Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
PLoS Pathog ; 16(2): e1008240, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32106253

RESUMEN

Cryptococcus neoformans is an opportunistic human pathogen, which causes serious disease in immunocompromised hosts. Infection with this pathogen is particularly relevant in HIV+ patients, where it leads to around 200,000 deaths per annum. A key feature of cryptococcal pathogenesis is the ability of the fungus to survive and replicate within the phagosome of macrophages, as well as its ability to be expelled from host cells via a novel non-lytic mechanism known as vomocytosis. Here we show that cryptococcal vomocytosis from macrophages is strongly enhanced by viral coinfection, without altering phagocytosis or intracellular proliferation of the fungus. This effect occurs with distinct, unrelated human viral pathogens and is recapitulated when macrophages are stimulated with the anti-viral cytokines interferon alpha or beta (IFNα or IFNß). Importantly, the effect is abrogated when type-I interferon signalling is blocked, thus underscoring the importance of type-I interferons in this phenomenon. Lastly, our data help resolve previous, contradictory animal studies on the impact of type I interferons on cryptococcal pathogenesis and suggest that secondary viral stimuli may alter patterns of cryptococcal dissemination in the host.


Asunto(s)
Coinfección , Criptococosis , Cryptococcus neoformans , Infecciones por VIH , VIH-1 , Macrófagos , Coinfección/inmunología , Coinfección/microbiología , Coinfección/patología , Coinfección/virología , Criptococosis/inmunología , Criptococosis/microbiología , Criptococosis/patología , Criptococosis/virología , Cryptococcus neoformans/inmunología , Cryptococcus neoformans/patogenicidad , Células HEK293 , Infecciones por VIH/inmunología , Infecciones por VIH/microbiología , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/patogenicidad , Humanos , Interferón-alfa/inmunología , Interferón beta/inmunología , Macrófagos/inmunología , Macrófagos/patología , Macrófagos/virología , Transducción de Señal/inmunología
2.
Cell Microbiol ; 22(2): e13145, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31730731

RESUMEN

Vomocytosis, or nonlytic exocytosis, has been reported for Cryptococcus neoformans since 2006. Since then, the repertoire of vomocytosing pathogens and host cells has increased and so have the molecular components linked to vomocytosis occurrence. Nonetheless, the mechanism underlying this phenomenon, whether it is triggered by the host or the pathogen, and how it affects disease progression are still unresolved. This review contains a summary of the main findings regarding vomocytosis and the outstanding questions puzzling scientists to this day.


Asunto(s)
Criptococosis/microbiología , Cryptococcus neoformans/metabolismo , Exocitosis , Macrófagos/inmunología , Animales , Línea Celular , Humanos , Macrófagos/citología
3.
J Immunol ; 200(10): 3539-3546, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29643192

RESUMEN

The pathogenic fungus Cryptococcus enters the human host via inhalation into the lung and is able to reside in a niche environment that is serum- (opsonin) limiting. Little is known about the mechanism by which nonopsonic phagocytosis occurs via phagocytes in such situations. Using a combination of soluble inhibitors of phagocytic receptors and macrophages derived from knockout mice and human volunteers, we show that uptake of nonopsonized Cryptococcus neoformans and C. gattii via the mannose receptor is dependent on macrophage activation by cytokines. However, although uptake of C. neoformans is via both dectin-1 and dectin-2, C. gattii uptake occurs largely via dectin-1. Interestingly, dectin inhibitors also blocked phagocytosis of unopsonized Cryptococci in wax moth (Galleria mellonella) larvae and partially protected the larvae from infection by both fungi, supporting a key role for host phagocytes in augmenting early disease establishment. Finally, we demonstrated that internalization of nonopsonized Cryptococci is not accompanied by the nuclear translocation of NF-κB or its concomitant production of proinflammatory cytokines such as TNF-α. Thus, nonopsonized Cryptococci are recognized by mammalian phagocytes in a manner that minimizes proinflammatory cytokine production and potentially facilitates fungal pathogenesis.


Asunto(s)
Criptococosis/metabolismo , Criptococosis/microbiología , Cryptococcus gattii/patogenicidad , Cryptococcus neoformans/patogenicidad , Macrófagos/metabolismo , Macrófagos/microbiología , Animales , Línea Celular , Citocinas/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Mariposas Nocturnas , FN-kappa B/metabolismo , Proteínas Opsoninas/metabolismo , Fagocitos/metabolismo , Fagocitos/microbiología , Fagocitosis/fisiología , Receptores de Superficie Celular/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
4.
Infect Immun ; 87(12)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31570562

RESUMEN

The larval stage of the cestode Echinococcus granulosus causes cystic echinococcosis in humans and livestock. This larva is protected by the millimeter-thick, mucin-based laminated layer (LL), from which materials have to be shed to allow parasite growth. We previously reported that dendritic cells (DCs) respond to microscopic pieces of the mucin gel of the LL (pLL) with unconventional maturation phenotypes, in the absence or presence of Toll-like receptor (TLR) agonists, including lipopolysaccharide (LPS). We also reported that the presence of pLL inhibited the activating phosphorylation of the phosphatidylinositol 3-kinase (PI3K) effector Akt induced by granulocyte-macrophage colony-stimulating factor or interleukin-4. We now show that the inhibitory effect of pLL extends to LPS as a PI3K activator, and results in diminished phosphorylation of GSK3 downstream from Akt. Functionally, the inhibition of Akt and GSK3 phosphorylation are linked to the blunted upregulation of CD40, a major feature of the unconventional maturation phenotype. Paradoxically, all aspects of unconventional maturation induced by pLL depend on PI3K class I. Additional components of the phagocytic machinery are needed, but phagocytosis of pLL particles is not required. These observations hint at a DC response mechanism related to receptor-independent mechanisms proposed for certain crystalline and synthetic polymer-based particles; this would fit the previously reported lack of detection of molecular-level motifs necessary of the effects of pLL on DCs. Finally, we report that DCs exposed to pLL are able to condition DCs not exposed to the material so that these cannot upregulate CD40 in full in response to LPS.


Asunto(s)
Antígenos CD40/biosíntesis , Células Dendríticas/inmunología , Echinococcus granulosus/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Células Cultivadas , Equinococosis/inmunología , Equinococosis/parasitología , Equinococosis/patología , Activación Enzimática/inmunología , Glucógeno Sintasa Quinasa 3/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-4/metabolismo , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Fagocitosis/fisiología , Fosforilación , Transducción de Señal/inmunología
5.
Exp Parasitol ; 158: 23-30, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25816974

RESUMEN

The laminated layer is the unique mucin-based extracellular matrix that protects Echinococcus larvae, and thus to an important extent, shapes host-parasite relationships in the larval echinococcoses. In 2011, we published twin reviews summarizing what was known about this structure. Since then, important advances have been made. Complete genomes and some RNAseq data are now available for E. multilocularis and E. granulosus, leading to the inference that the E. multilocularis LL is probably formed by a single type of mucin backbone, while a second apomucin subfamily additionally contributes to the E. granulosus LL. Previously suspected differences between E. granulosus and E. multilocularis in mucin glycan size have been confirmed and pinned down to the virtual absence of Galß1-3 chains in E. multilocularis. The LL carbohydrates from both species have been found to interact selectively with the Kupffer cell receptor expressed in rodent liver macrophages, highlighting the ancestral adaptations to rodents as intermediate hosts and to the liver as infection site. Finally, LL particles have been shown to possess carbohydrate-independent mechanisms profoundly conditioning non-liver-specific dendritic cells and macrophages. These advances are discussed in an integrated way, and in the context of the newly determined phylogeny of Echinococcus and its taenid relatives.


Asunto(s)
Echinococcus/fisiología , Mucinas/química , Animales , Evolución Biológica , Secuencia de Carbohidratos , Echinococcus/química , Echinococcus/genética , Echinococcus/inmunología , Mucinas Gástricas/química , Mucinas Gástricas/genética , Glicómica , Inmunidad Innata , Mucinas/genética , Mucinas/inmunología , Polisacáridos/química , Polisacáridos/genética
6.
Infect Immun ; 82(8): 3164-76, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24842926

RESUMEN

The larval stage of the cestode parasite Echinococcus granulosus causes hydatid disease in humans and livestock. This infection is characterized by the growth in internal organ parenchymae of fluid-filled structures (hydatids) that elicit surprisingly little inflammation in spite of their massive size and persistence. Hydatids are protected by a millimeter-thick layer of mucin-based extracellular matrix, termed the laminated layer (LL), which is thought to be a major factor determining the host response to the infection. Host cells can interact both with the LL surface and with materials that are shed from it to allow parasite growth. In this work, we analyzed the response of dendritic cells (DCs) to microscopic pieces of the native mucin-based gel of the LL (pLL). In vitro, this material induced an unusual activation state characterized by upregulation of CD86 without concomitant upregulation of CD40 or secretion of cytokines (interleukin 12 [IL-12], IL-10, tumor necrosis factor alpha [TNF-α], and IL-6). When added to Toll-like receptor (TLR) agonists, pLL-potentiated CD86 upregulation and IL-10 secretion while inhibiting CD40 upregulation and IL-12 secretion. In vivo, pLL also caused upregulation of CD86 and inhibited CD40 upregulation in DCs. Contrary to expectations, oxidation of the mucin glycans in pLL with periodate did not abrogate the effects on cells. Reduction of disulfide bonds, which are known to be important for LL structure, strongly diminished the impact of pLL on DCs without altering the particulate nature of the material. In summary, DCs respond to the LL mucin meshwork with a "semimature" activation phenotype, both in vitro and in vivo.


Asunto(s)
Antígenos Helmínticos/inmunología , Células Dendríticas/inmunología , Echinococcus granulosus/inmunología , Animales , Antígeno B7-2/análisis , Antígenos CD40/análisis , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Larva/inmunología , Ratones Endogámicos C57BL
8.
Cell Death Discov ; 9(1): 469, 2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38129373

RESUMEN

The NLRP3 inflammasome is a component of the inflammatory response to infection and injury, orchestrating the maturation and release of the pro-inflammatory cytokines interleukin-1ß (IL-1ß), IL-18, and triggering pyroptotic cell death. Appropriate levels of NLRP3 activation are needed to avoid excessive tissue damage while ensuring host protection. Here we report a role for symmetrical diarylsquaramides as selective K+ efflux-dependent NLRP3 inflammasome enhancers. Treatment of macrophages with squaramides potentiated IL-1ß secretion and ASC speck formation in response to K+ efflux-dependent NLRP3 inflammasome activators without affecting priming, endosome cargo trafficking, or activation of other inflammasomes. The squaramides lowered intracellular K+ concentration which enabled cells to respond to a below-threshold dose of the inflammasome activator nigericin. Taken together these data further highlight the role of ion flux in inflammasome activation and squaramides as an interesting platform for therapeutic development in conditions where enhanced NLRP3 activity could be beneficial.

9.
Sci Signal ; 16(773): eabm7134, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36809026

RESUMEN

Inflammation driven by the NLRP3 inflammasome is coordinated through multiple signaling pathways and is regulated by subcellular organelles. Here, we tested the hypothesis that NLRP3 senses disrupted endosome trafficking to trigger inflammasome formation and inflammatory cytokine secretion. NLRP3-activating stimuli disrupted endosome trafficking and triggered localization of NLRP3 to vesicles positive for endolysosomal markers and for the inositol lipid PI4P. Chemical disruption of endosome trafficking sensitized macrophages to the NLRP3 activator imiquimod, driving enhanced inflammasome activation and cytokine secretion. Together, these data suggest that NLRP3 can sense disruptions in the trafficking of endosomal cargoes, which may explain in part the spatial activation of the NLRP3 inflammasome. These data highlight mechanisms that could be exploited in the therapeutic targeting of NLRP3.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Caspasa 1/metabolismo , Macrófagos/metabolismo , Citocinas/metabolismo , Interleucina-1beta/metabolismo
10.
J Cell Biol ; 219(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33044555

RESUMEN

Diverse pathogen- and damage-associated stresses drive inflammation via activation of the multimolecular NLRP3-inflammasome complex. How the effects of diverse stimuli are integrated by the cell to regulate NLRP3 has been the subject of intense research, and yet an accepted unifying hypothesis for the control of NLRP3 remains elusive. Here, we review the literature on the effects of NLRP3-activating stimuli on subcellular organelles and conclude that a shared feature of NLRP3-activating stresses is an organelle dysfunction. In particular, we propose that the endosome may be more important than previously recognized as a signal-integrating hub for NLRP3 activation in response to many stimuli and may also link to the dysfunction of other organelles. In addition, NLRP3-inflammasome-activating stimuli trigger diverse posttranslational modifications of NLRP3 that are important in controlling its activation. Future research should focus on how organelles respond to specific NLRP3-activating stimuli, and how this relates to posttranslational modifications, to delineate the organellar control of NLRP3.


Asunto(s)
Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Orgánulos/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Estrés Fisiológico , Animales , Humanos , Inflamasomas/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Orgánulos/genética
11.
Immunobiology ; 224(5): 710-719, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31178241

RESUMEN

Persistent extracellular tissue-dwelling pathogens face the challenge of antibody-dependent activation of the classical complement pathway (CCP). A prime example of this situation is the larva of the cestode Echinococcus granulosus sensu lato, causing cystic echinococcosis. This tissue-dwelling, bladder-like larva is bounded by a cellular layer protected by the outermost acellular "laminated layer" (LL), to which host antibodies bind. The LL is made up of a mucin meshwork and interspersed nano-deposits of calcium inositol hexakisphosphate (calcium InsP6). We previously reported that calcium InsP6 bound C1q, apparently initiating CCP activation. The present work dissects CCP activation on the LL. Most of the C1 binding activity in the LL corresponded to calcium InsP6, and this binding was enhanced by partial proteolysis of the mucin meshwork. The remaining C1 binding activity was attributable to host antibodies, which included CCP-activating IgG isotypes. Calcium InsP6 made only a weak contribution to early CCP activation on the LL, suggesting inefficient C1 complex activation as reported for other polyanions. CCP activation on calcium InsP6 gave rise to a dominant population of C3b deposited onto calcium InsP6 itself that appeared to be quickly inactivated. Apparently as a result of inefficient initiation plus C3b inactivation, calcium InsP6 made no net contribution to C5 activation. We propose that the LL protects the underlying parasite cells from CCP activation through the combined effects of inefficient permeation of C1 through the mucins and C1 retention on calcium InsP6. This mechanism does not result in C5 activation, which is known to drive parasite-damaging inflammation.


Asunto(s)
Antígenos Helmínticos/inmunología , Vía Clásica del Complemento , Proteínas del Sistema Complemento/inmunología , Echinococcus granulosus/inmunología , Ácido Fítico/inmunología , Animales , Antígenos Helmínticos/química , Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Equinococosis/inmunología , Equinococosis/parasitología , Interacciones Huésped-Parásitos/inmunología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Ácido Fítico/química , Unión Proteica
12.
Sci Adv ; 3(8): e1700898, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28835924

RESUMEN

Vomocytosis, or nonlytic extrusion, is a poorly understood process through which macrophages release live pathogens that they have failed to kill back into the extracellular environment. Vomocytosis is conserved across vertebrates and occurs with a diverse range of pathogens, but to date, the host signaling events that underpin expulsion remain entirely unknown. We use a targeted inhibitor screen to identify the MAP kinase ERK5 as a critical suppressor of vomocytosis. Pharmacological inhibition or genetic manipulation of ERK5 activity significantly raises vomocytosis rates in human macrophages, whereas stimulation of the ERK5 signaling pathway inhibits vomocytosis. Lastly, using a zebrafish model of cryptococcal disease, we show that reducing ERK5 activity in vivo stimulates vomocytosis and results in reduced dissemination of infection. ERK5 therefore represents the first host signaling regulator of vomocytosis to be identified and a potential target for the future development of vomocytosis-modulating therapies.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Línea Celular , Citocinas/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Pez Cebra
13.
Sci Rep ; 6: 39204, 2016 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-27966637

RESUMEN

Proliferation of macrophages is a hallmark of inflammation in many type 2 settings including helminth infections. The cellular expansion is driven by the type 2 cytokine interleukin-4 (IL-4), as well as by M-CSF, which also controls homeostatic levels of tissue resident macrophages. Cystic echinococcosis, caused by the tissue-dwelling larval stage of the cestode Echinococcus granulosus, is characterised by normally subdued local inflammation. Infiltrating host cells make contact only with the acellular protective coat of the parasite, called laminated layer, particles of which can be ingested by phagocytic cells. Here we report that a particulate preparation from this layer (pLL) strongly inhibits the proliferation of macrophages in response to IL-4 or M-CSF. In addition, pLL also inhibits IL-4-driven up-regulation of Relm-α, without similarly affecting Chitinase-like 3 (Chil3/Ym1). IL-4-driven cell proliferation and up-regulation of Relm-α are both known to depend on the phosphatidylinositol (PI3K)/Akt pathway, which is dispensable for induction of Chil3/Ym1. Exposure to pLL in vitro inhibited Akt activation in response to proliferative stimuli, providing a potential mechanism for its activities. Our results suggest that the E. granulosus laminated layer exerts some of its anti-inflammatory properties through inhibition of PI3K/Akt activation and consequent limitation of macrophage proliferation.


Asunto(s)
Equinococosis/inmunología , Echinococcus granulosus/metabolismo , Proteínas del Helminto/inmunología , Macrófagos/inmunología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Regulación hacia Abajo , Femenino , Interleucina-4/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Fagocitosis , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA