Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Front Cell Dev Biol ; 10: 812244, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35652104

RESUMEN

Malaria affects the poorer regions of the world and is of tremendous health and economic burden for developing countries. Extracellular vesicles (EVs) are small vesicles released by almost any cells in the human body, including malaria infected red blood cells. Recent evidence shows that EVs might contribute to the pathogenesis of malaria. In addition, EVs hold considerable value in biomarker discovery. However, there are still significant gaps in our understanding of EV biology. So far most of our knowledge about EVs in malaria comes from in vitro work. More field studies are required to gain insight into their contribution to the disease and pathogenesis under physiological conditions. However, to perform research on EVs in low-income regions might be challenging due to the lack of appropriate equipment to isolate EVs. Therefore, there is a need to develop and validate EV extraction protocols applicable to poorly equipped laboratories. We established and validated two protocols for EV isolation from cell culture supernatants, rodent and human plasma. We compared polyethylene glycol (PEG) and salting out (SA) with sodium acetate for precipitation of EVs. We then characterized the EVs by Transmission Electron Microscopy (TEM), Western Blot, Size-exclusion chromatography (SEC), bead-based flow cytometry and protein quantification. Both protocols resulted in efficient purification of EVs without the need of expensive material or ultracentrifugation. Furthermore, the procedure is easily scalable to work with large and small sample volumes. Here, we propose that both of our approaches can be used in resource limited countries, therefore further helping to close the gap in knowledge of EVs during malaria.

2.
Eur J Immunol ; 52(8): 1273-1284, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35503749

RESUMEN

Endemic Burkitt lymphoma (eBL) is characterized by an oncogenic IGH/c-MYC translocation and Epstein-Barr virus (EBV) positivity, and is epidemiologically linked to Plasmodium falciparum malaria. Both EBV and malaria are thought to contribute to eBL by inducing the expression of activation-induced cytidine deaminase (AID), an enzyme involved in the IGH/c-MYC translocation. AID/apolipoprotein B mRNA editing catalytic polypeptide-like (AID/APOBEC) family enzymes have recently emerged as potent mutagenic sources in a variety of cancers, but apart from AID, their involvement in eBL and their regulation by EBV and P. falciparum is unknown. Here, we show that upon inoculation with EBV, human B cells strongly upregulate the expression of enzymatically active APOBEC3B and APOBEC3G. In addition, we found significantly increased levels of APOBEC3A in B cells of malaria patients, which correlated with parasite load. Interestingly, despite the fact that APOBEC3A, APOBEC3B, and APOBEC3G caused c-MYC mutations when overexpressed in HEK293T cells, a mutational enrichment in eBL tumors was only detected in AID motifs. This suggests that even though the EBV- and P. falciparum-directed immune response triggers the expression and activity of several AID/APOBEC members, only the upregulation of AID has oncogenic consequences, while the induction of the APOBEC3 subfamily may primarily have immunoprotective functions.


Asunto(s)
Desaminasas APOBEC , Linfoma de Burkitt , Citidina Desaminasa , Infecciones por Virus de Epstein-Barr , Malaria Falciparum , Desaminasas APOBEC/genética , Desaminasa APOBEC-3G , Linfoma de Burkitt/enzimología , Linfoma de Burkitt/genética , Citidina Desaminasa/genética , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Virus de Epstein-Barr/genética , Células HEK293 , Herpesvirus Humano 4 , Humanos , Malaria Falciparum/enzimología , Malaria Falciparum/genética , Antígenos de Histocompatibilidad Menor , Mutágenos
3.
Front Immunol ; 12: 750512, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34707614

RESUMEN

Cell-mediated cytotoxicity is an essential immune defense mechanism to fight against viral, bacterial or parasitic infections. Upon recognition of an infected target cell, killer lymphocytes form an immunological synapse to release the content of their cytotoxic granules. Cytotoxic granules of humans contain two membrane-disrupting proteins, perforin and granulysin, as well as a homologous family of five death-inducing serine proteases, the granzymes. The granzymes, after delivery into infected host cells by the membrane disrupting proteins, may contribute to the clearance of microbial pathogens through different mechanisms. The granzymes can induce host cell apoptosis, which deprives intracellular pathogens of their protective niche, therefore limiting their replication. However, many obligate intracellular pathogens have evolved mechanisms to inhibit programed cells death. To overcome these limitations, the granzymes can exert non-cytolytic antimicrobial activities by directly degrading microbial substrates or hijacked host proteins crucial for the replication or survival of the pathogens. The granzymes may also attack factors that mediate microbial virulence, therefore directly affecting their pathogenicity. Many mechanisms applied by the granzymes to eliminate infected cells and microbial pathogens rely on the induction of reactive oxygen species. These reactive oxygen species may be directly cytotoxic or enhance death programs triggered by the granzymes. Here, in the light of the latest advances, we review the antimicrobial activities of the granzymes in regards to their cytolytic and non-cytolytic activities to inhibit pathogen replication and invasion. We also discuss how reactive oxygen species contribute to the various antimicrobial mechanisms exerted by the granzymes.


Asunto(s)
Granzimas/inmunología , Animales , Muerte Celular , Humanos , Infecciones/inmunología , Especies Reactivas de Oxígeno/inmunología
4.
Front Immunol ; 12: 643746, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093532

RESUMEN

Malaria remains one of the most serious health problems in developing countries. The causative agent of malaria, Plasmodium spp., have a complex life cycle involving multiple developmental stages as well as different morphological, biochemical and metabolic requirements. We recently found that γδ T cells control parasite growth using pore-forming proteins to deliver their cytotoxic proteases, the granzymes, into blood residing parasites. Here, we follow up on the molecular mechanisms of parasite growth inhibition by human pore-forming proteins. We confirm that Plasmodium falciparum infection efficiently depletes the red blood cells of cholesterol, which renders the parasite surrounding membranes susceptible to lysis by prokaryotic membrane disrupting proteins, such as lymphocytic granulysin or the human cathelicidin LL-37. Interestingly, not the cholesterol depletion but rather the simultaneous exposure of phosphatidylserine, a negatively charged phospholipid, triggers resistance of late stage parasitized red blood cells towards the eukaryotic pore forming protein perforin. Overall, by revealing the molecular events we establish here a pathogen-host interaction that involves host cell membrane remodeling that defines the susceptibility towards cytolytic molecules.


Asunto(s)
Membrana Eritrocítica/inmunología , Hemólisis/inmunología , Malaria Falciparum/inmunología , Perforina/inmunología , Plasmodium falciparum/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Antígenos de Diferenciación de Linfocitos T , Péptidos Catiónicos Antimicrobianos/inmunología , Susceptibilidad a Enfermedades , Membrana Eritrocítica/parasitología , Humanos , Catelicidinas
5.
Sci Rep ; 11(1): 778, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436661

RESUMEN

The human leukemia cell line (HL-60) is an alternative to primary neutrophils in research studies. However, because HL-60 cells proliferate in an incompletely differentiated state, they must undergo differentiation before they acquire the functional properties of neutrophils. Here we provide evidence of swarming and chemotaxis in differentiated HL-60 neutrophil-like cells (dHL-60) using precise microfluidic assays. We found that dimethyl sulfoxide differentiated HL-60 cells (DdHL-60) have a larger size, increased length, and lower ability to squeeze through narrow channels compared to primary neutrophils. They migrate through tapered microfluidic channels slower than primary neutrophils, but faster than HL-60s differentiated by other protocols, e.g., using all-trans retinoic acid. We found that dHL-60 can swarm toward zymosan particle clusters, though they display disorganized migratory patterns and produce swarms of smaller size compared to primary neutrophils.


Asunto(s)
Factores Quimiotácticos/farmacología , Quimiotaxis/fisiología , Dimetilsulfóxido/farmacología , Neutrófilos/fisiología , Tretinoina/farmacología , Antineoplásicos/farmacología , Diferenciación Celular/fisiología , Crioprotectores/farmacología , Células HL-60 , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/fisiología , Neutrófilos/citología , Neutrófilos/efectos de los fármacos
6.
Blood ; 136(12): 1375-1376, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32941634
7.
iScience ; 23(3): 100932, 2020 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-32151975

RESUMEN

Pathogenic bacteria secrete virulence factors that interact with the human host to establish infections. The human immune system evolved multiple mechanisms to fight bacterial invaders, including immune proteases that were demonstrated to contribute crucially to antibacterial defense. Here we show that granzyme B degrades multiple secreted virulence mediators from Listeria monocytogenes, Salmonella typhimurium, and Mycobacteria tuberculosis. Pathogenic bacteria, when infected in the presence of granzyme B or granzyme-secreting killer cells, fail to grow in human macrophages and epithelial cells owing to their crippled virulence. A granzyme B-uncleavable mutant form of the major Listeria virulence factor, listeriolysin O, rescued the virulence defect in response to granzyme treatment. Hence, we link the degradation of a single factor with the observed decrease in virulent bacteria growth. Overall, we reveal here an innate immune barrier function of granzyme B by disrupting bacterial virulence to facilitate bacteria clearance by bystander immune and non-immune cells.

8.
Front Immunol ; 11: 22, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32082312

RESUMEN

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.


Asunto(s)
Comunicación Celular/inmunología , Vesículas Extracelulares/metabolismo , Malaria/inmunología , Plasmodium/crecimiento & desarrollo , Transducción de Señal/inmunología , Animales , Modelos Animales de Enfermedad , Eritrocitos/inmunología , Eritrocitos/metabolismo , Eritrocitos/parasitología , Vesículas Extracelulares/parasitología , Interacciones Huésped-Parásitos/inmunología , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/parasitología , Estadios del Ciclo de Vida , Malaria/parasitología , Ratones , ARN/metabolismo
9.
J Immunol ; 204(7): 1798-1809, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32066596

RESUMEN

Plasmodium spp., the causative agent of malaria, have a complex life cycle. The exponential growth of the parasites during the blood stage is responsible for almost all malaria-associated morbidity and mortality. Therefore, tight immune control of the intraerythrocytic replication of the parasite is essential to prevent clinical malaria. Despite evidence that the particular lymphocyte subset of γδ T cells contributes to protective immunity during the blood stage in naive hosts, their precise inhibitory mechanisms remain unclear. Using human PBMCs, we confirmed in this study that γδ T cells specifically and massively expanded upon activation with Plasmodium falciparum culture supernatant. We also demonstrate that these activated cells gain cytolytic potential by upregulating cytotoxic effector proteins and IFN-γ. The killer cells bound to infected RBCs and killed intracellular P. falciparum via the transfer of the granzymes, which was mediated by granulysin in a stage-specific manner. Several vital plasmodial proteins were efficiently destroyed by granzyme B, suggesting proteolytic degradation of these proteins as essential in the lymphocyte-mediated death pathway. Overall, these data establish a granzyme- and granulysin-mediated innate immune mechanism exerted by γδ T cells to kill late-stage blood-residing P. falciparum.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Granzimas/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Antígenos de Protozoos/inmunología , Células Cultivadas , Eritrocitos/inmunología , Humanos , Inmunidad Innata/inmunología , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/inmunología , Estadios del Ciclo de Vida/inmunología , Activación de Linfocitos/inmunología , Subgrupos de Linfocitos T/inmunología , Regulación hacia Arriba/inmunología
10.
Pathogens ; 9(1)2019 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-31878288

RESUMEN

Microglia are the chief immune cells of the brain and have been reported to be activated in severe malaria. Their activation may drive towards neuroinflammation in cerebral malaria. Malaria-infected red blood cell derived-extracellular vesicles (MiREVs) are produced during the blood stage of malaria infection. They mediate intercellular communication and immune regulation, among other functions. During cerebral malaria, the breakdown of the blood-brain barrier can promote the migration of substances such as MiREVs from the periphery into the brain, targeting cells such as microglia. Microglia and extracellular vesicle interactions in different pathological conditions have been reported to induce neuroinflammation. Unlike in astrocytes, microglia-extracellular vesicle interaction has not yet been described in malaria infection. Therefore, in this study, we aimed to investigate the uptake of MiREVs by human microglia cells and their cytokine response. Human blood monocyte-derived microglia (MoMi) were generated from buffy coats of anonymous healthy donors using Ficoll-Paque density gradient centrifugation. The MiREVs were isolated from the Plasmodium falciparum cultures. They were purified by ultracentrifugation and labeled with PKH67 green fluorescent dye. The internalization of MiREVs by MoMi was observed after 4 h of co-incubation on coverslips placed in a 24-well plate at 37 °C using confocal microscopy. Cytokine-gene expression was investigated using rt-qPCR, following the stimulation of the MoMi cells with supernatants from the parasite cultures at 2, 4, and 24 h, respectively. MiREVs were internalized by the microglia and accumulated in the perinuclear region. MiREVs-treated cells increased gene expression of the inflammatory cytokine TNFα and reduced gene expression of the immune suppressive IL-10. Overall, the results indicate that MiREVs may act on microglia, which would contribute to enhanced inflammation in cerebral malaria.

11.
Nat Commun ; 10(1): 1369, 2019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30911004

RESUMEN

Methods to diagnose malaria are of paramount interest to eradicate the disease. Current methods have severe limitations, as they are either costly or not sensitive enough to detect low levels of parasitemia. Here we report an ultrasensitive, yet low-resource chemical assay for the detection and quantification of hemozoin, a biomarker of all Plasmodium species. Solubilized hemozoin catalyzes the atom transfer radical polymerization of N-isopropylacrylamide above the lower critical solution temperature of poly(N-isopropylacrylamide). The solution becomes turbid, which can be observed by naked eye and quantified by UV-visible spectroscopy. The rate of turbidity increase is proportional to the concentration of hemozoin, with a detection limit of 0.85 ng mL-1. Malaria parasites in human blood can be detected down to 10 infected red blood cells µL-1. The assay could potentially be applied as a point-of-care test. The signal-amplification of an analyte by biocatalytic precipitation polymerization represents a powerful approach in biosensing.


Asunto(s)
Acrilamidas/química , Resinas Acrílicas/química , Bioensayo , Técnicas Biosensibles , Hemoproteínas/química , Malaria Falciparum/diagnóstico , Plasmodium falciparum/química , Biocatálisis , Eritrocitos/parasitología , Hemoproteínas/aislamiento & purificación , Humanos , Límite de Detección , Malaria Falciparum/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Polimerizacion , Espectrofotometría/métodos
12.
J Vis Exp ; (132)2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29553510

RESUMEN

Malaria is a life-threatening disease caused by Plasmodium parasites, with P. falciparum being the most prevalent on the African continent and responsible for most malaria-related deaths globally. Several factors including parasite sequestration in tissues, vascular dysfunction, and inflammatory responses influence the evolution of the disease in malaria-infected people. P. falciparum-infected red blood cells (iRBCs) release small extracellular vesicles (EVs) containing different kinds of cargo molecules that mediate pathogenesis and cellular communication between parasites and host. EVs are efficiently taken up by cells in which they modulate their function. Here we discuss strategies to address the role of EVs in parasite-host interactions. First, we describe a straightforward method for labeling and tracking EV internalization by endothelial cells, using a green cell linker dye. Second, we report a simple way to measure permeability across an endothelial cell monolayer by using a fluorescently labeled dextran. Finally, we show how to investigate the role of small non-coding RNA molecules in endothelial cell function.


Asunto(s)
Células Endoteliales/patología , Eritrocitos/patología , Eritrocitos/parasitología , Vesículas Extracelulares/patología , Malaria Falciparum/sangre , Animales , Células Endoteliales/metabolismo , Eritrocitos/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Microscopía Confocal
13.
Sci Rep ; 8(1): 884, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343745

RESUMEN

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.


Asunto(s)
Eritrocitos/parasitología , Vesículas Extracelulares/genética , Malaria/genética , ARN/genética , Comunicación Celular/genética , Diferenciación Celular/genética , Células Cultivadas , Células Endoteliales/parasitología , Recuento de Eritrocitos/métodos , Vesículas Extracelulares/parasitología , Humanos , Malaria/parasitología , Plasmodium falciparum/patogenicidad
14.
Nanomedicine ; 14(2): 601-607, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29155361

RESUMEN

Bone infections are difficult to treat and can lead to severe tissue destruction. Acute bone infections are usually caused by Staphylococcus aureus. Osteoclasts, which belong to the monocyte/macrophage lineage, are the key cells in bone infections. They are not well equipped for killing bacteria and may serve as a reservoir for bacterial pathogens. Silver has been known for centuries for its bactericidal activity. Here, we investigated the bactericidal effects of nano-silver particles in bacteria infected human osteoclasts. We found that nano-silver in per se non-toxic concentration enhanced the bactericidal activity in osteoclasts against intracellular Methicillin-resistant, virulent Staphylococcus aureus. The reduced bacterial survival in nano-silver pretreated cells correlated with increased reactive oxygen responses towards the invading pathogens. Overall, these results indicate that nano-silver compounds should be considered as an effective treatment and prevention option for bacterial bone and orthopedic implant infections.


Asunto(s)
Antibacterianos/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Osteoclastos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Plata/química , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/química , Células Cultivadas , Humanos , Nanopartículas del Metal/química , Osteoclastos/patología , Fagocitosis , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/aislamiento & purificación
15.
Methods Mol Biol ; 1660: 377-388, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28828673

RESUMEN

Growing attention is drawn toward the role of extracellular vesicles (EVs) in infectious diseases. EVs, which are small vesicles released by cells, are involved in cellular communication, immune regulation, and pathogenesis. EVs act as messenger carrying functional cargoes, including RNA, DNA, lipids and proteins from a donor cell to regulate the function of a recipient cell. In malaria, EVs play a key role in regulating the progression from the blood to the transmission stage by promoting the switch between asexual and sexual stages that are taken up by mosquitoes. In addition to their role in parasite communication, EVs modulate the immune system and regulate endothelial cell function.In this chapter, we describe protocols to isolate, purify and characterize EVs derived from Plasmodium falciparum infected red blood cell culture.


Asunto(s)
Eritrocitos/metabolismo , Eritrocitos/parasitología , Malaria/metabolismo , Malaria/parasitología , Comunicación Celular , Cromatografía en Gel , Cromatografía Liquida , Vesículas Extracelulares/metabolismo , Humanos , Plasmodium falciparum , Ultracentrifugación
16.
Methods Mol Biol ; 1535: 275-284, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27914086

RESUMEN

Bacterial pathogens represent a constant threat to human health that was exacerbated in recent years by a dramatic increase of strains resistant to last resort antibiotics. The immune system of higher vertebrates generally evolved several efficient innate and adaptive mechanisms to fight ubiquitous bacterial pathogens. Among those mechanisms, immune proteases were recognized to contribute essentially to antibacterial immune defense. The effector serine proteases of the adaptive immune system, the granzymes, exert potent antimicrobial activity when they are delivered into the bacterial cytosol by prokaryotic membrane disrupting proteins, such as granulysin.In this chapter, we are detailing experimental protocols to study the synergistic cytotoxic effects of human granzymes and granulysin on extracellular as well as on intracellular bacterial pathogens in vitro. In addition, we provide a simple and fast-forward method to biochemically purify native cytotoxic effector molecules necessary to perform this kind of investigations.


Asunto(s)
Antibacterianos/farmacología , Antígenos de Diferenciación de Linfocitos T/farmacología , Bacterias/efectos de los fármacos , Bacterias/inmunología , Citotoxicidad Inmunológica , Granzimas/farmacología , Perforina/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Espacio Extracelular/inmunología , Espacio Extracelular/microbiología , Humanos , Espacio Intracelular/inmunología , Espacio Intracelular/microbiología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
17.
Nat Commun ; 7: 12727, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27721445

RESUMEN

Malaria remains one of the greatest public health challenges worldwide, particularly in sub-Saharan Africa. The clinical outcome of individuals infected with Plasmodium falciparum parasites depends on many factors including host systemic inflammatory responses, parasite sequestration in tissues and vascular dysfunction. Production of pro-inflammatory cytokines and chemokines promotes endothelial activation as well as recruitment and infiltration of inflammatory cells, which in turn triggers further endothelial cell activation and parasite sequestration. Inflammatory responses are triggered in part by bioactive parasite products such as hemozoin and infected red blood cell-derived extracellular vesicles (iRBC-derived EVs). Here we demonstrate that such EVs contain functional miRNA-Argonaute 2 complexes that are derived from the host RBC. Moreover, we show that EVs are efficiently internalized by endothelial cells, where the miRNA-Argonaute 2 complexes modulate target gene expression and barrier properties. Altogether, these findings provide a mechanistic link between EVs and vascular dysfunction during malaria infection.


Asunto(s)
Proteínas Argonautas/metabolismo , Vasos Sanguíneos/metabolismo , Eritrocitos/parasitología , Vesículas Extracelulares/metabolismo , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , MicroARNs/metabolismo , Encéfalo/irrigación sanguínea , Línea Celular , Endocitosis , Células Endoteliales/metabolismo , Eritrocitos/ultraestructura , Vesículas Extracelulares/ultraestructura , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , MicroARNs/genética , Microvasos/citología , Complejo Silenciador Inducido por ARN/metabolismo
18.
Sci Rep ; 6: 21216, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26880111

RESUMEN

Infections with Plasmodium falciparum, the most pathogenic of the Plasmodium species affecting man, have been reduced in part due to artemisinin-based combination therapies. However, artemisinin resistant parasites have recently emerged in South-East Asia. Novel intervention strategies are therefore urgently needed to maintain the current momentum for control and elimination of this disease. In the present study we characterize the phenotypic and genetic properties of the multi drug resistant (MDR) P. falciparum Thai C2A parasite strain in the non-human Aotus primate model, and across multiple passages. Aotus infections with C2A failed to clear upon oral artesunate and mefloquine treatment alone or in combination, and ex vivo drug assays demonstrated reduction in drug susceptibility profiles in later Aotus passages. Further analysis revealed mutations in the pfcrt and pfdhfr loci and increased parasite multiplication rate (PMR) across passages, despite elevated pfmdr1 copy number. Altogether our experiments suggest alterations in parasite population structure and increased fitness during Aotus adaptation. We also present data of early treatment failures with an oral artemisinin combination therapy in a pre-artemisinin resistant P. falciparum Thai isolate in this animal model.


Asunto(s)
Adaptación Biológica , Antimaláricos/farmacología , Resistencia a Medicamentos , Interacciones Huésped-Patógeno , Malaria Falciparum/parasitología , Plasmodium falciparum/efectos de los fármacos , Animales , Antimaláricos/administración & dosificación , Aotidae , Artemisininas/administración & dosificación , Artemisininas/farmacología , Artesunato , Modelos Animales de Enfermedad , Malaria Falciparum/tratamiento farmacológico , Pruebas de Sensibilidad Parasitaria , Fenotipo , Plasmodium falciparum/genética , Primates , Sitios de Carácter Cuantitativo , Insuficiencia del Tratamiento
19.
PLoS One ; 11(1): e0144678, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26745887

RESUMEN

The identification of extracellular vesicles (EVs) as intercellular conveyors of biological information has recently emerged as a novel paradigm in signaling, leading to the exploitation of EVs and their contents as biomarkers of various diseases. However, whether there are diurnal variations in the size, number, and tissue of origin of blood EVs is currently not known, and could have significant implications when using EVs as biomarkers for disease progression. Currently available technologies for the measurement of EV size and number are either time consuming, require specialized equipment, or lack sufficient accuracy across a range of EV sizes. Flow cytometry represents an attractive alternative to these methods; however, traditional flow cytometers are only capable of measuring particles down to 500 nm, which is significantly larger than the average and median sizes of plasma EVs. Utilizing a Beckman Coulter MoFlo XDP flow cytometer with NanoView module, we employed nanoscale flow cytometry (termed nanoFCM) to examine the relative number and scatter distribution of plasma EVs at three different time points during the day in 6 healthy adults. Analysis of liposomes and plasma EVs proved that nanoFCM is capable of detecting biologically-relevant vesicles down to 100 nm in size. With this high resolution configuration, we observed variations in the relative size (FSC/SSC distributions) and concentration (proportions) of EVs in healthy adult plasma across the course of a day, suggesting that there are diurnal variations in the number and size distribution of circulating EV populations. The use of nanoFCM provides a valuable tool for the study of EVs in both health and disease; however, additional refinement of nanoscale flow cytometric methods is needed for use of these instruments for quantitative particle counting and sizing. Furthermore, larger scale studies are necessary to more clearly define the diurnal variations in circulating EVs, and thus further inform their use as biomarkers for disease.


Asunto(s)
Vesículas Extracelulares/fisiología , Citometría de Flujo , Adulto , Vesículas Extracelulares/química , Humanos , Liposomas/síntesis química , Liposomas/química , Microscopía de Fuerza Atómica , Tamaño de la Partícula
20.
Cell Host Microbe ; 18(3): 371-81, 2015 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-26355219

RESUMEN

During its life cycle, Plasmodium falciparum undergoes rapid proliferation fueled by de novo synthesis and acquisition of host cell lipids. Consistent with this essential role, Plasmodium lipid synthesis enzymes are emerging as potential drug targets. To explore their broader potential for therapeutic interventions, we assayed the global lipid landscape during P. falciparum sexual and asexual blood stage (ABS) development. Using liquid chromatography-mass spectrometry, we analyzed 304 lipids constituting 24 classes in ABS parasites, infected red blood cell (RBC)-derived microvesicles, gametocytes, and uninfected RBCs. Ten lipid classes were previously uncharacterized in P. falciparum, and 70%-75% of the lipid classes exhibited changes in abundance during ABS and gametocyte development. Utilizing compounds that target lipid metabolism, we affirmed the essentiality of major classes, including triacylglycerols. These studies highlight the interplay between host and parasite lipid metabolism and provide a comprehensive analysis of P. falciparum lipids with candidate pathways for drug discovery efforts.


Asunto(s)
Metabolismo de los Lípidos , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/metabolismo , Cromatografía Liquida , Lípidos/análisis , Espectrometría de Masas , Plasmodium falciparum/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA