong J, Zhang N, et al. Giardia duodenalis extracellular vesicles regulate the proinflammatory immune response in mouse macrophages in vitro via the MAPK, AKT and NF-?B pathways. Parasit Vectors. 2021; 14: 358. doi: 10.1186/s13071-021-04865-5.
https://doi.org/10.1186/s13071-021-04865...
and NLRP3 of the inflammasome, which possibly mediates IL-1β production.130130. Zhao P, Cao L, Wang X, Dong J, Zhang N, Li X, et al. Extracellular vesicles secreted by Giardia duodenalis regulate host cell innate immunity via TLR2 and NLRP3 inflammasome signaling pathways. PLoS Negl Trop Dis. 2021; 15(4): e0009304. doi: 10.1371/journal.pntd.0009304.
https://doi.org/10.1371/journal.pntd.000...

Finally, by evaluating the effect of EVs of G. duodenalis on commensal bacteria such as E. coli and Enterobacter cloacae, it was revealed that these vesicles could modulate growth, biofilm formation, motility, and adhesion to the epithelium,137137. Siddiq A, Dong G, Balan B, Harrison LG, Jex A, Olivier M, et al. A thermo-resistant and RNase-sensitive cargo from Giardia duodenalis extracellular vesicles modifies the behaviour of enterobacteria. J Extracell Biol. 2023; 2: e109. doi: 10.1002/jex2.109.
https://doi.org/10.1002/jex2.109...
,138138. Siddiq A, Allain T, Dong G, Olivier M, Buret A. Role of extracellular vesicles in Giardia-microbiota interactions. J Can Assoc Gastroenterol. 2021; 4(S1): 226-8. doi: 10.1093/jcag/gwab002.200.
https://doi.org/10.1093/jcag/gwab002.200...
which suggests new roles in the interaction with host microbiota.

Trichomonas vaginalis - Trichomoniasis is the most common non-viral sexually transmitted disease, which mainly affects women in reproductive age, but can also be symptomatic in men.139139. Poole DN, McClelland RS. Global epidemiology of Trichomonas vaginalis. Sex Transm Infect. 2013; 89(6): 418-22. doi: 10.1136/sextrans-2013-051075.
https://doi.org/10.1136/sextrans-2013-05...
The parasite causing the disease is the flagellate T. vaginalis. In the first description of EVs produced by trophozoites of this agent, an overlapping of protein composition compared to mammalian exosomes was concluded;140140. 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. PLoS Pathog. 2013; 9(7): e1003482. doi: 10.1371/journal.ppat.1003482.
https://doi.org/10.1371/journal.ppat.100...
this was similar to the findings published by Nievas et al.,141141. 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. 2018; 75: 2211-26. doi: 10.1007/s00018-017-2726-3.
https://doi.org/10.1007/s00018-017-2726-...
who reported a 56% of proteins homologous to those found in a fraction of human EVs. In addition, most proteins with signalling functions and metabolic enzymes were identified from those with identifiable domains.140140. 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. PLoS Pathog. 2013; 9(7): e1003482. doi: 10.1371/journal.ppat.1003482.
https://doi.org/10.1371/journal.ppat.100...
Using SEM, an increased protrusion of EVs from parasites due to the presence of CaCl2 was shown.141141. 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. 2018; 75: 2211-26. doi: 10.1007/s00018-017-2726-3.
https://doi.org/10.1007/s00018-017-2726-...

Other cargoes described in T. vaginalis-derived EVs are: surface proteins of the BspA family,140140. 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. PLoS Pathog. 2013; 9(7): e1003482. doi: 10.1371/journal.ppat.1003482.
https://doi.org/10.1371/journal.ppat.100...
,141141. 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. 2018; 75: 2211-26. doi: 10.1007/s00018-017-2726-3.
https://doi.org/10.1007/s00018-017-2726-...
which are molecules possibly involved in pathogenesis; ARF proteins, relevant for their relationship with formation, release and cargo selection;141141. 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. 2018; 75: 2211-26. doi: 10.1007/s00018-017-2726-3.
https://doi.org/10.1007/s00018-017-2726-...
tetraspanin TSP1140140. 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. PLoS Pathog. 2013; 9(7): e1003482. doi: 10.1371/journal.ppat.1003482.
https://doi.org/10.1371/journal.ppat.100...
,142142. Ong SC, Cheng WH, Ku FM, Tsai CY, Huang PJ, Lee CC, et al. Identification of endosymbiotic virus in small extracellular vesicles derived from Trichomonas vaginalis. Genes. 2022; 13: 531. doi: 10.3390/genes13030531.
https://doi.org/10.3390/genes13030531...
and VPS32, a molecule involved in the ESCRT III complex, which in T. vaginalis is related to the biogenic regulation of EVs, cargo sorting and parasite adhesion;143143. Salas N, Coceres VM, Melo TS, 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. 2022; 79: 11. doi: 10.1007/s00018-021-04083-3.
https://doi.org/10.1007/s00018-021-04083...
tRNA fragments4646. Artuyants A, Campos TL, Rai AK, Johnson PJ, Dauros-Singorenko P, Phillips A, et al. Extracellular vesicles produced by the protozoan parasite Trichomonas vaginalis contain a preferential cargo of tRNA-derived small RNAs. Int J Parasitol. 2020; 50: 1145-55. doi: 10.1016/j.ijpara.2020.07.003.
https://doi.org/10.1016/j.ijpara.2020.07...
and Trichomonasvirus particles, that might be transmitted to the host and is a possibly a critical element in disease development.142142. Ong SC, Cheng WH, Ku FM, Tsai CY, Huang PJ, Lee CC, et al. Identification of endosymbiotic virus in small extracellular vesicles derived from Trichomonas vaginalis. Genes. 2022; 13: 531. doi: 10.3390/genes13030531.
https://doi.org/10.3390/genes13030531...
,144144. Rada P, Hrdý I, Zdrha A, Narayanasamy RK, Smutná T, Horácková J, et al. Double-stranded RNA viruses are released from Trichomonas vaginalis inside small extracellular vesicles and modulate the exosomal cargo. Front Microbiol. 2022; 13: 893692. doi: 10.3389/fmicb.2022.893692.
https://doi.org/10.3389/fmicb.2022.89369...
Proteins involved in filopodia and in the formation of cytonemes (e.g., small actin-binding proteins, calreticulin and Rho/Ras family proteins) were found in EVs implicated in parasite-parasite communication.145145. Salas N, Pedreros MB, Melo TS, Maguire VG, Sha J, Wohlschlegel JA, et al. Role of cytoneme structures and extracellular vesicles in Trichomonas vaginalis parasite: parasite communication. Elife. 2023; 12. doi: 10.7554/eLife.86067.

Characterisation of the EVs uptake by host cells demonstrated the fusion with ectocervical cell membranes to release their contents140140. 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. PLoS Pathog. 2013; 9(7): e1003482. doi: 10.1371/journal.ppat.1003482.
https://doi.org/10.1371/journal.ppat.100...
and internalisation, with fluorescence and fluorimetry assays in BPH-1.146146. Rai AK, Johnson PJ. Trichomonas vaginalis extracellular vesicles are internalized by host cells using proteoglycans and caveolin-dependent endocytosis. Proc Natl Acad Sci USA. 2019; 116(43): 21354-60. doi: 10.1073/pnas.1912356116.
https://doi.org/10.1073/pnas.1912356116...
This uptake might be Ca2+-dependent, mediated by glycosaminoglycans and heparan sulphate in proteoglycans from host cells and 4-α-glucanotransferase homologues that act as ligands in EVs.146146. Rai AK, Johnson PJ. Trichomonas vaginalis extracellular vesicles are internalized by host cells using proteoglycans and caveolin-dependent endocytosis. Proc Natl Acad Sci USA. 2019; 116(43): 21354-60. doi: 10.1073/pnas.1912356116.
https://doi.org/10.1073/pnas.1912356116...
Entry by action of caveolin-1 and lipid raft dependent endocytosis has been established,4646. Artuyants A, Campos TL, Rai AK, Johnson PJ, Dauros-Singorenko P, Phillips A, et al. Extracellular vesicles produced by the protozoan parasite Trichomonas vaginalis contain a preferential cargo of tRNA-derived small RNAs. Int J Parasitol. 2020; 50: 1145-55. doi: 10.1016/j.ijpara.2020.07.003.
https://doi.org/10.1016/j.ijpara.2020.07...
which has been successfully inhibited by cholesterol depletion agents.146146. Rai AK, Johnson PJ. Trichomonas vaginalis extracellular vesicles are internalized by host cells using proteoglycans and caveolin-dependent endocytosis. Proc Natl Acad Sci USA. 2019; 116(43): 21354-60. doi: 10.1073/pnas.1912356116.
https://doi.org/10.1073/pnas.1912356116...

Regarding the pathogenic process, it could be pointed out that EVs (from a high adherent strain) increased adhesion by stimulating both host cells and parasites from less adherent strains;140140. 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. PLoS Pathog. 2013; 9(7): e1003482. doi: 10.1371/journal.ppat.1003482.
https://doi.org/10.1371/journal.ppat.100...
the same group demonstrated a positive outcome in survival and parasite burden when co-incubated with EVs, confirming a role in colonisation.147147. Molgora BM, Mukherjee SK, Baumel-Alterzon S, Santiago FM, Muratore KA, Sisk AE, et al. Trichomonas vaginalis adherence phenotypes and extracellular vesicles impact parasite survival in a novel in vivo model of pathogenesis. PLoS Negl Trop Dis. 2023; 17(10): e00011693. doi: 10.1371/journal.pntd.0011693.
https://doi.org/10.1371/journal.pntd.001...
Nitric oxide (NO) production in macrophages has been detected, indicating EVs-mediated activation.148148. 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. 2017; 39(6): e12426. doi: 10.1111/pim.12426.
https://doi.org/10.1111/pim.12426...
When animal and cellular models are pre-treated with EVs of T. vaginalis, an immune response has been determined, with a mitigating tendency that reduces mice oedema and inflammation and with significant increases in IL- 10;148148. 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. 2017; 39(6): e12426. doi: 10.1111/pim.12426.
https://doi.org/10.1111/pim.12426...
conversely, IL-6 is elevated to a lesser extent and no real regulation by EVs has been observed.140140. 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. PLoS Pathog. 2013; 9(7): e1003482. doi: 10.1371/journal.ppat.1003482.
https://doi.org/10.1371/journal.ppat.100...

Parasitic and free-living amoebae (FLA) - In amoebae such as Dictyostelium discoideum, EVs were described since 1998, as vesicular organelles of 100 - 300 nm.149149. Tatischeff I, Bomsel M, De Paillerets C, Durand H, Geny B, Segretain D, et al. Dictyostelium discoideum cells shed vesicles with associated DNA and vital stain Hoechst 33342. Cell Mol Life Sci. 1998; 54: 476-87. This organism has been tested as a eukaryotic model for the study of several diseases, cellular processes, and host-pathogen interactions, due to its easy manipulation and growth.150150. Tatischeff I. Dictyostelium: a model for studying the extracellular vesicle messengers involved in human health and disease. Cells. 2019; 8: 225. doi: 10.3390/cells8030225.
https://doi.org/10.3390/cells8030225...
Therefore, it has also been postulated as a potential model for research on the heterogeneity of EVs and the elucidation of their biological functions.150150. Tatischeff I. Dictyostelium: a model for studying the extracellular vesicle messengers involved in human health and disease. Cells. 2019; 8: 225. doi: 10.3390/cells8030225.
https://doi.org/10.3390/cells8030225...

Subsequently, with the escalate of interest in EVs in different research groups, work lines have been developed around the pathogenic intestinal amoeba such as Entamoeba histolytica, but also on free-living organisms with pathogenic potential (amphizoic) such as Acanthamoeba sp. and Naegleria fowleri, with highlights of the possible intervention of EVs in the mechanisms of damage and pathogenesis. Besides, an emerging issue is related to encapsulation of pathogenic bacteria (such as Legionella pneumophila) and respiratory viruses in EVs secreted by FLA, EVs serving as easy alveoli contact spreaders.151151. Dey R, Folkins MA, Ashbolt NJ. Extracellular amoebal-vesicles: potential transmission vehicles for respiratory viruses. NPJ Biofilms Microbiomes. 2021; 7: 25. doi: 10.1038/s41522-021-00201-y.
https://doi.org/10.1038/s41522-021-00201...
,152152. Ashbolt NJ. Conceptual model to inform Legionella-amoebae control, including the roles of extracellular vesicles in engineered water system infections. Front Cell Infect Microbiol. 2023; 13: 1200478. doi: 10.3389/fcimb.2023.1200478.
https://doi.org/10.3389/fcimb.2023.12004...

Entamoeba histolytica - The large intestine is an ideal habitat for colonisation by amoebae and, particularly, E. histolytica has been studied as a potentially invasive agent with complications such as ulcerative colitis and amoebic dysentery.153153. Christy NCV, Petri WA. Mechanisms of adherence, cytotoxicity and phagocytosis modulate the pathogenesis of Entamoeba histolytica. Future Microbiol. 2011; 6(12): 1501-19. doi: 10.2217/fmb.11.120.
https://doi.org/10.2217/fmb.11.120...
With the help of proteomic analyses, molecules involved in the pathogenesis of this amoeba have been identified, like adhesins and cysteine proteases; interestingly, membrane recycling has been suggested since surface membrane proteins were also identified in the excretion-secretion products.154154. Ujang JA, Kwan SH, Ismail MN, Lim BH, Noordin R, Othman N. Proteome analysis of excretory-secretory proteins of Entamoeba histolytica HM1:IMSS via LC-ESI-MS/MS and LC-MALDI-TOF/TOF. Clin Proteomics. 2016; 13: 33. doi: 10.1186/s12014-016-9135-8.
https://doi.org/10.1186/s12014-016-9135-...

Following a study of the endomembrane system, vesicles of 50 - 200 nm were known to be present inside the parasite with possible roles in a protein traffic system together with MVBs and endosomes, as well as the presence of mammalian Alix orthologues in the vesicles,155155. Perdomo D, Aït-Ammar N, Syan S, Sachse M, Jhingan GD, Guillén N. Data set for the proteomics analysis of the endomembrane system from the unicellular Entamoeba histolytica. J Proteomics. 2014; 112(2015): 125-40. doi: 10.1016/j.dib.2014.08.007.
https://doi.org/10.1016/j.dib.2014.08.00...
establishing a possible role of the ESCRT complex. Later, EVs of 125 nm were obtained from axenic culture of E. histolytica,156156. 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. Infect Immun. 2020; 88: 00349-20. doi: 10.1128/IAI.00349-20.
https://doi.org/10.1128/IAI.00349-20...
and a broader range of sizes (50 to less than 600 nm) has also been shown through TEM and NTA.157157. Díaz-Godínez C, Ríos-Valencia DG, García-Aguirre S, Martínez-Calvillo S, Carrero JC. Immunomodulatory effect of extracellular vesicles from Entamoeba histolytica trophozoites: regulation of NETs and respiratory burst during confrontation with human neutrophils. Front Cell Infect Microbiol. 2022; 12: 1018314. doi: 10.3389/fcimb.2022.1018314.
https://doi.org/10.3389/fcimb.2022.10183...
Amoebic EVs were enriched in cell surface galactose/N- acetyl galactosamine-binding lectins and an important part of proteins unveiled by mass spectrometry did not present signal peptide; also, selective small RNA packaging was described and compared to cellular RNA, denoted some differences156156. 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. Infect Immun. 2020; 88: 00349-20. doi: 10.1128/IAI.00349-20.
https://doi.org/10.1128/IAI.00349-20...
Packaging of tRNA fragments also occurs.158158. Sharma M, Zhang H, Ehrenkaufer G, Singh U. Stress response in Entamoeba histolytica is associated with robust processing of tRNA to tRNA halves. mBio. 2023; 14(2): 1-16. doi: 10.1128/mbio.03450-22.
https://doi.org/10.1128/mbio.03450-22...

Functional assays with neutrophils have demonstrated incorporation of amoebic EVs and effects over oxidative burst and NETosis,157157. Díaz-Godínez C, Ríos-Valencia DG, García-Aguirre S, Martínez-Calvillo S, Carrero JC. Immunomodulatory effect of extracellular vesicles from Entamoeba histolytica trophozoites: regulation of NETs and respiratory burst during confrontation with human neutrophils. Front Cell Infect Microbiol. 2022; 12: 1018314. doi: 10.3389/fcimb.2022.1018314.
https://doi.org/10.3389/fcimb.2022.10183...
and intercommunication between parasites in encystment processes.156156. 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. Infect Immun. 2020; 88: 00349-20. doi: 10.1128/IAI.00349-20.
https://doi.org/10.1128/IAI.00349-20...
The latter was seen in a model using Entamoeba invadens.

Acanthamoeba sp. - Amoebae of the genus Acanthamoeba are ubiquitous in nature and capable of generating a central nervous system condition such as amoebic granulomatous encephalitis, but also other more frequent diseases such as amoebic keratitis. The cases are typically associated to genotype T4 and, to a lesser extent, T3 and T11,159159. Maciver SK, Asif M, Simmen MW, Lorenzo-Morales J. A systematic analysis of Acanthamoeba genotype frequency correlated with source and pathogenicity: T4 is confirmed as a pathogen-rich genotype. Eur J Protistol. 2013; 49: 217-21. doi: 10.1016/j.ejop.2012.11.004.
https://doi.org/10.1016/j.ejop.2012.11.0...
among others. In environmental isolates, our research group has described organisms from these and other genotypes with pathogenic potential,160160. Moreira LR, Ramírez DV, Linares F, Ledezma AP, Garro AV, Osuna A, et al. Isolation of Acanthamoeba T5 from water: characterization of its pathogenic potential, including the production of extracellular vesicles. Pathogens. 2020; 9: 144. doi: 10.3390/pathogens9020144.
https://doi.org/10.3390/pathogens9020144...
,161161. Castro-Artavia E, Retana-Moreira L, Lorenzo-Morales J, Abrahams-Sandí E. Potentially pathogenic Acanthamoeba genotype T4 isolated from dental units and emergency combination showers. Mem Inst Oswaldo Cruz. 2017; 112(12): 817-21. doi: 10.1590/0074-02760170147.
https://doi.org/10.1590/0074-02760170147...
,162162. Alvarado-Ocampo J, Retana-Moreira L, Abrahams-Sandí E. In vitro effects of environmental isolates of Acanthamoeba T4 and T5 over human erythrocytes and platelets. Exp Parasitol. 2020; 210: 107842. doi: 10.1016/j.exppara.2020.107842.
https://doi.org/10.1016/j.exppara.2020.1...
including the secretion of EVs with serine and cysteine protease activity in Acanthamoeba T5.160160. Moreira LR, Ramírez DV, Linares F, Ledezma AP, Garro AV, Osuna A, et al. Isolation of Acanthamoeba T5 from water: characterization of its pathogenic potential, including the production of extracellular vesicles. Pathogens. 2020; 9: 144. doi: 10.3390/pathogens9020144.
https://doi.org/10.3390/pathogens9020144...
Coincidentally, another study found that serine proteases are predominant in four strains of environmental (genotypes T1, T2 and T11) and clinical (genotype T4) origin.163163. Costa AO, Chagas IAR, de 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. 2021; 45: 1060-71. doi: 10.1002/cbin.11551.
https://doi.org/10.1002/cbin.11551...
Aminopeptidase activity has also been determined in EVs of Acanthamoeba.164164. Lin WC, Tsai CY, Huang JM, Wu SR, Chu LJ, Huang KY. Quantitative proteomic analysis and functional characterization of Acanthamoeba castellanii exosome-like vesicles. Parasit Vectors. 2019; 12: 467. doi: 10.1186/s13071-019-3725-z.
https://doi.org/10.1186/s13071-019-3725-...

A previous study with Acanthamoeba castellanii described evaginating vesicles from plasma membrane using SEM, and great diversity in mean diameter estimations (Table): 117 nm by TEM and 287,7 and 365,1 nm using DLS,165165. Gonçalves DS, Ferreira MS, 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. 2018; 9(1): 818-36. doi: 10.1080/21505594.2018.1451184.
https://doi.org/10.1080/21505594.2018.14...
a range that embraces sizes reported in posterior works (166,7 nm using NTA).164164. Lin WC, Tsai CY, Huang JM, Wu SR, Chu LJ, Huang KY. Quantitative proteomic analysis and functional characterization of Acanthamoeba castellanii exosome-like vesicles. Parasit Vectors. 2019; 12: 467. doi: 10.1186/s13071-019-3725-z.
https://doi.org/10.1186/s13071-019-3725-...
When analysing two culture conditions through a qualitative proteomic characterisation of the secretome (one in rich medium PYG and the other under nutritional stress), most of the proteins belonged to the miscellaneous or undefined categories.165165. Gonçalves DS, Ferreira MS, 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. 2018; 9(1): 818-36. doi: 10.1080/21505594.2018.1451184.
https://doi.org/10.1080/21505594.2018.14...
However, the exoproteome under stress identified more proteins related to cellular stress and oxidative, protein and amino acid metabolism, with a rich enzymatic profile for carbohydrate metabolism (amylases, glycosyl hydrolases, alpha-1,4-glucan phosphorylases),165165. Gonçalves DS, Ferreira MS, 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. 2018; 9(1): 818-36. doi: 10.1080/21505594.2018.1451184.
https://doi.org/10.1080/21505594.2018.14...
which draws attention for its potential use in biotechnological applications.166166. Gonçalves DS, Ferreira MS, Guimarães AJ. Extracellular vesicles from the protozoa Acanthamoeba castellanii: their role in pathogenesis, environmental adaptation and potential applications. Bioengineering. 2019; 6: 13. doi: 10.3390/bioengineering6010013.
https://doi.org/10.3390/bioengineering60...
While, in abundance, more locomotion and signalling proteins were found,165165. Gonçalves DS, Ferreira MS, 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. 2018; 9(1): 818-36. doi: 10.1080/21505594.2018.1451184.
https://doi.org/10.1080/21505594.2018.14...
other proteomic analyses of quantitative type support that the largest families of proteins found are hydrolases and oxidoreductases.164164. Lin WC, Tsai CY, Huang JM, Wu SR, Chu LJ, Huang KY. Quantitative proteomic analysis and functional characterization of Acanthamoeba castellanii exosome-like vesicles. Parasit Vectors. 2019; 12: 467. doi: 10.1186/s13071-019-3725-z.
https://doi.org/10.1186/s13071-019-3725-...
On the other hand, characterisation of lipid composition has shown the presence of sterols, phospholipids, free fatty acids, and sterol esters.165165. Gonçalves DS, Ferreira MS, 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. 2018; 9(1): 818-36. doi: 10.1080/21505594.2018.1451184.
https://doi.org/10.1080/21505594.2018.14...

TABLE
Isolation and characterisation methods commonly employed for the collection of extracellular vesicles (EVs) from extracellular protozoan pathogens, including sizes ranges reported in literature

EVs of A. castellanii have been shown to interact with different cell lines such as Chinese hamster ovary (CHO) cells, glioblastoma T98G and rat glial C6 cells, adhering to the membrane and terminating in all cases with their internalisation. Likewise, in vitro cytopathic effect assays have yielded positive results.164164. Lin WC, Tsai CY, Huang JM, Wu SR, Chu LJ, Huang KY. Quantitative proteomic analysis and functional characterization of Acanthamoeba castellanii exosome-like vesicles. Parasit Vectors. 2019; 12: 467. doi: 10.1186/s13071-019-3725-z.
https://doi.org/10.1186/s13071-019-3725-...
,165165. Gonçalves DS, Ferreira MS, 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. 2018; 9(1): 818-36. doi: 10.1080/21505594.2018.1451184.
https://doi.org/10.1080/21505594.2018.14...
It has been further determined that A. castellanii EVs are also able to elicit an immune response in THP-1 cells, after detecting the expression and production of IL-6, IL-12164164. Lin WC, Tsai CY, Huang JM, Wu SR, Chu LJ, Huang KY. Quantitative proteomic analysis and functional characterization of Acanthamoeba castellanii exosome-like vesicles. Parasit Vectors. 2019; 12: 467. doi: 10.1186/s13071-019-3725-z.
https://doi.org/10.1186/s13071-019-3725-...
and TNF-α.163163. Costa AO, Chagas IAR, de 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. 2021; 45: 1060-71. doi: 10.1002/cbin.11551.
https://doi.org/10.1002/cbin.11551...
In murine macrophages, activation level after the stimulation with EVs of Acanthamoeba has been measured through the production of NO, demonstrating that, of those tested, the main receptor is TLR4, followed by TLR2.163163. Costa AO, Chagas IAR, de 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. 2021; 45: 1060-71. doi: 10.1002/cbin.11551.
https://doi.org/10.1002/cbin.11551...
Protease inhibitors have exerted a negative effect on both, the concretion of the cytopathic effect,165165. Gonçalves DS, Ferreira MS, 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. 2018; 9(1): 818-36. doi: 10.1080/21505594.2018.1451184.
https://doi.org/10.1080/21505594.2018.14...
as well as NO production,163163. Costa AO, Chagas IAR, de 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. 2021; 45: 1060-71. doi: 10.1002/cbin.11551.
https://doi.org/10.1002/cbin.11551...
pointing to a preponderant role of these as virulence factors associated with EVs.

Naegleria fowleri - The infectious disease given by N. fowleri, primary amoebic meningoencephalitis, is a severe fulminant pathology with high mortality rate, in which the amoeba employs contact-dependent (adhesion and phagocytosis) and contact-independent (matrix metalloproteinases and pore-forming proteins) mechanisms to produce brain tissue damage and destruction.167167. Güémez A, García E. Primary amoebic meningoencephalitis by Naegleria fowleri: pathogenesis and treatments. Biomolecules. 2021; 11: 1320. doi: 10.3390/biom11091320.
https://doi.org/10.3390/biom11091320...

Pathophysiological mechanisms are under constant review and two pioneer investigation groups have confirmed the production of EVs by trophozoites of this amoeba. In this sense, it has been reported cup-shaped vesicles observed via TEM, comprising two subpopulations of 156,8 nm and 141 nm;168168. Lertjuthaporn S, Somkird J, Lekmanee K, Atipimonpat A, Sukapirom K, Sawasdipokin H, et al. Extracellular vesicles from Naegleria fowleri induce IL-8 response in THP-1 macrophage. Pathogens. 2022; 11: 632. doi: 10.3390/pathogens11060632.
https://doi.org/10.3390/pathogens1106063...
a more comprehensive characterisation of these EVs was performed by Retana Moreira et al.,169169. Retana-Moreira L, Espinoza MFS, Camacho NC, Cornet-Gomez A, Sáenz-Arce G, Osuna A, et al. Characterization of extracellular vesicles secreted by a clinical isolate of Naegleria fowleri and identification of immunogenic components within their protein cargo. Biology. 2022; 11: 983. doi: 10.3390/biology11070983.
https://doi.org/10.3390/biology11070983...
who measured size through TEM, NTA and DLS, obtaining means ranging from 24,24 nm to 227,13 nm (Table). Z-potential of -12,228 mV was also determined.169169. Retana-Moreira L, Espinoza MFS, Camacho NC, Cornet-Gomez A, Sáenz-Arce G, Osuna A, et al. Characterization of extracellular vesicles secreted by a clinical isolate of Naegleria fowleri and identification of immunogenic components within their protein cargo. Biology. 2022; 11: 983. doi: 10.3390/biology11070983.
https://doi.org/10.3390/biology11070983...
Then, clustered release of EVs was reported by Russell et al.170170. Russell AC, Bush P, Grigorean G, Kyle DE. Characterization of the extracellular vesicles, ultrastructural morphology, and intercellular interactions of multiple clinical isolates of the brain-eating amoeba, Naegleria fowleri. Front Microbiol. 2023; 14: 1264348. doi: 10.3389/fmicb.2023.1264348.
https://doi.org/10.3389/fmicb.2023.12643...
and Retana Moreira et al.171171. Retana-Moreira L, Cornet-Gomez A, Sepulveda MR, Molina-Castro S, Alvarado-Ocampo J, Monge FC, et al. Providing an in vitro depiction of microglial cells challenged with immunostimulatory extracellular vesicles of Naegleria fowleri. Front Microbiol. 2024; 15: 1346021. doi: 10.3389/fmicb.2024.1346021.
https://doi.org/10.3389/fmicb.2024.13460...

Proteome analysis has found almost half of proteins are still uncharacterised, but also identified actin, Rho GTPases, dehydrogenases, and two potential pathogenesis-related factors: leucin aminopeptidase and fowlerpain (a cysteine protease).169169. Retana-Moreira L, Espinoza MFS, Camacho NC, Cornet-Gomez A, Sáenz-Arce G, Osuna A, et al. Characterization of extracellular vesicles secreted by a clinical isolate of Naegleria fowleri and identification of immunogenic components within their protein cargo. Biology. 2022; 11: 983. doi: 10.3390/biology11070983.
https://doi.org/10.3390/biology11070983...
Besides, protease activity of EVs of N. fowleri has been found, mainly by serine proteases, although to a lesser extent than the whole trophozoite extract.169169. Retana-Moreira L, Espinoza MFS, Camacho NC, Cornet-Gomez A, Sáenz-Arce G, Osuna A, et al. Characterization of extracellular vesicles secreted by a clinical isolate of Naegleria fowleri and identification of immunogenic components within their protein cargo. Biology. 2022; 11: 983. doi: 10.3390/biology11070983.
https://doi.org/10.3390/biology11070983...
Afterwards, Russell et al.170170. Russell AC, Bush P, Grigorean G, Kyle DE. Characterization of the extracellular vesicles, ultrastructural morphology, and intercellular interactions of multiple clinical isolates of the brain-eating amoeba, Naegleria fowleri. Front Microbiol. 2023; 14: 1264348. doi: 10.3389/fmicb.2023.1264348.
https://doi.org/10.3389/fmicb.2023.12643...
identified 2270 proteins, 150 of which overlapped with Retana Moreira et al.169169. Retana-Moreira L, Espinoza MFS, Camacho NC, Cornet-Gomez A, Sáenz-Arce G, Osuna A, et al. Characterization of extracellular vesicles secreted by a clinical isolate of Naegleria fowleri and identification of immunogenic components within their protein cargo. Biology. 2022; 11: 983. doi: 10.3390/biology11070983.
https://doi.org/10.3390/biology11070983...
findings.

Regarding functional analysis, cellular effects of N. fowleri EVs have been featured by PKH26-monitored internalisation in the THP-1 monocytic cell line, with no subsequent apoptosis and stimulation of IL-8 gene expression, cytokine that was later identified 48 h and 72 h post-activation of macrophages.168168. Lertjuthaporn S, Somkird J, Lekmanee K, Atipimonpat A, Sukapirom K, Sawasdipokin H, et al. Extracellular vesicles from Naegleria fowleri induce IL-8 response in THP-1 macrophage. Pathogens. 2022; 11: 632. doi: 10.3390/pathogens11060632.
https://doi.org/10.3390/pathogens1106063...
Uptake by other mammalian cells (e.g., Vero, HFF, A549, B103 rat neuroblastoma cells) and other amoebae has been proved via EVs-R18 staining.170170. Russell AC, Bush P, Grigorean G, Kyle DE. Characterization of the extracellular vesicles, ultrastructural morphology, and intercellular interactions of multiple clinical isolates of the brain-eating amoeba, Naegleria fowleri. Front Microbiol. 2023; 14: 1264348. doi: 10.3389/fmicb.2023.1264348.
https://doi.org/10.3389/fmicb.2023.12643...
A cytokine/chemokine proinflammatory profile was described on BV-2 microglial cells stimulated by N. fowleri EVs, showing the possibility of a contact independent immunopathogenic mechanism.172172. Lê HG, Kang JM, Võ TC, Yoo WG, Na BK. Naegleria fowleri extracellular vesicles induce proinflammatory immune responses in BV-2 microglial cells. Int J Mol Sci. 2023; 24: 13623. doi: 10.3390/ijms241713623.
https://doi.org/10.3390/ijms241713623...

In this sense, our group has just confirmed the induction of diverse effectors (e.g., iNOS, IL-6, IL-23, TNF-α, IL-10) on primary microglia and BV-2 cells by EVs secreted by trophozoites of two clinic isolates of Naegleria fowleri. We also noted morphological changes in cells to an amoeboid-like morphology after the contact with these vesicles. Moreover, specific N. fowleri DNA was found in EVs fractions, according to our quantitative polymerase chain reaction (qPCR) results,171171. Retana-Moreira L, Cornet-Gomez A, Sepulveda MR, Molina-Castro S, Alvarado-Ocampo J, Monge FC, et al. Providing an in vitro depiction of microglial cells challenged with immunostimulatory extracellular vesicles of Naegleria fowleri. Front Microbiol. 2024; 15: 1346021. doi: 10.3389/fmicb.2024.1346021.
https://doi.org/10.3389/fmicb.2024.13460...
a promising finding for diagnostic purposes.

Limitations and future perspectives

There are still many biological questions regarding EVs and their purposes; whether they respond to a stimulus, a selective process or an incidental release must be elucidated.173173. Drurey C, Maizels RM. Helminth extracellular vesicles: Interactions with the host immune system. Mol Immunol. 2021; 137: 124-33. doi: 10.1016/j.molimm.2021.06.017.
https://doi.org/10.1016/j.molimm.2021.06...
,174174. Olajide JS, Cai J. Perils and promises of pathogenic protozoan extracellular vesicles. Front Cell Infect Microbiol. 2020; 10: 371. doi: 10.3389/fcimb.2020.00371.
https://doi.org/10.3389/fcimb.2020.00371...
In parasites of medical importance, it remains to be clarified if the change in the profile of biomolecules depends on the parasite stage and what mechanisms of cargo manipulation exist in pathophysiological contexts to lead to more or less virulence.1919. Carrera-Bravo C, Koh EY, Tan KSW. The roles of parasite-derived extracellular vesicles in disease and host-parasite communication. Parasitol Int. 2021; 83: 102373. doi: 10.1016/j.parint.2021.102373.
https://doi.org/10.1016/j.parint.2021.10...
,175175. Alfandari D, Cadury S, Morandi MI, Regev-Rudzki N. Transforming parasites into their own foes: parasitic extracellular vesicles as a vaccine platform. Trends Parasitol. 2023; 39(11): 913-28. doi: 10.1016/j.pt.2023.08.009.
https://doi.org/10.1016/j.pt.2023.08.009...

The discovery and description of the interactions between EVs and host cells supposes the integration of new knowledge in the understanding of the phenomenon of parasitism. Furthermore, as cellular inducers, EVs immunomodulation has been widely proven. In fact, in biomedical application, the advantages offered using EVs as platforms for immunisation are being studied since they could represent stable carriers of various antigens, which would prevent the development of tolerance. However, aspects of logistics, formulation, safety, and effectiveness in suitable models cannot be ignored given still unpredictable responses.175175. Alfandari D, Cadury S, Morandi MI, Regev-Rudzki N. Transforming parasites into their own foes: parasitic extracellular vesicles as a vaccine platform. Trends Parasitol. 2023; 39(11): 913-28. doi: 10.1016/j.pt.2023.08.009.
https://doi.org/10.1016/j.pt.2023.08.009...

ACKNOWLEDGEMENTS

To all former students that have trusted our research group for developing their skills in parasitology and studies of extracellular vesicles.

REFERENCES

  • 1
    Heijnen H, Schiel AE, Fijnheer R, Geuze HJ, Sixma JJ. Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and a-granules. Blood. 1999; 94(11): 3791-9.
  • 2
    Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015; 25(6): 364-72. doi: 10.1016/j.tcb.2015.01.004.
    » https://doi.org/10.1016/j.tcb.2015.01.004
  • 3
    Mathieu M, Névo N, Jouve M, Valenzuela JI, Maurin M, Verweij FJ, et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nat Commun. 2021; 12: 4389. doi: 10.1038/s41467-021-24384-2.
    » https://doi.org/10.1038/s41467-021-24384-2
  • 4
    Cruz Camacho A, Alfandari D, Kozela E, Regev-Rudzki N. Biogenesis of extracellular vesicles in protozoan parasites: the ESCRT complex in the trafficking fast lane? PLoS Pathog. 2023; 19(2): e1011140. doi: 10.1371/journal.ppat.1011140.
    » https://doi.org/10.1371/journal.ppat.1011140
  • 5
    Abels ER, Breakefield XO. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 2016; 36(3): 301-12. doi: 10.1007/s10571-016-0366-z.
    » https://doi.org/10.1007/s10571-016-0366-z
  • 6
    Théry C, Boussac M, Véron P, Ricciardi-Castagnoli P, Raposo G, Garin J, et al. Proteomic analysis of dendritic cell-derived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J Immunol. 2001; 166: 7309-18. doi: 10.4049/jimmunol.166.12.7309.
    » https://doi.org/10.4049/jimmunol.166.12.7309
  • 7
    Palmisano G, Jensen SS, Le Bihan MC, Lainé J, McGuire JN, Pociot F, et al. Characterization of membrane-shed microvesicles from cytokine-stimulated ß-cells using proteomics strategies. Mol Cell Proteomics. 2012; 11(8): 230-43. doi: 10.1074/mcp.M111.012732.
    » https://doi.org/10.1074/mcp.M111.012732
  • 8
    Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles. 2024; 13: e12404. doi: 10.1002/jev2.12404.
    » https://doi.org/10.1002/jev2.12404
  • 9
    Doyle LM, Wang MZ. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 2019; 8: 727. doi: 10.3390/cells8070727.
    » https://doi.org/10.3390/cells8070727
  • 10
    Chulpanova DS, Kitaeva KV, James V, Rizvanov AA, Solovyeva VV. Therapeutic prospects of extracellular vesicles in cancer treatment. Front Immunol. 2018; 9: 1534. doi: 10.3389/fimmu.2018.01534.
    » https://doi.org/10.3389/fimmu.2018.01534
  • 11
    Kok VC, Yu CC. Cancer-derived exosomes: their role in cancer biology and biomarker development. Int J Nanomedicine. 2020; 15: 8019-36. doi: 10.2147/IJN.S272378.
    » https://doi.org/10.2147/IJN.S272378
  • 12
    Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016; 126(4): 1208-15. doi: 10.1172/JCI81135.
    » https://doi.org/10.1172/JCI81135
  • 13
    Kodam SP, Ullah M. Diagnostic and therapeutic potential of extracellular vesicles. Technol Cancer Res Treat. 2021; 20: 1-10. doi: 10.1177/15330338211041203.
    » https://doi.org/10.1177/15330338211041203
  • 14
    Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep. 2015; 16(1): 24-43. doi: 10.15252/embr.201439363.
    » https://doi.org/10.15252/embr.201439363
  • 15
    Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013; 200(4): 373-83. doi: 10.1083/jcb.201211138.
    » https://doi.org/10.1083/jcb.201211138
  • 16
    Bielska E, May RC. Extracellular vesicles of human pathogenic fungi. Curr Opin Microbiol. 2019; 52: 90-9. doi: 10.1016/j.mib.2019.05.007.
    » https://doi.org/10.1016/j.mib.2019.05.007
  • 17
    Kim JH, Lee J, Park J, Gho YS. Gram-negative and Gram-positive bacterial extracellular vesicles. Semin Cell Dev Biol. 2015; 40: 97-104. doi: 10.1016/j.semcdb.2015.02.006.
    » https://doi.org/10.1016/j.semcdb.2015.02.006
  • 18
    Urbanelli L, Buratta S, Tancini B, Sagini K, Delo F, Porcellati S, et al. The role of extracellular vesicles in viral infection and transmission. Vaccines. 2019; 7: 102. doi: 10.3390/vaccines7030102.
    » https://doi.org/10.3390/vaccines7030102
  • 19
    Carrera-Bravo C, Koh EY, Tan KSW. The roles of parasite-derived extracellular vesicles in disease and host-parasite communication. Parasitol Int. 2021; 83: 102373. doi: 10.1016/j.parint.2021.102373.
    » https://doi.org/10.1016/j.parint.2021.102373
  • 20
    Ofir-Birin Y, Heidenreich M, Regev-Rudzki N. Pathogen-derived extracellular vesicles coordinate social behaviour and host manipulation. Semin Cell Dev Biol. 2017; 67: 83-90. doi: 10.1016/j.semcdb.2017.03.004.
    » https://doi.org/10.1016/j.semcdb.2017.03.004
  • 21
    Duncan L, Yoshioka M, Chandad F, Grenier D. Loss of lipopolysaccharide receptor CD14 from the surface of human macrophage-like cells mediated by Porphyromonas gingivalis outer membrane vesicles. Microb Pathog. 2004; 36: 319-25. doi: 10.1016/j.micpath.2004.02.004.
    » https://doi.org/10.1016/j.micpath.2004.02.004
  • 22
    Bhatnagar S, Shinagawa K, Castellino FJ, Schorey JS. Exosomes released from macrophages infected with intracellular pathogens stimulate a proinflammatory response in vitro and in vivo. Blood. 2007; 110(9): 3234-44. doi: 10.1182/blood-2007-03-079152.
    » https://doi.org/10.1182/blood-2007-03-079152
  • 23
    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. 2014; 193(2): 71-4. doi: 10.1016/j.molbiopara.2014.02.004.
    » https://doi.org/10.1016/j.molbiopara.2014.02.004
  • 24
    Cipriano MJ, Hajduk SL. Drivers of persistent infection: pathogen-induced extracellular vesicles. Essays Biochem. 2018; 62: 135-47. doi: 10.1042/EBC20170083.
    » https://doi.org/10.1042/EBC20170083
  • 25
    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. 2014; 3: 25040. doi: 10.3402/jev.v3.25040.
    » https://doi.org/10.3402/jev.v3.25040
  • 26
    Arenaccio C, Anticoli S, Manfredi F, Chiozzini C, Olivetta E, Federico M. Latent HIV-1 is activated by exosomes from cells infected with either replication-competent or defective HIV-1. Retrovirology. 2015; 12: 87. doi: 10.1186/s12977-015-0216-y.
    » https://doi.org/10.1186/s12977-015-0216-y
  • 27
    Mack M, Kleinschmidt A, Bruhl H, Klier C, Nelson P, Cihak J, et al. Transfer of the chemokine receptor CCR5 between cells by membrane-derived microparticles: a mechanism for cellular human immunodeficiency virus 1 infection. Nat Med. 2000; 6(7): 769-75.
  • 28
    Hassanpour M, Rezaie J, Nouri M, Panahi Y. The role of extracellular vesicles in COVID-19 virus infection. Infect Genet Evol. 2020; 85: 104422. doi: 10.1016/j.meegid.2020.104422.
    » https://doi.org/10.1016/j.meegid.2020.104422
  • 29
    Bukong TN, Momen-Heravi F, Kodys K, Bala S, Szabo G. Exosomes from Hepatitis C infected patients transmit HCV infection and contain replication competent viral RNA in complex with Ago2-miR122-HSP90. PLoS Pathog. 2014; 10(10): e1004424. doi: 10.1371/journal.ppat.1004424.
    » https://doi.org/10.1371/journal.ppat.1004424
  • 30
    Longatti A, Boyd B, Chisari FV. Virion-independent transfer of replication-competent Hepatitis C virus RNA between permissive cells. J Virol. 2015; 89(5): 2956-61. doi: 10.1128/jvi.02721-14.
    » https://doi.org/10.1128/jvi.02721-14
  • 31
    Kim YS, Choi EJ, Lee WH, Choi SJ, Roh TY, Park J, et al. Extracellular vesicles, especially derived from Gram-negative bacteria, in indoor dust induce neutrophilic pulmonary inflammation associated with both Th1 and Th17 cell responses. Clin Exp Allergy. 2013; 43: 443-54. doi: 10.1111/cea.12085.
    » https://doi.org/10.1111/cea.12085
  • 32
    Yang D, Chen X, Wang J, Lou Q, Lou Y, Li L, et al. Dysregulated lung commensal bacteria drive interleukin-17B production to promote pulmonary fibrosis through their outer membrane vesicles. Immunity. 2019; 50(3): 692-706. doi: 10.1016/j.immuni.2019.02.001.
    » https://doi.org/10.1016/j.immuni.2019.02.001
  • 33
    Briaud P, Carroll RK. Extracellular vesicle biogenesis and functions in gram-positive bacteria. Infect Immun. 2020; 8(12): e00433-20. doi: 10.1128/IAI.00433-20.
    » https://doi.org/10.1128/IAI.00433-20
  • 34
    Kulp AJ, Sun B, Ai T, Manning AJ, Orench-Rivera N, Schmid AK, et al. Genome-wide assessment of outer membrane vesicle production in Escherichia coli. PLoS One. 2015; 10(9): e0139200. doi: 10.1371/journal.pone.0139200.
    » https://doi.org/10.1371/journal.pone.0139200
  • 35
    Nevermann J, Silva A, Otero C, Oyarzún DP, Barrera B, Gil F, et al. Identification of genes involved in biogenesis of outer membrane vesicles (OMVs) in Salmonella enterica Serovar Typhi. Front Microbiol. 2019; 10: 104. doi: 10.3389/fmicb.2019.00104.
    » https://doi.org/10.3389/fmicb.2019.00104
  • 36
    Roier S, Zingl FG, Cakar F, Durakovic S, Kohl P, Eichmann TO, et al. A novel mechanism for the biogenesis of outer membrane vesicles in Gram-negative bacteria. Nat Commun. 2016; 7: 10515. doi: 10.1038/ncomms10515.
    » https://doi.org/10.1038/ncomms10515
  • 37
    Schlatterer K, Beck C, Hanzelmann D, Lebtig M, Fehrenbacher B, Schaller M, et al. The mechanism behind bacterial lipoprotein release: Phenol-soluble modulins mediate toll-like receptor 2 activation via extracellular vesicle release from Staphylococcus aureus. mBio. 2018; 9(6): e01851-18. doi: 10.1128/mbio.01851-18.
    » https://doi.org/10.1128/mbio.01851-18
  • 38
    Turnbull L, Toyofuku M, Hynen AL, Kurosawa M, Pessi G, Petty NK, et al. Explosive cell lysis as a mechanism for the biogenesis of bacterial membrane vesicles and biofilms. Nat Commun. 2016; 7: 11220. doi: 10.1038/ncomms11220.
    » https://doi.org/10.1038/ncomms11220
  • 39
    Wang X, Thompson CD, Weidenmaier C, Lee JC. Release of Staphylococcus aureus extracellular vesicles and their application as a vaccine platform. Nat Commun. 2018; 9: 1379. doi: 10.1038/s41467-018-03847-z.
    » https://doi.org/10.1038/s41467-018-03847-z
  • 40
    Schaar V, De Vries SPW, Perez Vidakovics MLA, Bootsma HJ, Larsson L, Hermans PWM, et al. Multicomponent Moraxella catarrhalis outer membrane vesicles induce an inflammatory response and are internalized by human epithelial cells. Cell Microbiol. 2011; 13(3): 432-49. doi: 10.1111/j.1462-5822.2010.01546.x.
    » https://doi.org/10.1111/j.1462-5822.2010.01546.x
  • 41
    Van Bergenhenegouwen J, Kraneveld AD, Rutten L, Kettelarij N, Garssen J, Vos AP. Extracellular vesicles modulate host-microbe responses by altering TLR2 activity and phagocytosis. PLoS One. 2014; 9(2): e89121. doi: 10.1371/journal.pone.0089121.
    » https://doi.org/10.1371/journal.pone.0089121
  • 42
    Ciofu O, Beveridge TJ, Kadurugamuwa J, Walther-Rasmussen J, Høiby N. Chromosomal ß-lactamase is packaged into membrane vesicles and secreted from Pseudomonas aeruginosa. J Antimicrob Chemother. 2000; 45: 9-13.
  • 43
    Rivera J, Cordero RJB, Nakouzi AS, Frases S, Nicola A, Casadevall A. Bacillus anthracis produces membrane-derived vesicles containing biologically active toxins. Proc Natl Acad Sci USA. 2010; 107(44): 19002-7. doi: 10.1073/pnas.1008843107.
    » https://doi.org/10.1073/pnas.1008843107
  • 44
    Vanaja SK, Russo AJ, Behl B, Banerjee I, Yankova M, Deshmukh SD, et al. Bacterial outer membrane vesicles mediate cytosolic localization of LPS and Caspase-11 activation. Cell. 2016; 165: 1106-19. doi: 10.1016/j.cell.2016.04.015.
    » https://doi.org/10.1016/j.cell.2016.04.015
  • 45
    Ñahui Palomino RA, Vanpouille C, Costantini PE, Margolis L. Microbiota-host communications: bacterial extracellular vesicles as a common language. PLoS Pathog. 2021; 17(5): e1009508. doi: 10.1371/journal.ppat.1009508.
    » https://doi.org/10.1371/journal.ppat.1009508
  • 46
    Artuyants A, Campos TL, Rai AK, Johnson PJ, Dauros-Singorenko P, Phillips A, et al. Extracellular vesicles produced by the protozoan parasite Trichomonas vaginalis contain a preferential cargo of tRNA-derived small RNAs. Int J Parasitol. 2020; 50: 1145-55. doi: 10.1016/j.ijpara.2020.07.003.
    » https://doi.org/10.1016/j.ijpara.2020.07.003
  • 47
    Brown L, Wolf JM, Prados-Rosales R, Casadevall A. Through the wall: extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi. Nat Rev Microbiol. 2015; 13(10): 620-30. doi: 10.1038/nrmicro3480.
    » https://doi.org/10.1038/nrmicro3480
  • 48
    Lieberman LA. Outer membrane vesicles: a bacterial-derived vaccination system. Front Microbiol. 2022; 13: 1029146. doi: 10.3389/fmicb.2022.1029146.
    » https://doi.org/10.3389/fmicb.2022.1029146
  • 49
    Rodrigues ML, Nimrichter L, Oliveira DL, Frases S, Miranda K, Zaragoza O, et al. Vesicular polysaccharide export in Cryptococcus neoformans is a eukaryotic solution to the problem of fungal trans-cell wall transport. Eukaryot Cell. 2007; 6(1): 48-59. doi: 10.1128/EC.00318-06.
    » https://doi.org/10.1128/EC.00318-06
  • 50
    Ikeda MAK, De Almeida JRF, Jannuzzi GP, Cronemberger-Andrade A, Torrecilhas ACT, Moretti NS, et al. 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. doi: 10.3389/fmicb.2018.02286.
    » https://doi.org/10.3389/fmicb.2018.02286
  • 51
    Liebana-Jordan M, Brotons B, Falcon-Perez JM, Gonzalez E. Extracellular vesicles in the fungi kingdom. Int J Mol Sci. 2021; 22: 7221. doi: 10.3390/ijms22137221.
    » https://doi.org/10.3390/ijms22137221
  • 52
    Rizzo J, Chaze T, Miranda K, Roberson RW, Gorgette O, Nimrichter L, et al. Characterization of extracellular vesicles produced by Aspergillus fumigatus protoplasts. mSphere. 2020; 5(4): e00476-20. doi: 10.1128/msphere.00476-20.
    » https://doi.org/10.1128/msphere.00476-20
  • 53
    Albuquerque PC, Nakayasu ES, Rodrigues ML, Frases S, Casadevall A, Zancope-Oliveira RM, et al. Vesicular transport in Histoplasma capsulatum: an effective mechanism for trans-cell wall transfer of proteins and lipids in ascomycetes. Cell Microbiol. 2008; 10(8): 1695-710. doi: 10.1111/j.1462-5822.2008.01160.x.
    » https://doi.org/10.1111/j.1462-5822.2008.01160.x
  • 54
    Piffer AC, Kuczera D, Rodrigues ML, Nimrichter L. The paradoxical and still obscure properties of fungal extracellular vesicles. Mol Immunol. 2021; 135: 137-46. doi: 10.1016/j.molimm.2021.04.009.
    » https://doi.org/10.1016/j.molimm.2021.04.009
  • 55
    Evans-Osses I, Reichembach LH, Ramirez MI. Exosomes or microvesicles? Two kinds of extracellular vesicles with different routes to modify protozoan-host cell interaction. Parasitol Res. 2015; 114: 3567-75. doi: 10.1007/s00436-015-4659-9.
    » https://doi.org/10.1007/s00436-015-4659-9
  • 56
    Barteneva NS, Maltsev N, Vorobjev IA. Microvesicles and intercellular communication in the context of parasitism. Front Cell Infect Microbiol. 2013; 3: 49. doi: 10.3389/fcimb.2013.00049.
    » https://doi.org/10.3389/fcimb.2013.00049
  • 57
    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; 13(3): e1006245. doi: 10.1371/journal.ppat.1006245.
    » https://doi.org/10.1371/journal.ppat.1006245
  • 58
    Sharma M, Lozano-Amado D, Chowdhury D, Singh U. Extracellular Vesicles and Their impact on the biology of protozoan parasites. Trop Med Infect Dis. 2023: 8: 448. doi: 10.3390/tropicalmed8090448.
    » https://doi.org/10.3390/tropicalmed8090448
  • 59
    Nawaz M, Malik MI, Hameed M, Zhou J. Research progress on the composition and function of parasite-derived exosomes. Acta Trop. 2019; 196: 30-6. doi: 10.1016/j.actatropica.2019.05.004.
    » https://doi.org/10.1016/j.actatropica.2019.05.004
  • 60
    Wang Y, Yuan W, Kimber M, Lu M, Dong L. Rapid differentiation of host and parasitic exosome vesicles using microfluidic photonic crystal biosensor. ACS Sens. 2018; 3: 1616-21. doi: 10.1021/acssensors.8b00360.
    » https://doi.org/10.1021/acssensors.8b00360
  • 61
    Kifle DW, Chaiyadet S, Waardenberg AJ, Wise I, Cooper M, Becker L, et al. Uptake of Schistosoma mansoni extracellular vesicles by human endothelial and monocytic cell lines and impact on vascular endothelial cell gene expression. Int J Parasitol. 2020; 50: 685-96. doi: 10.1016/j.ijpara.2020.05.005.
    » https://doi.org/10.1016/j.ijpara.2020.05.005
  • 62
    Chaiyadet S, Sotillo J, Krueajampa W, Thongsen S, Smout M, Brindley PJ, et al. Silencing of Opisthorchis viverrini tetraspanin gene expression results in reduced secretion of extracellular vesicles. Front Cell Infect Microbiol. 2022; 12: 827521. doi: 10.3389/fcimb.2022.827521.
    » https://doi.org/10.3389/fcimb.2022.827521
  • 63
    Wu Z, Wang L, Li J, Wang L, Wu Z, Sun X. Extracellular vesicle-mediated communication within host-parasite interactions. Front Immunol. 2019; 9: 3066. doi: 10.3389/fimmu.2018.03066.
    » https://doi.org/10.3389/fimmu.2018.03066
  • 64
    Sultana H, Neelakanta G. Arthropod exosomes as bubbles with message(s) to transmit vector-borne diseases. Curr Opin Insect Sci. 2020; 40: 39-47. doi: 10.1016/j.cois.2020.05.017.
    » https://doi.org/10.1016/j.cois.2020.05.017
  • 65
    Hackenberg M, Kotsyfakis M. Exosome-mediated pathogen transmission by arthropod vectors. Trends Parasitol. 2018; 34(7): 549-52. doi: 10.1016/j.pt.2018.04.001.
    » https://doi.org/10.1016/j.pt.2018.04.001
  • 66
    Zhou W, Woodson M, Neupane B, Bai F, Sherman MB, Choi KH, et al. Exosomes serve as novel modes of tick-borne flavivirus transmission from arthropod to human cells and facilitates dissemination of viral RNA and proteins to the vertebrate neuronal cells. PLoS Pathog. 2018; 14(1): e1006764. doi: 10.1371/journal.ppat.1006764.
    » https://doi.org/10.1371/journal.ppat.1006764
  • 67
    Vora A, Zhou W, Londono-Renteria B, Woodson M, Sherman MB, Colpitts TM, et al. Arthropod EVs mediate dengue virus transmission through interaction with a tetraspanin domain containing glycoprotein Tsp29Fb. Proc Natl Acad Sci USA. 2018; 115(28): E6604-13. doi: 10.1073/pnas.1720125115.
    » https://doi.org/10.1073/pnas.1720125115
  • 68
    Reyes-Ruiz JM, Osuna-Ramos JF, De Jesús-González LA, Hurtado-Monzón AM, Farfan-Morales CN, Cervantes-Salazar M, et al. Isolation and characterization of exosomes released from mosquito cells infected with dengue virus. Virus Res. 2019; 266: 1-14. doi: 10.1016/j.virusres.2019.03.015.
    » https://doi.org/10.1016/j.virusres.2019.03.015
  • 69
    Oliva Chávez AS, Wang X, Marnin L, Archer NK, Hammond HL, Carroll EEMC, et al. Tick extracellular vesicles enable arthropod feeding and promote distinct outcomes of bacterial infection. Nat Commun. 2021; 12: 3696. doi: 10.1038/s41467-021-23900-8.
    » https://doi.org/10.1038/s41467-021-23900-8
  • 70
    Butler LR, Gonzalez J, Pedra JHF, Chavez ASO. Tick extracellular vesicles in host skin immunity and pathogen transmission. Trends Parasitol. 2023; 39(10): 873-85. doi: 10.1016/j.pt.2023.07.009.
    » https://doi.org/10.1016/j.pt.2023.07.009
  • 71
    Loghry HJ, Kwon H, Smith RC, Sondjaja NA, Minkler SJ, Young S, et al. Extracellular vesicles secreted by Brugia malayi microfilariae modulate the melanization pathway in the mosquito host. Sci Rep. 2023; 13: 8778. doi: 10.1038/s41598-023-35940-9.
    » https://doi.org/10.1038/s41598-023-35940-9
  • 72
    Yang T, Xu Z, Yu J, Liu J, Wang W, Hong S. Exosomes derived from Dermatophagoides farinae induce allergic airway inflammation. Microbiol Spectr. 2023; 11(4): 1-19. doi: 10.1128/spectrum.05054-22.
    » https://doi.org/10.1128/spectrum.05054-22
  • 73
    Duque-Correa MA, Schreiber F, Rodgers FH, Goulding D, Forrest S, White R, et al. Development of caecaloids to study host-pathogen interactions: new insights into immunoregulatory functions of Trichuris muris extracellular vesicles in the caecum. Int J Parasitol. 2020; 50: 707-18. doi: 10.1016/j.ijpara.2020.06.001.
    » https://doi.org/10.1016/j.ijpara.2020.06.001
  • 74
    Harischandra H, Yuan W, Loghry HJ, Zamanian M, Kimber MJ. Profiling extracellular vesicle release by the filarial nematode Brugia malayi reveals sex-specific differences in cargo and a sensitivity to ivermectin. PLoS Negl Trop Dis. 2018; 12(4): e0006438. doi: 10.1371/journal.pntd.0006438.
    » https://doi.org/10.1371/journal.pntd.0006438
  • 75
    de la Torre-Escudero E, Gerlach JQ, Bennett APS, Cwiklinski K, Jewhurst HL, Huson KM, et al. Surface molecules of extracellular vesicles secreted by the helminth pathogen Fasciola hepatica direct their internalisation by host cells. PLoS Negl Trop Dis. 2019; 13(1): e0007087. doi: 10.1371/journal.pntd.0007087.
    » https://doi.org/10.1371/journal.pntd.0007087
  • 76
    Di Maggio LS, Fischer K, Yates D, Curtis KC, Rosa BA, Martin J, et al. The proteome of extracellular vesicles of the lung fluke Paragonimus kellicotti produced in vitro and in the lung cyst. Sci Rep. 2023; 13: 13726. doi: 10.1038/s41598-023-39966-x.
    » https://doi.org/10.1038/s41598-023-39966-x
  • 77
    Liu C, Cao J, Zhang H, Field MC, Yin J. Extracellular vesicles secreted by Echinococcus multilocularis: important players in angiogenesis promotion. Microbes Infect. 2023; 25: 105147. doi: 10.1016/j.micinf.2023.105147.
    » https://doi.org/10.1016/j.micinf.2023.105147
  • 78
    Kosanovic M, Cvetkovic J, Gruden-Movsesijan A, Vasilev S, Svetlana M, Ilic N, et al. Trichinella spiralis muscle larvae release extracellular vesicles with immunomodulatory properties. Parasite Immunol. 2019; 41: e12665. doi: 10.1111/pim.12665.
    » https://doi.org/10.1111/pim.12665
  • 79
    Murphy A, Cwiklinski K, Lalor R, O'connell B, Robinson MW, Gerlach J, et al. Fasciola hepatica extracellular vesicles isolated from excretory-secretory products using a gravity flow method modulate dendritic cell phenotype and activity. PLoS Negl Trop Dis. 2020; 14(9): e0008626. doi: 10.1371/journal.pntd.0008626.
    » https://doi.org/10.1371/journal.pntd.0008626
  • 80
    Sánchez-López CM, González-Arce A, Soler C, Ramírez-Toledo V, Trelis M, Bernal D, et al. Extracellular vesicles from the trematodes Fasciola hepatica and Dicrocoelium dendriticum trigger different responses in human THP-1 macrophages. J Extracell Vesicles. 2023; 12(4): e12317. doi: 10.1002/jev2.12317.
    » https://doi.org/10.1002/jev2.12317
  • 81
    Ricciardi A, Bennuru S, Tariq S, Kaur S, Wu W, Elkahloun AG, et al. Extracellular vesicles released from the filarial parasite Brugia malayi downregulate the host mTOR pathway. PLoS Negl Trop Dis. 2021; 15(1): e0008884. doi: 10.1371/journal.pntd.0008884.
    » https://doi.org/10.1371/journal.pntd.0008884
  • 82
    Meningher T, Lerman G, Regev-Rudzki N, Gold D, Ben-Dov IZ, Sidi Y, et al. Schistosomal microRNAs isolated from extracellular vesicles in sera of infected patients: a new tool for diagnosis and follow-up of human schistosomiasis. J Infect Dis. 2017; 215: 378-86. doi: 10.1093/infdis/jiw539.
    » https://doi.org/10.1093/infdis/jiw539
  • 83
    Eichenberger RM, Talukder MH, Field MA, Wangchuk P, Giacomin P, Loukas A, et al. Characterization of Trichuris muris secreted proteins and extracellular vesicles provides new insights into host-parasite communication. J Extracell Vesicles. 2018; 7: 1428004. doi: 10.1080/20013078.2018.1428004.
    » https://doi.org/10.1080/20013078.2018.1428004
  • 84
    Eichenberger RM, Ryan S, Jones L, Buitrago G, Polster R, de Oca MM, et al. Hookworm secreted extracellular vesicles interact with host cells and prevent inducible colitis in mice. Front Immunol. 2018; 9: 850. doi: 10.3389/fimmu.2018.00850.
    » https://doi.org/10.3389/fimmu.2018.00850
  • 85
    Hansen EP, Fromm B, Andersen SD, Marcilla A, Andersen KL, Borup A, et al. Exploration of extracellular vesicles from Ascaris suum provides evidence of parasite-host cross talk. J Extracell Vesicles. 2019; 8: 1578116. doi: 10.1080/20013078.2019.1578116.
    » https://doi.org/10.1080/20013078.2019.1578116
  • 86
    Drurey C, Coakley G, Maizels RM. Extracellular vesicles: new targets for vaccines against helminth parasites. Int J Parasitol. 2020; 50: 623-33. doi: 10.1016/j.ijpara.2020.04.011.
    » https://doi.org/10.1016/j.ijpara.2020.04.011
  • 87
    Sheng ZA, Wu CL, Wang DY, Zhong SH, Yang X, Rao GS, et al. Proteomic analysis of exosome-like vesicles from Fasciola gigantica adult worm provides support for new vaccine targets against fascioliasis. Parasit Vectors. 2023; 16: 62. doi: 10.1186/s13071-023-05659-7.
    » https://doi.org/10.1186/s13071-023-05659-7
  • 88
    Gonzáles WHR, Coelho GR, Pimenta DC, de Paula FM, Gryschek RCB. Proteomic analysis of the excretory-secretory products from Strongyloides venezuelensis infective larvae: new insights for the immunodiagnosis of human strongyloidiasis. Parasitol Res. 2022; 121(11): 3155-70. doi: 10.1007/s00436-022-07636-y.
    » https://doi.org/10.1007/s00436-022-07636-y
  • 89
    Li C, Li C, Xu F, Wang H, Jin X, Zhang Y, et al. Identification of antigens in the Trichinella spiralis extracellular vesicles for serological detection of early stage infection in swine. Parasit Vectors. 2023; 16: 387. doi: 10.1186/s13071-023-06013-7.
    » https://doi.org/10.1186/s13071-023-06013-7
  • 90
    Babatunde KA, Subramanian BY, Ahouidi AD, Murillo PM, Walch M, Mantel PY. Role of extracellular vesicles in cellular cross talk in malaria. Front Immunol. 2020; 11: 22. doi: 10.3389/fimmu.2020.00022.
    » https://doi.org/10.3389/fimmu.2020.00022
  • 91
    Varikuti S, Jha BK, Holcomb EA, McDaniel JC, Karpurapu M, Srivastava N, et al. The role of vascular endothelium and exosomes in human protozoan parasitic diseases. Vessel Plus. 2020; 4: 28. doi: 10.20517/2574-1209.2020.27.
    » https://doi.org/10.20517/2574-1209.2020.27
  • 92
    de Souza W, Barrias ES. Membrane-bound extracellular vesicles secreted by parasitic protozoa: cellular structures involved in the communication between cells. Parasitol Res. 2020; 119: 2005-23. doi: 10.1007/s00436-020-06691-7.
    » https://doi.org/10.1007/s00436-020-06691-7
  • 93
    Kato K, Sugi T, Iwanaga T. Roles of Apicomplexan protein kinases at each life cycle stage. Parasitol Int. 2012; 61: 224-34. doi: 10.1016/j.parint.2011.12.002.
    » https://doi.org/10.1016/j.parint.2011.12.002
  • 94
    Rossi IV, Nunes MAF, Vargas-Otalora S, Ferreira TCS, Cortez M, Ramirez MI. Extracellular vesicles during TriTryps infection: complexity and future challenges. Mol Immunol. 2021; 132: 172-83. doi: 10.1016/j.molimm.2021.01.008.
    » https://doi.org/10.1016/j.molimm.2021.01.008
  • 95
    Abdi A, Yu L, Goulding D, Rono MK, Bejon P, Choudhary J, et al. Proteomic analysis of extracellular vesicles from a Plasmodium falciparum Kenyan clinical isolate defines a core parasite secretome. Wellcome Open Res. 2017; 2: 50. doi: 10.12688/wellcomeopenres.11910.1.
    » https://doi.org/10.12688/wellcomeopenres.11910.1
  • 96
    Vimonpatranon S, Roytrakul S, Phaonakrop N, Lekmanee K, Atipimonpat A, Srimark N, et al. Extracellular vesicles derived from early and late stage Plasmodium falciparum-infected red blood cells contain invasion-associated proteins. J Clin Med. 2022; 11: 4250. doi: 10.3390/jcm11144250.
    » https://doi.org/10.3390/jcm11144250
  • 97
    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. 2018; 13(10): 1157-68. doi: 10.2217/nnm-2018-0035.
    » https://doi.org/10.2217/nnm-2018-0035
  • 98
    Quiarim TM, Maia MM, da Cruz AB, Taniwaki NN, Namiyama GM, Pereira-Chioccola VL. Characterization of extracellular vesicles isolated from types I, II and III strains of Toxoplasma gondii. Acta Trop. 2021; 219: 105915. doi: 10.1016/j.actatropica.2021.105915.
    » https://doi.org/10.1016/j.actatropica.2021.105915
  • 99
    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. 2018; 40(9): e12571. doi: 10.1111/pim.12571.
    » https://doi.org/10.1111/pim.12571
  • 100
    Gómez-Chávez F, Murrieta-Coxca JM, Caballero-Ortega H, Morales-Prieto DM, Markert UR. Host-pathogen interactions mediated by extracellular vesicles in Toxoplasma gondii infection during pregnancy. J Reprod Immunol. 2023; 158: 103957. doi: 10.1016/j.jri.2023.103957.
    » https://doi.org/10.1016/j.jri.2023.103957
  • 101
    Maia MM, da Cruz AB, Taniwaki NN, Namiyama GM, Gava R, Gomes AHS, et al. Immunization with extracellular vesicles excreted by Toxoplasma gondii confers protection in murine infection, activating cellular and humoral responses. Int J Parasitol. 2021; 51: 559-69. doi: 10.1016/j.ijpara.2020.11.010.
    » https://doi.org/10.1016/j.ijpara.2020.11.010
  • 102
    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: e190217. doi: 10.1590/0074-02760190217.
    » https://doi.org/10.1590/0074-02760190217
  • 103
    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. 2015; 13(5): 957-67. doi: 10.1016/j.celrep.2015.09.058.
    » https://doi.org/10.1016/j.celrep.2015.09.058
  • 104
    Serafim TD, Coutinho-Abreu IV, Dey R, Kissinger R, Valenzuela JG, Oliveira F, et al. Leishmaniasis: the act of transmission. Trends Parasitol. 2021; 37(11): 976-87. doi: 10.1016/j.pt.2021.07.003.
    » https://doi.org/10.1016/j.pt.2021.07.003
  • 105
    Barbosa FMC, Dupin TV, Toledo MS, Reis NFC, Ribeiro K, Cronemberger-Andrade A, et al. Extracellular vesicles released by Leishmania (Leishmania) amazonensis promote disease progression and induce the production of different cytokines in macrophages and B-1 Cells. Front Microbiol. 2018; 9: 3056. doi: 10.3389/fmicb.2018.03056.
    » https://doi.org/10.3389/fmicb.2018.03056
  • 106
    Silverman JM, Clos J, Horakova E, Wang AY, Wiesgigl M, Kelly I, et al. Leishmania exosomes modulate innate and adaptive immune responses through effects on monocytes and dendritic cells. J Immunol. 2010; 185: 5011-22. doi: 10.4049/jimmunol.1000541.
    » https://doi.org/10.4049/jimmunol.1000541
  • 107
    Dong G, Wagner V, Minguez-Menendez A, Fernandez-Prada C, Olivier M. Extracellular vesicles and leishmaniasis: current knowledge and promising avenues for future development. Mol Immunol. 2021; 135: 73-83. doi: 10.1016/j.molimm.2021.04.003.
    » https://doi.org/10.1016/j.molimm.2021.04.003
  • 108
    Hassani K, Shio MT, Martel C, Faubert D, Olivier M. Absence of metalloprotease GP63 alters the protein content of Leishmania exosomes. PLoS One. 2014; 9(4): e95007. doi: 10.1371/journal.pone.0095007.
    » https://doi.org/10.1371/journal.pone.0095007
  • 109
    Lambertz U, Ovando MEO, Vasconcelos EJR, 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. 2015; 16: 151. doi: 10.1186/s12864-015-1260-7.
    » https://doi.org/10.1186/s12864-015-1260-7
  • 110
    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; 19: e12672. doi: 10.1111/cmi.12672.
    » https://doi.org/10.1111/cmi.12672
  • 111
    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; 13(2): e0007163. doi: 10.1371/journal.pntd.0007163.
    » https://doi.org/10.1371/journal.pntd.0007163
  • 112
    Torrecilhas ACT, 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; 11: 29-39. doi: 10.1016/j.micinf.2008.10.003.
    » https://doi.org/10.1016/j.micinf.2008.10.003
  • 113
    Lozano IMD, 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; 7: 44451. doi: 10.1038/srep44451.
    » https://doi.org/10.1038/srep44451
  • 114
    Choudhuri S, Garg NJ. PARP1-cGAS-NF-?B pathway of proinflammatory macrophage activation by extracellular vesicles released during Trypanosoma cruzi infection and Chagas disease. PLoS Pathog. 2020; 16(4): e1008474. doi: 10.1371/journal.ppat.1008474.
    » https://doi.org/10.1371/journal.ppat.1008474
  • 115
    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; 22: 5183. doi: 10.3390/ijms22105183.
    » https://doi.org/10.3390/ijms22105183
  • 116
    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. doi: 10.3389/fcimb.2020.602502.
    » https://doi.org/10.3389/fcimb.2020.602502
  • 117
    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. 2022; 40: 111121. doi: 10.1016/j.celrep.2022.111121.
    » https://doi.org/10.1016/j.celrep.2022.111121
  • 118
    Rossi IV, Nunes MAF, Sabatke B, Ribas HT, Winnischofer SMB, Ramos ASP, et al. An induced population of Trypanosoma cruzi epimastigotes more resistant to complement lysis promotes a phenotype with greater differentiation, invasiveness, and release of extracellular vesicles. Front Cell Infect Microbiol. 2022; 12: 1046681. doi: 10.3389/fcimb.2022.1046681.
    » https://doi.org/10.3389/fcimb.2022.1046681
  • 119
    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; 164(0): 246-57. doi: 10.1016/j.cell.2015.11.051.
    » https://doi.org/10.1016/j.cell.2015.11.051
  • 120
    Dias-Guerreiro T, Palma-Marques J, Mourata-Gonçalves P, Alexandre-Pires G, Valério-Bolas A, Gabriel A, et al. African trypanosomiasis: extracellular vesicles shed by Trypanosoma brucei brucei manipulate host mononuclear cells. Biomedicines. 2021; 9: 1056. doi: 10.3390/biomedicines9081056.
    » https://doi.org/10.3390/biomedicines9081056
  • 121
    Fernandez-Becerra C, Xander P, Alfandari D, Dong G, Aparici-Herraiz I, Rosenhek-Goldian I, et al. Guidelines for the purification and characterization of extracellular vesicles of parasites. J Extracell Biol. 2023; 2: e117. doi: 10.1002/jex2.117.
    » https://doi.org/10.1002/jex2.117
  • 122
    Buret AG. Pathophysiology of enteric infections with Giardia duodenalis. Parasite. 2008; 15: 261-5. doi: 10.1051/parasite/2008153261.
    » https://doi.org/10.1051/parasite/2008153261
  • 123
    Lanfredi-Rangel A, Attias M, Reiner DS, Gillin FD, De Souza W. Fine structure of the biogenesis of Giardia lamblia encystation secretory vesicles. J Struct Biol. 2003; 143: 153-63. doi: 10.1016/S1047-8477(03)00123-0.
    » https://doi.org/10.1016/S1047-8477(03)00123-0
  • 124
    Benchimol M. The release of secretory vesicle in encysting Giardia lamblia. FEMS Microbiol Lett. 2004; 235: 81-7. doi: 10.1016/j.femsle.2004.04.014.
    » https://doi.org/10.1016/j.femsle.2004.04.014
  • 125
    Ma'ayeh SY, Liu J, Peirasmaki D, Hörnaeus K, Lind SB, Grabherr M, et al. Characterization of the Giardia intestinalis secretome during interaction with human intestinal epithelial cells: the impact on host cells. PLoS Negl Trop Dis. 2017; 11(12): e0006120. doi: 10.1371/journal.pntd.0006120.
    » https://doi.org/10.1371/journal.pntd.0006120
  • 126
    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. 2017; 96: 131-42. doi: 10.1016/j.ejcb.2017.01.005.
    » https://doi.org/10.1016/j.ejcb.2017.01.005
  • 127
    Sana A, Rossi IV, Sabatke B, Bonato LB, Medeiros LCS, Ramirez MI. An improved method to enrich large extracellular vesicles derived from Giardia intestinalis through differential centrifugation. Life. 2023; 13: 1799. doi: 10.3390/life13091799.
    » https://doi.org/10.3390/life13091799
  • 128
    Faria CP, Ferreira B, Lourenço A, Guerra I, Melo T, Domingues P, et al. Lipidome of extracellular vesicles from Giardia lamblia. PLoS One. 2023; 18(9): e0291292. doi: 10.1371/journal.pone.0291292.
    » https://doi.org/10.1371/journal.pone.0291292
  • 129
    Siddiq A, Allain T, Dong G, Olivier M, Buret A. Giardia extracellular vesicles disrupt intestinal epithelial and inhibit the growth of commensal bacterial while increasing their swimming motility. FASEB J. 2020; 34(S1): 1-1. doi: 10.1096/fasebj.2020.34.s1.00515.
    » https://doi.org/10.1096/fasebj.2020.34.s1.00515
  • 130
    Zhao P, Cao L, Wang X, Dong J, Zhang N, Li X, et al. Extracellular vesicles secreted by Giardia duodenalis regulate host cell innate immunity via TLR2 and NLRP3 inflammasome signaling pathways. PLoS Negl Trop Dis. 2021; 15(4): e0009304. doi: 10.1371/journal.pntd.0009304.
    » https://doi.org/10.1371/journal.pntd.0009304
  • 131
    Gavinho B, Sabatke B, Feijoli V, Rossi IV, da Silva JM, Evans-Osses I, et al. Peptidyl arginine 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. 2020; 10: 417. doi: 10.3389/fcimb.2020.00417.
    » https://doi.org/10.3389/fcimb.2020.00417
  • 132
    Ferreira B, Lourenço A, Sousa MC. Protozoa-derived extracellular vesicles on intercellular communication with special emphasis on Giardia lamblia. Microorganisms. 2022; 10: 2422. doi: 10.3390/microorganisms10122422.
    » https://doi.org/10.3390/microorganisms10122422
  • 133
    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. 2018; 97: 44-62. doi: 10.1016/j.ejcb.2017.11.004.
    » https://doi.org/10.1016/j.ejcb.2017.11.004
  • 134
    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. 2019; 207: 301-11. doi: 10.1016/j.jsb.2019.07.002.
    » https://doi.org/10.1016/j.jsb.2019.07.002
  • 135
    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. 2019; 8: 1600. doi: 10.3390/cells8121600.
    » https://doi.org/10.3390/cells8121600
  • 136
    Zhao P, Cao L, Wang X, Li J, Dong J, Zhang N, et al. Giardia duodenalis extracellular vesicles regulate the proinflammatory immune response in mouse macrophages in vitro via the MAPK, AKT and NF-?B pathways. Parasit Vectors. 2021; 14: 358. doi: 10.1186/s13071-021-04865-5.
    » https://doi.org/10.1186/s13071-021-04865-5
  • 137
    Siddiq A, Dong G, Balan B, Harrison LG, Jex A, Olivier M, et al. A thermo-resistant and RNase-sensitive cargo from Giardia duodenalis extracellular vesicles modifies the behaviour of enterobacteria. J Extracell Biol. 2023; 2: e109. doi: 10.1002/jex2.109.
    » https://doi.org/10.1002/jex2.109
  • 138
    Siddiq A, Allain T, Dong G, Olivier M, Buret A. Role of extracellular vesicles in Giardia-microbiota interactions. J Can Assoc Gastroenterol. 2021; 4(S1): 226-8. doi: 10.1093/jcag/gwab002.200.
    » https://doi.org/10.1093/jcag/gwab002.200
  • 139
    Poole DN, McClelland RS. Global epidemiology of Trichomonas vaginalis. Sex Transm Infect. 2013; 89(6): 418-22. doi: 10.1136/sextrans-2013-051075.
    » https://doi.org/10.1136/sextrans-2013-051075
  • 140
    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. PLoS Pathog. 2013; 9(7): e1003482. doi: 10.1371/journal.ppat.1003482.
    » https://doi.org/10.1371/journal.ppat.1003482
  • 141
    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. 2018; 75: 2211-26. doi: 10.1007/s00018-017-2726-3.
    » https://doi.org/10.1007/s00018-017-2726-3
  • 142
    Ong SC, Cheng WH, Ku FM, Tsai CY, Huang PJ, Lee CC, et al. Identification of endosymbiotic virus in small extracellular vesicles derived from Trichomonas vaginalis. Genes. 2022; 13: 531. doi: 10.3390/genes13030531.
    » https://doi.org/10.3390/genes13030531
  • 143
    Salas N, Coceres VM, Melo TS, 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. 2022; 79: 11. doi: 10.1007/s00018-021-04083-3.
    » https://doi.org/10.1007/s00018-021-04083-3
  • 144
    Rada P, Hrdý I, Zdrha A, Narayanasamy RK, Smutná T, Horácková J, et al. Double-stranded RNA viruses are released from Trichomonas vaginalis inside small extracellular vesicles and modulate the exosomal cargo. Front Microbiol. 2022; 13: 893692. doi: 10.3389/fmicb.2022.893692.
    » https://doi.org/10.3389/fmicb.2022.893692
  • 145
    Salas N, Pedreros MB, Melo TS, Maguire VG, Sha J, Wohlschlegel JA, et al. Role of cytoneme structures and extracellular vesicles in Trichomonas vaginalis parasite: parasite communication. Elife. 2023; 12. doi: 10.7554/eLife.86067.
  • 146
    Rai AK, Johnson PJ. Trichomonas vaginalis extracellular vesicles are internalized by host cells using proteoglycans and caveolin-dependent endocytosis. Proc Natl Acad Sci USA. 2019; 116(43): 21354-60. doi: 10.1073/pnas.1912356116.
    » https://doi.org/10.1073/pnas.1912356116
  • 147
    Molgora BM, Mukherjee SK, Baumel-Alterzon S, Santiago FM, Muratore KA, Sisk AE, et al. Trichomonas vaginalis adherence phenotypes and extracellular vesicles impact parasite survival in a novel in vivo model of pathogenesis. PLoS Negl Trop Dis. 2023; 17(10): e00011693. doi: 10.1371/journal.pntd.0011693.
    » https://doi.org/10.1371/journal.pntd.0011693
  • 148
    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. 2017; 39(6): e12426. doi: 10.1111/pim.12426.
    » https://doi.org/10.1111/pim.12426
  • 149
    Tatischeff I, Bomsel M, De Paillerets C, Durand H, Geny B, Segretain D, et al. Dictyostelium discoideum cells shed vesicles with associated DNA and vital stain Hoechst 33342. Cell Mol Life Sci. 1998; 54: 476-87.
  • 150
    Tatischeff I. Dictyostelium: a model for studying the extracellular vesicle messengers involved in human health and disease. Cells. 2019; 8: 225. doi: 10.3390/cells8030225.
    » https://doi.org/10.3390/cells8030225
  • 151
    Dey R, Folkins MA, Ashbolt NJ. Extracellular amoebal-vesicles: potential transmission vehicles for respiratory viruses. NPJ Biofilms Microbiomes. 2021; 7: 25. doi: 10.1038/s41522-021-00201-y.
    » https://doi.org/10.1038/s41522-021-00201-y
  • 152
    Ashbolt NJ. Conceptual model to inform Legionella-amoebae control, including the roles of extracellular vesicles in engineered water system infections. Front Cell Infect Microbiol. 2023; 13: 1200478. doi: 10.3389/fcimb.2023.1200478.
    » https://doi.org/10.3389/fcimb.2023.1200478
  • 153
    Christy NCV, Petri WA. Mechanisms of adherence, cytotoxicity and phagocytosis modulate the pathogenesis of Entamoeba histolytica. Future Microbiol. 2011; 6(12): 1501-19. doi: 10.2217/fmb.11.120.
    » https://doi.org/10.2217/fmb.11.120
  • 154
    Ujang JA, Kwan SH, Ismail MN, Lim BH, Noordin R, Othman N. Proteome analysis of excretory-secretory proteins of Entamoeba histolytica HM1:IMSS via LC-ESI-MS/MS and LC-MALDI-TOF/TOF. Clin Proteomics. 2016; 13: 33. doi: 10.1186/s12014-016-9135-8.
    » https://doi.org/10.1186/s12014-016-9135-8
  • 155
    Perdomo D, Aït-Ammar N, Syan S, Sachse M, Jhingan GD, Guillén N. Data set for the proteomics analysis of the endomembrane system from the unicellular Entamoeba histolytica. J Proteomics. 2014; 112(2015): 125-40. doi: 10.1016/j.dib.2014.08.007.
    » https://doi.org/10.1016/j.dib.2014.08.007
  • 156
    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. Infect Immun. 2020; 88: 00349-20. doi: 10.1128/IAI.00349-20.
    » https://doi.org/10.1128/IAI.00349-20
  • 157
    Díaz-Godínez C, Ríos-Valencia DG, García-Aguirre S, Martínez-Calvillo S, Carrero JC. Immunomodulatory effect of extracellular vesicles from Entamoeba histolytica trophozoites: regulation of NETs and respiratory burst during confrontation with human neutrophils. Front Cell Infect Microbiol. 2022; 12: 1018314. doi: 10.3389/fcimb.2022.1018314.
    » https://doi.org/10.3389/fcimb.2022.1018314
  • 158
    Sharma M, Zhang H, Ehrenkaufer G, Singh U. Stress response in Entamoeba histolytica is associated with robust processing of tRNA to tRNA halves. mBio. 2023; 14(2): 1-16. doi: 10.1128/mbio.03450-22.
    » https://doi.org/10.1128/mbio.03450-22
  • 159
    Maciver SK, Asif M, Simmen MW, Lorenzo-Morales J. A systematic analysis of Acanthamoeba genotype frequency correlated with source and pathogenicity: T4 is confirmed as a pathogen-rich genotype. Eur J Protistol. 2013; 49: 217-21. doi: 10.1016/j.ejop.2012.11.004.
    » https://doi.org/10.1016/j.ejop.2012.11.004
  • 160
    Moreira LR, Ramírez DV, Linares F, Ledezma AP, Garro AV, Osuna A, et al. Isolation of Acanthamoeba T5 from water: characterization of its pathogenic potential, including the production of extracellular vesicles. Pathogens. 2020; 9: 144. doi: 10.3390/pathogens9020144.
    » https://doi.org/10.3390/pathogens9020144
  • 161
    Castro-Artavia E, Retana-Moreira L, Lorenzo-Morales J, Abrahams-Sandí E. Potentially pathogenic Acanthamoeba genotype T4 isolated from dental units and emergency combination showers. Mem Inst Oswaldo Cruz. 2017; 112(12): 817-21. doi: 10.1590/0074-02760170147.
    » https://doi.org/10.1590/0074-02760170147
  • 162
    Alvarado-Ocampo J, Retana-Moreira L, Abrahams-Sandí E. In vitro effects of environmental isolates of Acanthamoeba T4 and T5 over human erythrocytes and platelets. Exp Parasitol. 2020; 210: 107842. doi: 10.1016/j.exppara.2020.107842.
    » https://doi.org/10.1016/j.exppara.2020.107842
  • 163
    Costa AO, Chagas IAR, de 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. 2021; 45: 1060-71. doi: 10.1002/cbin.11551.
    » https://doi.org/10.1002/cbin.11551
  • 164
    Lin WC, Tsai CY, Huang JM, Wu SR, Chu LJ, Huang KY. Quantitative proteomic analysis and functional characterization of Acanthamoeba castellanii exosome-like vesicles. Parasit Vectors. 2019; 12: 467. doi: 10.1186/s13071-019-3725-z.
    » https://doi.org/10.1186/s13071-019-3725-z
  • 165
    Gonçalves DS, Ferreira MS, 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. 2018; 9(1): 818-36. doi: 10.1080/21505594.2018.1451184.
    » https://doi.org/10.1080/21505594.2018.1451184
  • 166
    Gonçalves DS, Ferreira MS, Guimarães AJ. Extracellular vesicles from the protozoa Acanthamoeba castellanii: their role in pathogenesis, environmental adaptation and potential applications. Bioengineering. 2019; 6: 13. doi: 10.3390/bioengineering6010013.
    » https://doi.org/10.3390/bioengineering6010013
  • 167
    Güémez A, García E. Primary amoebic meningoencephalitis by Naegleria fowleri: pathogenesis and treatments. Biomolecules. 2021; 11: 1320. doi: 10.3390/biom11091320.
    » https://doi.org/10.3390/biom11091320
  • 168
    Lertjuthaporn S, Somkird J, Lekmanee K, Atipimonpat A, Sukapirom K, Sawasdipokin H, et al. Extracellular vesicles from Naegleria fowleri induce IL-8 response in THP-1 macrophage. Pathogens. 2022; 11: 632. doi: 10.3390/pathogens11060632.
    » https://doi.org/10.3390/pathogens11060632
  • 169
    Retana-Moreira L, Espinoza MFS, Camacho NC, Cornet-Gomez A, Sáenz-Arce G, Osuna A, et al. Characterization of extracellular vesicles secreted by a clinical isolate of Naegleria fowleri and identification of immunogenic components within their protein cargo. Biology. 2022; 11: 983. doi: 10.3390/biology11070983.
    » https://doi.org/10.3390/biology11070983
  • 170
    Russell AC, Bush P, Grigorean G, Kyle DE. Characterization of the extracellular vesicles, ultrastructural morphology, and intercellular interactions of multiple clinical isolates of the brain-eating amoeba, Naegleria fowleri. Front Microbiol. 2023; 14: 1264348. doi: 10.3389/fmicb.2023.1264348.
    » https://doi.org/10.3389/fmicb.2023.1264348
  • 171
    Retana-Moreira L, Cornet-Gomez A, Sepulveda MR, Molina-Castro S, Alvarado-Ocampo J, Monge FC, et al. Providing an in vitro depiction of microglial cells challenged with immunostimulatory extracellular vesicles of Naegleria fowleri. Front Microbiol. 2024; 15: 1346021. doi: 10.3389/fmicb.2024.1346021.
    » https://doi.org/10.3389/fmicb.2024.1346021
  • 172
    Lê HG, Kang JM, Võ TC, Yoo WG, Na BK. Naegleria fowleri extracellular vesicles induce proinflammatory immune responses in BV-2 microglial cells. Int J Mol Sci. 2023; 24: 13623. doi: 10.3390/ijms241713623.
    » https://doi.org/10.3390/ijms241713623
  • 173
    Drurey C, Maizels RM. Helminth extracellular vesicles: Interactions with the host immune system. Mol Immunol. 2021; 137: 124-33. doi: 10.1016/j.molimm.2021.06.017.
    » https://doi.org/10.1016/j.molimm.2021.06.017
  • 174
    Olajide JS, Cai J. Perils and promises of pathogenic protozoan extracellular vesicles. Front Cell Infect Microbiol. 2020; 10: 371. doi: 10.3389/fcimb.2020.00371.
    » https://doi.org/10.3389/fcimb.2020.00371
  • 175
    Alfandari D, Cadury S, Morandi MI, Regev-Rudzki N. Transforming parasites into their own foes: parasitic extracellular vesicles as a vaccine platform. Trends Parasitol. 2023; 39(11): 913-28. doi: 10.1016/j.pt.2023.08.009.
    » https://doi.org/10.1016/j.pt.2023.08.009

Publication Dates

  • Publication in this collection
    23 Sept 2024
  • Date of issue
    2024

History

  • Received
    01 Apr 2024
  • Accepted
    09 July 2024
Instituto Oswaldo Cruz, Ministério da Saúde Av. Brasil, 4365 - Pavilhão Mourisco, Manguinhos, 21040-900 Rio de Janeiro RJ Brazil, Tel.: (55 21) 2562-1222, Fax: (55 21) 2562 1220 - Rio de Janeiro - RJ - Brazil
E-mail: memorias@fiocruz.br