Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA