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











Base de datos
Intervalo de año de publicación
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.
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.

3.
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
4.
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
5.
J Vis Exp ; (100): e52911, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26132420

RESUMEN

When cytotoxic T lymphocytes (CTL) or natural killer (NK) cells recognize tumor cells or cells infected with intracellular pathogens, they release their cytotoxic granule content to eliminate the target cells and the intracellular pathogen. Death of the host cells and intracellular pathogens is triggered by the granule serine proteases, granzymes (Gzms), delivered into the host cell cytosol by the pore forming protein perforin (PFN) and into bacterial pathogens by the prokaryotic membrane disrupting protein granulysin (GNLY). To investigate the molecular mechanisms of target cell death mediated by the Gzms in experimental in-vitro settings, protein expression and purification systems that produce high amounts of active enzymes are necessary. Mammalian secreted protein expression systems imply the potential to produce correctly folded, fully functional protein that bears posttranslational modification, such as glycosylation. Therefore, we used a cost-efficient calcium precipitation method for transient transfection of HEK293T cells with human Gzms cloned into the expression plasmid pHLsec. Gzm purification from the culture supernatant was achieved by immobilized nickel affinity chromatography using the C-terminal polyhistidine tag provided by the vector. The insertion of an enterokinase site at the N-terminus of the protein allowed the generation of active protease that was finally purified by cation exchange chromatography. The system was tested by producing high levels of cytotoxic human Gzm A, B and M and should be capable to produce virtually every enzyme in the human body in high yields.


Asunto(s)
Granzimas/biosíntesis , Transfección/métodos , Calcio/química , Técnicas de Cultivo de Célula/métodos , Cromatografía de Afinidad/métodos , Enteropeptidasa/química , Granzimas/genética , Granzimas/aislamiento & purificación , Células HEK293 , Humanos , Plásmidos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA