Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur J Immunol ; 54(5): e2350515, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38361219

RESUMEN

Caspase-1 location in cells has been studied with fluorochrome-labeled inhibitors of caspase-1 (FLICA reagents). We report that FLICA reagents have limited cell-membrane permeability. This impacts experimental design as cells with intact membranes, including caspase-1 knockout cells, are not appropriate controls for cells with inflammasome-induced gasdermin D membrane pores.


Asunto(s)
Caspasa 1 , Inhibidores de Caspasas , Permeabilidad de la Membrana Celular , Colorantes Fluorescentes , Inflamasomas , Macrófagos , Caspasa 1/metabolismo , Animales , Macrófagos/inmunología , Macrófagos/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ratones , Inflamasomas/metabolismo , Inhibidores de Caspasas/farmacología , Ratones Noqueados , Proteínas de Unión a Fosfato/metabolismo , Humanos
2.
Immunity ; 45(2): 333-45, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27533014

RESUMEN

Many pathogens, including Plasmodium spp., exploit the interaction of programmed death-1 (PD-1) with PD-1-ligand-1 (PD-L1) to "deactivate" T cell functions, but the role of PD-L2 remains unclear. We studied malarial infections to understand the contribution of PD-L2 to immunity. Here we have shown that higher PD-L2 expression on blood dendritic cells, from Plasmodium falciparum-infected individuals, correlated with lower parasitemia. Mechanistic studies in mice showed that PD-L2 was indispensable for establishing effective CD4(+) T cell immunity against malaria, because it not only inhibited PD-L1 to PD-1 activity but also increased CD3 and inducible co-stimulator (ICOS) expression on T cells. Importantly, administration of soluble multimeric PD-L2 to mice with lethal malaria was sufficient to dramatically improve immunity and survival. These studies show immuno-regulation by PD-L2, which has the potential to be translated into an effective treatment for malaria and other diseases where T cell immunity is ineffective or short-lived due to PD-1-mediated signaling.


Asunto(s)
Antígeno B7-H1/metabolismo , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Adamantano/análogos & derivados , Adamantano/uso terapéutico , Adulto , Animales , Antimaláricos/uso terapéutico , Antígeno B7-H1/genética , Células Cultivadas , Ensayos Clínicos como Asunto , Células Dendríticas/parasitología , Femenino , Humanos , Inmunidad Celular , Activación de Linfocitos , Malaria Falciparum/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Parasitemia/inmunología , Peróxidos/uso terapéutico , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/genética , Pirimidinas/uso terapéutico , Triazoles/uso terapéutico , Adulto Joven
3.
EMBO J ; 39(17): e106202, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32869315

RESUMEN

Shigella, a major cause of bacterial dysentery, knows when it is not wanted. To generate and maintain its niche within host cells, this unwelcome guest injects several dozen virulence factors via a type 3 secretion system (T3SS). In this issue, Ashida et al (2020) have elucidated the role of two such factors from Shigella flexneri-OspC1 and OspD3-that together counteract apoptotic and necroptotic death pathways in colonised epithelial cells. As a result, Shigella can replicate to high levels within the colonic epithelium, leading to the substantial epithelial damage in shigellosis and efficient bacterial release for faecal transmission.


Asunto(s)
Disentería Bacilar , Shigella , Caspasa 8 , Muerte Celular , Células Epiteliales , Humanos , Shigella/genética , Shigella flexneri/genética
4.
Eur J Immunol ; 53(7): e2250056, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37058370

RESUMEN

TLRs engage numerous adaptor proteins and signaling molecules, enabling a complex series of post-translational modifications (PTMs) to mount inflammatory responses. TLRs themselves are post-translationally modified following ligand-induced activation, with this being required to relay the full spectrum of proinflammatory signaling responses. Here, we reveal indispensable roles for TLR4 Y672 and Y749 phosphorylation in mounting optimal LPS-inducible inflammatory responses in primary mouse macrophages. LPS promotes phosphorylation at both tyrosine residues, with Y749 phosphorylation being required for maintenance of total TLR4 protein levels and Y672 phosphorylation exerting its pro-inflammatory effects more selectively by initiating ERK1/2 and c-FOS phosphorylation. Our data also support a role for the TLR4-interacting membrane proteins SCIMP and the SYK kinase axis in mediating TLR4 Y672 phosphorylation to permit downstream inflammatory responses in murine macrophages. The corresponding residue in human TLR4 (Y674) is also required for optimal LPS signaling responses. Our study, thus, reveals how a single PTM on one of the most widely studied innate immune receptors orchestrates downstream inflammatory responses.


Asunto(s)
Citocinas , Lipopolisacáridos , Humanos , Animales , Ratones , Fosforilación , Citocinas/metabolismo , Lipopolisacáridos/farmacología , Receptor Toll-Like 4 , Tirosina/metabolismo , Tirosina/farmacología , Macrófagos
5.
J Virol ; 97(11): e0125123, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37850747

RESUMEN

IMPORTANCE: Dengue virus, an arbovirus, causes an estimated 100 million symptomatic infections annually and is an increasing threat as the mosquito range expands with climate change. Dengue epidemics are a substantial strain on local economies and health infrastructure, and an understanding of what drives severe disease may enable treatments to help reduce hospitalizations. Factors exacerbating dengue disease are debated, but gut-related symptoms are much more frequent in severe than mild cases. Using mouse models of dengue infection, we have shown that inflammation and damage are earlier and more severe in the gut than in other tissues. Additionally, we observed impairment of the gut mucus layer and propose that breakdown of the barrier function exacerbates inflammation and promotes severe dengue disease. This idea is supported by recent data from human patients showing elevated bacteria-derived molecules in dengue patient serum. Therapies aiming to maintain gut integrity may help to abrogate severe dengue disease.


Asunto(s)
Virus del Dengue , Dengue Grave , Animales , Humanos , Ratones , Culicidae , Virus del Dengue/fisiología , Inflamación/virología , Dengue Grave/patología , Cinética
6.
Mol Psychiatry ; 28(7): 2878-2893, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36316366

RESUMEN

Coronavirus disease-2019 (COVID-19) is primarily a respiratory disease, however, an increasing number of reports indicate that SARS-CoV-2 infection can also cause severe neurological manifestations, including precipitating cases of probable Parkinson's disease. As microglial NLRP3 inflammasome activation is a major driver of neurodegeneration, here we interrogated whether SARS-CoV-2 can promote microglial NLRP3 inflammasome activation. Using SARS-CoV-2 infection of transgenic mice expressing human angiotensin-converting enzyme 2 (hACE2) as a COVID-19 pre-clinical model, we established the presence of virus in the brain together with microglial activation and NLRP3 inflammasome upregulation in comparison to uninfected mice. Next, utilising a model of human monocyte-derived microglia, we identified that SARS-CoV-2 isolates can bind and enter human microglia in the absence of viral replication. This interaction of virus and microglia directly induced robust inflammasome activation, even in the absence of another priming signal. Mechanistically, we demonstrated that purified SARS-CoV-2 spike glycoprotein activated the NLRP3 inflammasome in LPS-primed microglia, in a ACE2-dependent manner. Spike protein also could prime the inflammasome in microglia through NF-κB signalling, allowing for activation through either ATP, nigericin or α-synuclein. Notably, SARS-CoV-2 and spike protein-mediated microglial inflammasome activation was significantly enhanced in the presence of α-synuclein fibrils and was entirely ablated by NLRP3-inhibition. Finally, we demonstrate SARS-CoV-2 infected hACE2 mice treated orally post-infection with the NLRP3 inhibitory drug MCC950, have significantly reduced microglial inflammasome activation, and increased survival in comparison with untreated SARS-CoV-2 infected mice. These results support a possible mechanism of microglial innate immune activation by SARS-CoV-2, which could explain the increased vulnerability to developing neurological symptoms akin to Parkinson's disease in COVID-19 infected individuals, and a potential therapeutic avenue for intervention.


Asunto(s)
COVID-19 , Enfermedad de Parkinson , Humanos , Ratones , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Microglía/metabolismo , alfa-Sinucleína/metabolismo , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo , COVID-19/metabolismo , Ratones Transgénicos
7.
Mol Cell ; 64(2): 236-250, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27746017

RESUMEN

Caspase-8 activation can be triggered by death receptor-mediated formation of the death-inducing signaling complex (DISC) and by the inflammasome adaptor ASC. Caspase-8 assembles with FADD at the DISC and with ASC at the inflammasome through its tandem death effector domain (tDED), which is regulated by the tDED-containing cellular inhibitor cFLIP and the viral inhibitor MC159. Here we present the caspase-8 tDED filament structure determined by cryoelectron microscopy. Extensive assembly interfaces not predicted by the previously proposed linear DED chain model were uncovered, and were further confirmed by structure-based mutagenesis in filament formation in vitro and Fas-induced apoptosis and ASC-mediated caspase-8 recruitment in cells. Structurally, the two DEDs in caspase-8 use quasi-equivalent contacts to enable assembly. Using the tDED filament structure as a template, structural analyses reveal the interaction surfaces between FADD and caspase-8 and the distinct mechanisms of regulation by cFLIP and MC159 through comingling and capping, respectively.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/química , Caspasa 8/química , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/química , Proteína de Dominio de Muerte Asociada a Fas/química , Proteínas Virales/química , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Sitios de Unión , Proteínas Adaptadoras de Señalización CARD , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Microscopía por Crioelectrón , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Dominio Efector de Muerte , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Expresión Génica , Humanos , Células Jurkat , Plásmidos/química , Plásmidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Proteínas Virales/genética , Proteínas Virales/metabolismo , Receptor fas/farmacología
8.
J Enzyme Inhib Med Chem ; 39(1): 2313055, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38416868

RESUMEN

Toll-like receptor (TLR) innate immunity signalling protects against pathogens, but excessive or prolonged signalling contributes to a range of inflammatory conditions. Structural information on the TLR cytoplasmic TIR (Toll/interleukin-1 receptor) domains and the downstream adaptor proteins can help us develop inhibitors targeting this pathway. The small molecule o-vanillin has previously been reported as an inhibitor of TLR2 signalling. To study its mechanism of action, we tested its binding to the TIR domain of the TLR adaptor MAL/TIRAP (MALTIR). We show that o-vanillin binds to MALTIR and inhibits its higher-order assembly in vitro. Using NMR approaches, we show that o-vanillin forms a covalent bond with lysine 210 of MAL. We confirm in mouse and human cells that o-vanillin inhibits TLR2 but not TLR4 signalling, independently of MAL, suggesting it may covalently modify TLR2 signalling complexes directly. Reactive aldehyde-containing small molecules such as o-vanillin may target multiple proteins in the cell.


Asunto(s)
Benzaldehídos , Lisina , Receptor Toll-Like 2 , Humanos , Animales , Ratones , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores Toll-Like/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Interleucina-1/metabolismo
9.
J Biol Chem ; 298(12): 102666, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36334634

RESUMEN

Conventional assays to probe signaling protein interactions and function involve measurement of luciferase reporter expression within the bulk cell population, with lack of control over target-protein expression level. To address this issue, we have developed a rapid and robust flow cytometric assay for analysis of signaling protein function. A fluorescent reporter and fluorescent tagging of the target protein enables simultaneous assessment of protein expression and signaling within individual cells. We have applied our technique to the analysis of variants of the lipopolysaccharide receptor Toll-like receptor 4 (TLR4) and its adapter protein MyD88, using a NF-кB-responsive promoter driving mScarlet-I expression. The assay enables exclusion of nontransfected cells and overexpressing cells that signal spontaneously. Additionally, our assay allows the identification of protein variants that fail to express. We found that the assays were highly sensitive, with cells expressing an appropriate level of GFP-MyD88 showing approximately 200-fold induction of mScarlet-I by lipopolysaccharide, and we can detect subtle protein concentration-dependent effects of mutations. Importantly, the assay is adaptable to various signaling pathways.


Asunto(s)
Lipopolisacáridos , Factor 88 de Diferenciación Mieloide , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal , Humanos
10.
EMBO Rep ; 20(9): e48891, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31379068

RESUMEN

The non-canonical inflammasome mediates pyroptotic cell death in response to bacterial lipopolysaccharide (LPS) found in the cytosol. Understanding the mechanism and regulation of this system is of great interest, given its central role in mouse models of bacterial septic shock. In this issue of EMBO Reports, Benaoudia and colleagues sought to discover extra players in the human non-canonical inflammasome using a CRISPR library screen; the only strongly positive hit apart from the known components caspase-4 and gasdermin D was interferon regulatory factor-2 (IRF2) [1 ]. IRF2 was found to be a transcriptional activator of caspase-4, and in its absence, induction of IRF1 could substitute to maintain caspase-4 expression.


Asunto(s)
Inflamasomas , Lipopolisacáridos , Animales , Humanos , Factor 1 Regulador del Interferón , Factor 2 Regulador del Interferón , Ratones
11.
Immunol Cell Biol ; 97(1): 17-28, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30052286

RESUMEN

Inflammasomes are protein complexes activated by infection and cellular stress that promote caspase-1 activation and subsequent inflammatory cytokine processing and cell death. It has been anticipated that inflammasome activity contributes to autoimmunity. However, we previously showed that macrophages from autoimmune New Zealand Black (NZB) mice lack NLRP3 inflammasome function, and their absent in melanoma 2 (AIM2) inflammasome responses are compromised by high expression of the AIM2 antagonist protein p202. Here we found that the point mutation leading to lack of NLRP3 expression occurred early in the NZB strain establishment, as it is shared with the related obese strain New Zealand Obese, but not with the unrelated New Zealand White (NZW) strain. The first cross progeny of NZB and NZW mice develop more severe lupus nephritis than the NZB strain. We have compared AIM2 and NLRP3 inflammasome function in macrophages from NZB, NZW, and NZB/W F1 mice. The NZW parental strain showed strong inflammasome function, whereas the NZB/W F1 have haploinsufficient expression of NLRP3 and show reduced NLRP3 and AIM2 inflammasome responses, particularly at low stimulus strength. It remains to be established whether the low inflammasome function could contribute to loss of tolerance and the onset of autoimmunity in NZB and NZB/W F1. However, with amplifying inflammatory stimuli through the course of disease, the NLRP3 response in the NZB/W F1 may be sufficient to contribute to kidney damage at later stages of disease.


Asunto(s)
Autoinmunidad , Proteínas de Unión al ADN/deficiencia , Inflamasomas , Macrófagos , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Animales , Autoinmunidad/genética , Proteínas de Unión al ADN/inmunología , Femenino , Inflamasomas/genética , Nefritis Lúpica/genética , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos NZB , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Mutación Puntual
12.
Immunol Cell Biol ; 96(10): 1120-1130, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30003588

RESUMEN

Outer membrane vesicles (OMVs) are constitutively produced by Gram-negative bacteria both in vivo and in vitro. These lipid-bound structures carry a range of immunogenic components derived from the parent cell, which are transported into host target cells and activate the innate immune system. Recent advances in the field have shed light on some of the multifaceted roles of OMVs in host-pathogen interactions. In this study, we investigated the ability of OMVs from two clinically important pathogens, Pseudomonas aeruginosa and Helicobacter pylori, to activate canonical and noncanonical inflammasomes. P. aeruginosa OMVs induced inflammasome activation in mouse macrophages, as evidenced by "speck" formation, as well as the cleavage and secretion of interleukin-1ß and caspase-1. These responses were independent of AIM2 and NLRC4 canonical inflammasomes, but dependent on the noncanonical caspase-11 pathway. Moreover, P. aeruginosa OMVs alone were able to activate the inflammasome in a TLR-dependent manner, without requiring an exogenous priming signal. In contrast, H. pylori OMVs were not able to induce inflammasome activation in macrophages. Using CRISPR/Cas9 knockout THP-1 cells lacking the human caspase-11 homologs, caspase-4 and -5,we demonstrated that caspase-5 but not caspase-4 is required for inflammasome activation by P. aeruginosa OMVs in human monocytes. In contrast, free P. aeruginosa lipopolysaccharide (LPS) transfected into cells induced inflammasome responses via caspase-4. This suggests that caspase-4 and caspase-5 differentially recognize LPS depending on its physical form or route of delivery into the cell. These findings have relevance to Gram-negative infections in humans and the use of OMVs as novel vaccines.


Asunto(s)
Caspasas/metabolismo , Vesículas Extracelulares/metabolismo , Inflamasomas/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiología , Caspasa 1/metabolismo , Línea Celular , Humanos , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Infecciones por Pseudomonas/microbiología , Transducción de Señal
13.
Adv Exp Med Biol ; 1062: 89-106, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29845527

RESUMEN

Recent structural and functional advances provide fresh insight into the biology of the dengue virus non-structural protein, NS1 and suggest new avenues of research. The work of our lab and others have shown that the secreted, hexameric form of NS1 has a systemic toxic effect, inducing inflammatory cytokines and acting directly on endothelial cells to produce the hallmark of dengue disease, vascular leak. We also demonstrated that NS1 exerts its toxic activity through recognition by the innate immune receptor TLR4, mimicking the bacterial endotoxin LPS. This monograph covers the background underpinning these new findings and discusses new avenues for antiviral and vaccine intervention.


Asunto(s)
Virus del Dengue/inmunología , Dengue Grave/virología , Proteínas no Estructurales Virales/inmunología , Animales , Citocinas/genética , Citocinas/inmunología , Virus del Dengue/química , Virus del Dengue/genética , Células Endoteliales/inmunología , Células Endoteliales/virología , Humanos , Receptores Virales/genética , Receptores Virales/inmunología , Dengue Grave/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
14.
Immunol Cell Biol ; 95(5): 491-495, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28220810

RESUMEN

The secreted hexameric form of the dengue virus (DENV) non-structural protein 1 (NS1) has recently been shown to elicit inflammatory cytokine release and disrupt endothelial cell monolayer integrity. This suggests that circulating NS1 contributes to the vascular leak that plays a major role in the pathology of dengue haemorrhagic fever and shock. Pathways activated by NS1 are thus of great interest as potential therapeutic targets. Recent works have separately implicated both toll-like receptor 4 (TLR4) and the TLR2/6 heterodimer in immune cell activation by NS1. Here we have used mouse gene knockout macrophages and antibodies blocking TLR function in human peripheral blood mononuclear cells to show that recombinant NS1, expressed and purified from eukaryotic cells, induces cytokine production via TLR4 but not TLR2/6. Furthermore, the commercial Escherichia coli-derived recombinant NS1 preparation used in other work to implicate TLR2/6 in the response is not correctly folded and appears to be contaminated by several microbial TLR ligands. Thus TLR4 remains a therapeutic target for DENV infections, with TLR4 antagonists holding promise for the treatment of dengue disease.


Asunto(s)
Virus del Dengue/inmunología , Leucocitos/virología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 6/metabolismo , Proteínas no Estructurales Virales/inmunología , Animales , Virus del Dengue/efectos de los fármacos , Escherichia coli/metabolismo , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/patología , Lipopolisacáridos , Ratones Endogámicos C57BL , Polimixina B/farmacología , Multimerización de Proteína/efectos de los fármacos
15.
FASEB J ; 30(5): 1901-12, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26839376

RESUMEN

We aimed to characterize antimicrobial zinc trafficking within macrophages and to determine whether the professional intramacrophage pathogen Salmonella enterica serovar Typhimurium (S Typhimurium) subverts this pathway. Using both Escherichia coli and S Typhimurium, we show that TLR signaling promotes the accumulation of vesicular zinc within primary human macrophages. Vesicular zinc is delivered to E. coli to promote microbial clearance, whereas S. Typhimurium evades this response via Salmonella pathogenicity island (SPI)-1. Even in the absence of SPI-1 and the zinc exporter ZntA, S Typhimurium resists the innate immune zinc stress response, implying the existence of additional host subversion mechanisms. We also demonstrate the combinatorial antimicrobial effects of zinc and copper, a pathway that S. Typhimurium again evades. Our use of complementary tools and approaches, including confocal microscopy, direct assessment of intramacrophage bacterial zinc stress responses, specific E. coli and S Typhimurium mutants, and inductively coupled plasma mass spectroscopy, has enabled carefully controlled characterization of this novel innate immune antimicrobial pathway. In summary, our study provides new insights at the cellular level into the well-documented effects of zinc in promoting host defense against infectious disease, as well as the complex host subversion strategies employed by S Typhimurium to combat this pathway.-Kapetanovic, R., Bokil, N. J., Achard, M. E. S., Ong, C.-L. Y., Peters, K. M., Stocks, C. J., Phan, M.-D., Monteleone, M., Schroder, K., Irvine, K. M., Saunders, B. M., Walker, M. J., Stacey, K. J., McEwan, A. G., Schembri, M. A., Sweet, M. J. Salmonella employs multiple mechanisms to subvert the TLR-inducible zinc-mediated antimicrobial response of human macrophages.


Asunto(s)
Macrófagos/inmunología , Macrófagos/metabolismo , Salmonella typhimurium/fisiología , Salmonella/fisiología , Receptores Toll-Like/metabolismo , Zinc/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Células Cultivadas , Cobre , Vesículas Citoplasmáticas/química , Vesículas Citoplasmáticas/metabolismo , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Toll-Like/genética
16.
J Immunol ; 194(1): 455-62, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25404358

