Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Exp Parasitol ; 231: 108173, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34742714

ABSTRACT

Extracellular vesicles (EVs) from parasitic helminths play an important role in immunomodulation. However, EVs are little studied in the important parasite Fasciola gigantica. Here the ability of EVs from F. gigantica to induce cellular response to stress (reactive oxygen species generation, autophage and DNA damage response) in human intrahepatic biliary epithelial cells (HIBEC) was investigated. F. gigantica-derived EVs were isolated by ultracentrifugation, and identified with transmission electron microscopy, nanoparticle size analysis and parasite-derived EV markers. Internalization of EVs by HIBEC was determined by confocal immunofluorescence microscopy and flow cytometry. ROS levels in HIBEC were detected by molecular probing. EVs-induced autophagy and DNA-damaging effects were determined by evaluating expression levels of light chain 3B protein (LC3B), phosphor- H2A.X and phosphor-Chk1, respectively. Results revealed that EVs with sizes predominately ranging from 39 to 110 nm in diameter were abundant in adult F. gigantica and contained the parasite-derived marker proteins enolase and 14-3-3, and EVs were internalized by HIBEC. Further, uptake of EVs into HIBEC was associated with increased levels of reactive oxygen species, LC3Ⅱ, phosphor-H2A.X and phosphor-Chk1, suggesting EVs are likely to induce autophagy and DNA damage & repair processes. These results indicate F. gigantica EVs are associated with modulations of host cell responses and have a potential important role in the host-parasite interactions.


Subject(s)
Extracellular Vesicles/physiology , Fasciola/physiology , Immunomodulation/physiology , Animals , Antibodies, Helminth/immunology , Antibodies, Helminth/isolation & purification , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Autophagy/physiology , Blotting, Western , Buffaloes/parasitology , Cell Line , Extracellular Vesicles/parasitology , Fasciola/ultrastructure , Flow Cytometry , Host-Parasite Interactions , Humans , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Liver/parasitology , Microscopy, Confocal , Microscopy, Fluorescence , Rabbits , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism
3.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068436

ABSTRACT

Extracellular vesicles (EVs) are small lipid vesicles released by either any prokaryotic or eukaryotic cell, or both, with a biological role in cell-to-cell communication. In this work, we characterize the proteomes and nanomechanical properties of EVs released by tissue-culture cell-derived trypomastigotes (mammalian infective stage; (TCT)) and epimastigotes (insect stage; (E)) of Trypanosoma cruzi, the etiologic agent of Chagas disease. EVs of each stage were isolated by differential centrifugation and analyzed using liquid chromatography with tandem mass spectrometry (LC-MS/MS), dynamic light scattering (DLS), nanoparticle tracking analysis (NTA), electron microscopy and atomic force microscopy (AFM). Measurements of zeta-potential were also included. Results show marked differences in the surface molecular cargos of EVs between both stages, with a noteworthy expansion of all groups of trans-sialidase proteins in trypomastigote's EVs. In contrast, chromosomal locations of trans-sialidases of EVs of epimastigotes were dramatically reduced and restricted to subtelomeric regions, indicating a possible regulatable expression of these proteins between both stages of the parasite. Regarding mechanical properties, EVs of trypomastigotes showed higher adhesion compared to the EVs of epimastigotes. These findings demonstrate the remarkable surface remodeling throughout the life cycle of T. cruzi, which shapes the physicochemical composition of the extracellular vesicles and could have an impact in the ability of these vesicles to participate in cell communication in completely different niches of infection.


Subject(s)
Chagas Disease/metabolism , Extracellular Vesicles/metabolism , Life Cycle Stages , Proteome/metabolism , Protozoan Proteins/metabolism , Trypanosoma cruzi/metabolism , Animals , Chagas Disease/parasitology , Chlorocebus aethiops , Extracellular Vesicles/parasitology , Host-Parasite Interactions , Male , Mice , Mice, Inbred BALB C , Proteome/analysis , Vero Cells
4.
Trends Parasitol ; 37(8): 690-693, 2021 08.
Article in English | MEDLINE | ID: mdl-34154932

ABSTRACT

Helminth infections impact the composition of the mammalian gut microbiota; however, the mechanisms underpinning these interactions are, thus far, unknown. In this article, we propose that microbiota-derived extracellular vesicles might represent key players in host-helminth-microbiome crosstalk, and outline future directions to elucidate their role(s) in host-parasite relationships.


Subject(s)
Extracellular Vesicles/metabolism , Extracellular Vesicles/parasitology , Gastrointestinal Microbiome/physiology , Helminthiasis/microbiology , Helminthiasis/parasitology , Host-Parasite Interactions , Animals , Helminths/physiology , Humans
5.
Front Immunol ; 12: 662944, 2021.
Article in English | MEDLINE | ID: mdl-33959131

ABSTRACT

Extracellular vesicles (EVs) have garnered significant interest in recent years due to their contributions to cell-to-cell communication and disease processes. EVs are composed of a complex profile of bioactive molecules, which include lipids, nucleic acids, metabolites, and proteins. Although the biogenesis of EVs released by cells under various normal and abnormal conditions has been well-studied, there is incomplete knowledge about how infection influences EV biogenesis. EVs from infected cells contain specific molecules of both host and pathogen origin that may contribute to pathogenesis and the elicitation of the host immune response. Intracellular pathogens exhibit diverse lifestyles that undoubtedly dictate the mechanisms by which their molecules enter the cell's exosome biogenesis schemes. We will discuss the current understanding of the mechanisms used during infection to traffic molecules from their vacuolar niche to host EVs by selected intravacuolar pathogens. We initially review general exosome biogenesis schemes and then discuss what is known about EV biogenesis in Mycobacterium, Plasmodium, Toxoplasma, and Leishmania infections, which are pathogens that reside within membrane delimited compartments in phagocytes at some time in their life cycle within mammalian hosts. The review includes discussion of the need for further studies into the biogenesis of EVs to better understand the contributions of these vesicles to host-pathogen interactions, and to uncover potential therapeutic targets to control these pathogens.


Subject(s)
Extracellular Vesicles/metabolism , Host-Pathogen Interactions/immunology , Virulence Factors/metabolism , Animals , Biological Transport , Cell Communication , Exosomes , Extracellular Vesicles/microbiology , Extracellular Vesicles/parasitology , Gene Expression Regulation , Host-Parasite Interactions , Host-Pathogen Interactions/genetics , Humans , Intracellular Space/immunology , Intracellular Space/metabolism , Intracellular Space/microbiology , Intracellular Space/parasitology , Protein Processing, Post-Translational , Signal Transduction
6.
Mol Immunol ; 133: 34-43, 2021 05.
Article in English | MEDLINE | ID: mdl-33621941

ABSTRACT

The anaerobic or microaerophilic protozoan parasites such as the enteric human pathogens Entamoeba histolytica, Giardia intestinalis, Cryptosporidium parvum, Blastocystis hominis and urogenital tract parasites Trichomonas vaginalis are able to survival in an environment with oxygen deprivation. Despite living in hostile environments these pathogens adopted different strategies to survive within the hosts. Among them, the release of extracellular vesicles (EVs) has become an active endeavor in the study of pathogenesis for these parasites. EVs are heterogenous, membrane-limited structures that have played important roles in cellular communication, transferring information through cargo and modulating the immune system of the host. In this review, we described several aspects of the recently characterized EVs of the anaerobic protozoa, including their role in adhesion, modulation of the immune response and omics analysis to understand the potential of these EVs in the pathogenesis of these diseases caused by anaerobic parasites.


Subject(s)
Exosomes/parasitology , Extracellular Vesicles/parasitology , Host-Parasite Interactions/physiology , Protozoan Infections/pathology , Anaerobiosis/physiology , Blastocystis hominis/growth & development , Cell Adhesion/physiology , Cryptosporidium parvum/growth & development , Entamoeba histolytica/growth & development , Extracellular Vesicles/immunology , Giardia lamblia/growth & development , Humans , Protozoan Infections/parasitology , Trichomonas vaginalis/growth & development
7.
Mol Immunol ; 132: 172-183, 2021 04.
Article in English | MEDLINE | ID: mdl-33601226

ABSTRACT

The trypanosomatid pathogens Leishmania spp., Trypanosoma cruzi, and Trypanosoma brucei, currently grouped as TriTryps, have evolved through the time to overcome the upfront innate immune response and establish the infection in humans adapting many aspects of the parasite-cell host interaction. Extracellular vesicles (EVs) emerge as critical structures carrying different key molecules from parasites and target cells that interact continuously during infection. Current information regarding the structure and composition of these vesicles provide new insights into the primary role of TriTryps-EVs reviewed in this work. Expanding knowledge about these critical vesicular structures will promote advances in basic sciences and in translational applications controlling pathogenesis in the neglected tropical diseases caused by TriTryps.


Subject(s)
Extracellular Vesicles/immunology , Leishmania major/immunology , Protozoan Infections/immunology , Trypanosoma brucei brucei/immunology , Trypanosoma cruzi/immunology , Animals , Extracellular Vesicles/parasitology , Host-Parasite Interactions/immunology , Humans , Immunity, Innate/immunology , Protozoan Infections/parasitology
8.
Parasitol Res ; 119(12): 4297-4302, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33089360

ABSTRACT

Malaria is caused by unicellular parasites of the genus Plasmodium, which reside in erythrocytes during the clinically relevant stage of infection. To separate parasite from host cell material, haemolytic agents such as saponin are widely used. Previous electron microscopy studies on saponin-treated parasites reported both, parasites enclosed by the erythrocyte membrane and liberated from the host cell. These ambiguous reports prompted us to investigate haemolysis by live-cell time-lapse microscopy. Using either saponin or streptolysin O to lyse Plasmodium falciparum-infected erythrocytes, we found that ring-stage parasites efficiently exit the erythrocyte upon haemolysis. For late-stage parasites, we found that only approximately half were freed, supporting the previous electron microscopy studies. Immunofluorescence imaging indicated that freed parasites were surrounded by the parasitophorous vacuolar membrane. These results may be of interest for future work using haemolytic agents to enrich for parasite material.


Subject(s)
Erythrocytes/parasitology , Hemolysis/drug effects , Plasmodium falciparum/physiology , Saponins/pharmacology , Streptolysins/pharmacology , Bacterial Proteins/pharmacology , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/parasitology , Erythrocytes/drug effects , Extracellular Vesicles/parasitology , Humans , Life Cycle Stages/physiology , Microscopy , Plasmodium falciparum/growth & development
9.
Mol Biochem Parasitol ; 238: 111296, 2020 07.
Article in English | MEDLINE | ID: mdl-32603736

ABSTRACT

Eggs, schistosomula and adult Schistosoma worms are known to release extracellular vesicles (EV) during in vitro incubations and these EVs are postulated to affect the host responses. So far only those EVs released during in vitro incubations of schistosomes have been studied and it is unknown whether in blood of infected hosts the schistosomal EVs can be detected amidst all the circulating EVs of the host itself. In this study we analyzed the protein as well as the phospholipid composition of EVs circulating in blood plasma of S. mansoni infected hamsters and compared those with the EVs circulating in blood of non-infected hamsters. Although neither proteins nor lipids specific for schistosomes could be detected in the circulating EVs of the infected hamsters, the infection with schistosomes had a marked effect on the circulating EVs of the host, as the protein as well as the lipid composition of EVs circulating in infected hamsters were different from the EVs of uninfected hamsters. The observed changes in the EV lipid and protein content suggest that more EVs are released by the diseased liver, the affected erythrocytes and activated immune cells.


Subject(s)
Blood Proteins/metabolism , Extracellular Vesicles/chemistry , Host-Parasite Interactions , Mesocricetus/parasitology , Phospholipids/blood , Schistosoma mansoni/metabolism , Schistosomiasis mansoni/parasitology , Animals , Chromatography, Liquid , Extracellular Vesicles/metabolism , Extracellular Vesicles/parasitology , Lipidomics , Mesocricetus/blood , Principal Component Analysis , Proteome/metabolism , Protozoan Proteins/blood , Schistosoma mansoni/pathogenicity , Schistosomiasis mansoni/blood , Tandem Mass Spectrometry
10.
Nat Commun ; 11(1): 2761, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32487994

ABSTRACT

Plasmodium vivax is the most widely distributed human malaria parasite. Previous studies have shown that circulating microparticles during P. vivax acute attacks are indirectly associated with severity. Extracellular vesicles (EVs) are therefore major components of circulating plasma holding insights into pathological processes. Here, we demonstrate that plasma-derived EVs from Plasmodium vivax patients (PvEVs) are preferentially uptaken by human spleen fibroblasts (hSFs) as compared to the uptake of EVs from healthy individuals. Moreover, this uptake induces specific upregulation of ICAM-1 associated with the translocation of NF-kB to the nucleus. After this uptake, P. vivax-infected reticulocytes obtained from patients show specific adhesion properties to hSFs, reversed by inhibiting NF-kB translocation to the nucleus. Together, these data provide physiological EV-based insights into the mechanisms of human malaria pathology and support the existence of P. vivax-adherent parasite subpopulations in the microvasculature of the human spleen.


Subject(s)
Extracellular Vesicles/metabolism , Fibroblasts/metabolism , NF-kappa B/metabolism , Plasma , Plasmodium vivax/physiology , Reticulocytes/metabolism , Spleen/metabolism , Animals , Cell Adhesion , Cell-Derived Microparticles , Disease Models, Animal , Extracellular Vesicles/parasitology , Fibroblasts/pathology , Host-Parasite Interactions/physiology , Humans , Intercellular Adhesion Molecule-1/metabolism , Malaria, Vivax/parasitology , Male , Mice , Mice, Inbred C57BL , Microvessels/parasitology , Proteomics , Reticulocytes/parasitology , Spleen/pathology
11.
Front Immunol ; 11: 22, 2020.
Article in English | MEDLINE | ID: mdl-32082312

ABSTRACT

Malaria infection caused by the Plasmodium species is a complex disease in which a fine balance between host and parasite factors determine the disease severity. While in some individuals, the infection will trigger only a mild and uncomplicated disease, other individuals will develop severe complications which lead to death. Extracellular vesicles (EVs) secreted by infected red blood cells (iRBCs), as well as other host cells, are important regulators of the balance that determines the disease outcome. In addition, EVs constitute a robust mode of cell-to-cell communication by transferring signaling cargoes between parasites, and between parasites and host, without requiring cellular contact. The transfer of membrane and cytosolic proteins, lipids, DNA, and RNA through EVs not only modulate the immune response, it also mediates cellular communication between parasites to synchronize the transmission stage. Here, we review the recent progress in understanding EV roles during malaria.


Subject(s)
Cell Communication/immunology , Extracellular Vesicles/metabolism , Malaria/immunology , Plasmodium/growth & development , Signal Transduction/immunology , Animals , Disease Models, Animal , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/parasitology , Extracellular Vesicles/parasitology , Host-Parasite Interactions/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/parasitology , Life Cycle Stages , Malaria/parasitology , Mice , RNA/metabolism
12.
J Cell Sci ; 132(6)2019 03 18.
Article in English | MEDLINE | ID: mdl-30886004

ABSTRACT

Vector-borne diseases cause over 700,000 deaths annually and represent 17% of all infectious illnesses worldwide. This public health menace highlights the importance of understanding how arthropod vectors, microbes and their mammalian hosts interact. Currently, an emphasis of the scientific enterprise is at the vector-host interface where human pathogens are acquired and transmitted. At this spatial junction, arthropod effector molecules are secreted, enabling microbial pathogenesis and disease. Extracellular vesicles manipulate signaling networks by carrying proteins, lipids, carbohydrates and regulatory nucleic acids. Therefore, they are well positioned to aid in cell-to-cell communication and mediate molecular interactions. This Review briefly discusses exosome and microvesicle biogenesis, their cargo, and the role that nanovesicles play during pathogen spread, host colonization and disease pathogenesis. We then focus on the role of extracellular vesicles in dictating microbial pathogenesis and host immunity during transmission of vector-borne pathogens.


Subject(s)
Arthropod Vectors , Extracellular Vesicles , Vector Borne Diseases , Amebiasis/parasitology , Amebiasis/transmission , Animals , Arthropod Vectors/microbiology , Arthropod Vectors/parasitology , Culicidae/microbiology , Culicidae/parasitology , Disease Vectors , Exosomes/immunology , Exosomes/microbiology , Exosomes/parasitology , Extracellular Vesicles/immunology , Extracellular Vesicles/microbiology , Extracellular Vesicles/parasitology , Filariasis/parasitology , Filariasis/transmission , Hemiptera/microbiology , Hemiptera/parasitology , Host-Parasite Interactions/immunology , Host-Parasite Interactions/physiology , Humans , Immunomodulation , Leishmaniasis/parasitology , Leishmaniasis/transmission , Malaria/parasitology , Malaria/transmission , Psychodidae/microbiology , Psychodidae/parasitology , Trypanosomiasis/parasitology , Trypanosomiasis/transmission , Vector Borne Diseases/microbiology , Vector Borne Diseases/parasitology , Vector Borne Diseases/transmission , Virus Diseases/microbiology , Virus Diseases/transmission
14.
Biochim Biophys Acta Gen Subj ; 1863(2): 325-331, 2019 02.
Article in English | MEDLINE | ID: mdl-30339917

ABSTRACT

BACKGROUND: Malaria is a serious parasitic infection affecting millions of people worldwide each year. Cerebral malaria is the most severe complication of Plasmodium infections, predominantly affecting children. Extracellular vesicles are essential mediators of intercellular communication and include apoptotic bodies, microvesicles and exosomes. Microvesicle numbers increase during disease pathogenesis and inhibition of their release can prevent brain pathology and mortality. SCOPE OF REVIEW: We explore the current knowledge on microvesicles and exosomes in cerebral malaria pathogenesis. MAJOR CONCLUSIONS: Microvesicles and exosomes are implicated in cerebral malaria pathogenesis, in the modulation of host immunity to Plasmodium, and in cell-cell communication. Blocking their production is protective in models of cerebral malaria, both in vivo and in vitro. GENERAL SIGNIFICANCE: While anti-malarial treatments exist to combat Plasmodium infections, increasing drug resistance presents a major challenge. In order to improve diagnosis and treatment outcomes, further research is required to better appreciate extracellular vesicle involvement in cerebral malaria.


Subject(s)
Extracellular Vesicles/pathology , Extracellular Vesicles/parasitology , Malaria, Cerebral/pathology , Malaria, Cerebral/parasitology , Plasmodium/pathogenicity , Antimalarials/pharmacology , Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Humans , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Plasmodium/drug effects
15.
Parasitology ; 145(12): 1521-1530, 2018 10.
Article in English | MEDLINE | ID: mdl-29986788

ABSTRACT

Extracellular vesicles (EVs) are released by a wide number of cells including blood cells, immune system cells, tumour cells, adult and embryonic stem cells. EVs are a heterogeneous group of vesicles (~30-1000 nm) including microvesicles and exosomes. The physiological release of EVs represents a normal state of the cell, raising a metabolic equilibrium between catabolic and anabolic processes. Moreover, when the cells are submitted to stress with different inducers or in pathological situations (malignancies, chronic diseases, infectious diseases.), they respond with an intense and dynamic release of EVs. The EVs released from stimulated cells vs those that are released constitutively may themselves differ, both physically and in their cargo. EVs contain protein, lipids, nucleic acids and biomolecules that can alter cell phenotypes or modulate neighbouring cells. In this review, we have summarized findings involving EVs in certain protozoan diseases. We have commented on strategies to study the communicative roles of EVs during host-pathogen interaction and hypothesized on the use of EVs for diagnostic, preventative and therapeutic purposes in infectious diseases. This kind of communication could modulate the innate immune system and reformulate concepts in parasitism. Moreover, the information provided within EVs could produce alternatives in translational medicine.


Subject(s)
Extracellular Vesicles/parasitology , Host-Pathogen Interactions , Leishmania/physiology , Plasmodium/physiology , Trypanosoma/physiology , Exosomes/parasitology , Humans , Phenotype
16.
Parasite Immunol ; 40(9): e12571, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29974519

ABSTRACT

This study established a protocol to purify Toxoplasma gondii tachyzoite microvesicles and exosomes, called as extracellular vesicles (EVs). In addition, the investigations were conducted to determine the kinetic of EV release by tachyzoites and whether EV proteins are able to modulate the host immune response. The particle size and concentration released by tachyzoites in culture medium at different incubation-period were characterized by nanoparticle tracking analysis. Tachyzoites (1 × 106 ) released around 4.37 ± 0.81 × 108 EVs/mL/h, with size varying between 138.2 and 171.9 nm. EVs released into the medium were purified by gel-exclusion chromatography and screened by ELISA, using a pool of human positive sera for toxoplasmosis. EV-fractions contained high concentration of proteins, and EVs were analyzed by scanning and transmission electron microscopies. Tachyzoites released EVs into the culture medium throughout all membrane surface, and these vesicles contain small RNAs/miRNA. Pooled sera from chronically infected human or mice (infected with 2 different T. gondii strains) recognized distinct EV electrophoretic patterns in immunoblotting. T. gondii EVs significantly induced IL-10, TNF-α and iNOS in murine macrophages. In conclusion, this study shows that T. gondii secrete/excrete EVs (microvesicles and exosomes) contain miRNA and they were immunologically recognized by host immune response.


Subject(s)
Extracellular Vesicles/immunology , Toxoplasma/immunology , Toxoplasmosis/parasitology , Animals , Enzyme-Linked Immunosorbent Assay , Exosomes/immunology , Exosomes/parasitology , Extracellular Vesicles/parasitology , Humans , Immunoblotting , Interleukin-10/genetics , Interleukin-10/immunology , Macrophages/immunology , Macrophages/parasitology , Mice , Toxoplasma/genetics , Toxoplasmosis/genetics , Toxoplasmosis/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
17.
PLoS Negl Trop Dis ; 12(5): e0006475, 2018 05.
Article in English | MEDLINE | ID: mdl-29727453

ABSTRACT

TcTASV-C is a protein family of about 15 members that is expressed only in the trypomastigote stage of Trypanosoma cruzi. We have previously shown that TcTASV-C is located at the parasite surface and secreted to the medium. Here we report that the expression of different TcTASV-C genes occurs simultaneously at the trypomastigote stage and while some secreted and parasite-associated products are found in both fractions, others are different. Secreted TcTASV-C are mainly shedded through trypomastigote extracellular vesicles, of which they are an abundant constituent, despite its scarce expression on culture-derived trypomastigotes. In contrast, TcTASV-C is highly expressed in bloodstream trypomastigotes; its upregulation in bloodstream parasites was observed in different T. cruzi strains and was specific for TcTASV-C, suggesting that some host-molecules trigger TcTASV-C expression. TcTASV-C is also strongly secreted by bloodstream parasites. A DNA prime-protein boost immunization scheme with TcTASV-C was only partially effective to control the infection in mice challenged with a highly virulent T. cruzi strain. Vaccination triggered a strong humoral response that delayed the appearance of bloodstream trypomastigotes at the early phase of the infection. Linear epitopes recognized by vaccinated mice were mapped within the TcTASV-C family motif, suggesting that blockade of secreted TcTASV-C impacts on the settlement of infection. Furthermore, although experimental and naturally T. cruzi-infected hosts did not react with antigens from extracellular vesicles, vaccinated and challenged mice recognized not only TcTASV-C but also other vesicle-antigens. We hypothesize that TcTASV-C is involved in the establishment of the initial T. cruzi infection in the mammalian host. Altogether, these results point towards TcTASV-C as a novel secreted virulence factor of T. cruzi trypomastigotes.


Subject(s)
Blood/parasitology , Chagas Disease/parasitology , Extracellular Vesicles/parasitology , Protozoan Proteins/metabolism , Trypanosoma cruzi/growth & development , Trypanosoma cruzi/metabolism , Virulence Factors/metabolism , Animals , Chagas Disease/blood , Chagas Disease/metabolism , Extracellular Vesicles/metabolism , Humans , Mice , Mice, Inbred C3H , Multigene Family , Protein Transport , Protozoan Proteins/genetics , Trypanosoma cruzi/genetics , Virulence Factors/genetics
18.
Sci Rep ; 8(1): 884, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343745

ABSTRACT

The parasite Plasmodium falciparum causes the most severe form of malaria. Cell communication between parasites is an important mechanism to control population density and differentiation. The infected red blood cells (iRBCs) release small extracellular vesicles (EVs) that transfer cargoes between cells. The EVs synchronize the differentiation of the asexual parasites into gametocytes to initiate the transmission to the mosquito. Beside their role in parasite communication, EVs regulate vascular function. So far, the exact cargoes responsible for cellular communication remain unknown. We isolated EVs from cultured iRBCs to determine their small RNA content. We identified several types of human and plasmodial regulatory RNAs. While the miRNAs and tRNA-derived fragments were the most abundant human RNAs, we also found Y-RNAs, vault RNAs, snoRNAs and piRNAs. Interestingly, we found about 120 plasmodial RNAs, including mRNAs coding for exported proteins and proteins involved in drug resistance, as well as non-coding RNAs, such as rRNAs, small nuclear (snRNAs) and tRNAs. These data show, that iRBC-EVs carry small regulatory RNAs. A role in cellular communication is possible since the RNAs were transferred to endothelial cells. Furthermore, the presence of Plasmodium RNAs, in EVs suggests that they may be used as biomarker to track and detect disease.


Subject(s)
Erythrocytes/parasitology , Extracellular Vesicles/genetics , Malaria/genetics , RNA/genetics , Cell Communication/genetics , Cell Differentiation/genetics , Cells, Cultured , Endothelial Cells/parasitology , Erythrocyte Count/methods , Extracellular Vesicles/parasitology , Humans , Malaria/parasitology , Plasmodium falciparum/pathogenicity
19.
J Infect Dis ; 215(3): 378-386, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-28362903

ABSTRACT

BACKGROUND: Schistosomiasis traditionally has been diagnosed by detecting eggs in stool or urine. However, the sensitivity of these examinations is limited, especially in travelers with a low worm burden. Serologic tests have a greater sensitivity, but their results remain positive regardless of treatment and thus cannot be used for follow-up of patients. We hypothesized that detection of worm microRNAs (miRNAs) in serum can overcome the drawbacks of the existing diagnostic methods. METHODS AND RESULTS: Twenty-six returning travelers with schistosomiasis (based on positive results of serologic tests or detection of ova) and 17 healthy controls were included in the study. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) amplification of miRNA extracted directly from 500 µL of serum had limited sensitivity and specificity. However, qRT-PCR analysis of RNA extracted from 200 µL of serum extracellular vesicles detected 4 schistosomal miRNAs; the sensitivity and specificity of the 2 highest expressed miRNAs (bantam and miR-2c-3p) were 86% and 84%, respectively. In 7 patients with posttreatment serum available for analysis, we observed outcomes ranging from a reduction in the schistosomal miRNA level to full recovery from disease. CONCLUSIONS: qRT-PCR of pathogen miRNAs isolated from extracellular vesicles in sera from infected individuals may provide a new tool for diagnosing schistosomiasis in patients with a low parasite burden. This assay could also be used for evaluating the outcome of therapy, as well as disease-control programs.


Subject(s)
Extracellular Vesicles/parasitology , MicroRNAs/blood , RNA, Helminth/blood , Schistosoma mansoni/genetics , Schistosomiasis/diagnosis , Adult , Animals , Female , Follow-Up Studies , Humans , Male , MicroRNAs/isolation & purification , Middle Aged , Real-Time Polymerase Chain Reaction , Schistosoma mansoni/isolation & purification , Schistosomiasis/blood , Schistosomiasis/parasitology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...