RESUMEN

Inflammasomes are large protein complexes induced by a wide range of microbial, stress, and environmental stimuli that function to induce cell death and inflammatory cytokine processing. Formation of an inflammasome involves dramatic relocalization of the inflammasome adapter protein apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) into a single speck. We have developed a flow cytometric assay for inflammasome formation, time of flight inflammasome evaluation, which detects the change in ASC distribution within the cell. The transit of ASC into the speck is detected by a decreased width or increased height of the pulse of emitted fluorescence. This assay can be used to quantify native inflammasome formation in subsets of mixed cell populations ex vivo. It can also provide a rapid and sensitive technique for investigating molecular interactions in inflammasome formation, by comparison of wild-type and mutant proteins in inflammasome reconstitution experiments.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Citometría de Flujo/métodos , Inflamasomas/inmunología , Animales , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/genética , Células de la Médula Ósea/inmunología , Proteínas Adaptadoras de Señalización CARD/inmunología , Caspasa 1/genética , Línea Celular , Células HEK293 , Humanos , Inflamasomas/análisis , Mediadores de Inflamación/inmunología , Macrófagos/inmunología , Ratones , Ratones Noqueados
17.
J Immunol ; 195(3): 1233-41, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26116505

RESUMEN

Inflammasomes are protein complexes that promote caspase activation, resulting in processing of IL-1ß and cell death, in response to infection and cellular stresses. Inflammasomes have been anticipated to contribute to autoimmunity. The New Zealand Black (NZB) mouse develops anti-erythrocyte Abs and is a model of autoimmune hemolytic anemia. These mice also develop anti-nuclear Abs typical of lupus. In this article, we show that NZB macrophages have deficient inflammasome responses to a DNA virus and fungal infection. Absent in melanoma 2 (AIM2) inflammasome responses are compromised in NZB by high expression of the AIM 2 antagonist protein p202, and consequently NZB cells had low IL-1ß output in response to both transfected DNA and mouse CMV infection. Surprisingly, we also found that a second inflammasome system, mediated by the NLR family, pyrin domain containing 3 (NLRP3) initiating protein, was completely lacking in NZB cells. This was due to a point mutation in an intron of the Nlrp3 gene in NZB mice, which generates a novel splice acceptor site. This leads to incorporation of a pseudoexon with a premature stop codon. The lack of full-length NLRP3 protein results in NZB being effectively null for Nlrp3, with no production of bioactive IL-1ß in response to NLRP3 stimuli, including infection with Candida albicans. Thus, this autoimmune strain harbors two inflammasome deficiencies, mediated through quite distinct mechanisms. We hypothesize that the inflammasome deficiencies in NZB alter the interaction of the host with both microflora and pathogens, promoting prolonged production of cytokines that contribute to development of autoantibodies.


Asunto(s)
Anemia Hemolítica Autoinmune/genética , Proteínas Portadoras/genética , Proteínas de Unión al ADN/genética , Inflamasomas/genética , Macrófagos/inmunología , Anemia Hemolítica Autoinmune/inmunología , Animales , Anticuerpos Antinucleares/inmunología , Autoinmunidad/genética , Autoinmunidad/inmunología , Candida albicans/inmunología , Candidiasis/inmunología , Candidiasis/microbiología , Proteínas Portadoras/inmunología , Caspasa 1/genética , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/inmunología , Inflamasomas/inmunología , Interleucina-1beta/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Ratones , Ratones Endogámicos NZB , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal/genética , Transducción de Señal/inmunología
18.
J Biol Chem ; 290(49): 29217-30, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26468282

RESUMEN

Inflammasomes mediate inflammatory and cell death responses to pathogens and cellular stress signals via activation of procaspases-1 and -8. During inflammasome assembly, activated receptors of the NLR or PYHIN family recruit the adaptor protein ASC and initiate polymerization of its pyrin domain (PYD) into filaments. We show that ASC filaments in turn nucleate procaspase-8 death effector domain (DED) filaments in vitro and in vivo. Interaction between ASC PYD and procaspase-8 tandem DEDs optimally required both DEDs and represents an unusual heterotypic interaction between domains of the death fold superfamily. Analysis of ASC PYD mutants showed that interaction surfaces that mediate procaspase-8 interaction overlap with those required for ASC self-association and interaction with the PYDs of inflammasome initiators. Our data indicate that multiple types of death fold domain filaments form at inflammasomes and that PYD/DED and homotypic PYD interaction modes are similar. Interestingly, we observed condensation of procaspase-8 filaments containing the catalytic domain, suggesting that procaspase-8 interactions within and/or between filaments may be involved in caspase-8 activation. Procaspase-8 filaments may also be relevant to apoptosis induced by death receptors.


Asunto(s)
Caspasa 8/metabolismo , Proteínas del Citoesqueleto/metabolismo , Inflamasomas/metabolismo , Apoptosis , Proteínas Adaptadoras de Señalización CARD , Caspasa 1/metabolismo , Dominio Catalítico , Muerte Celular , Células HEK293 , Humanos , Inflamación , Microscopía Fluorescente , Mutación , Unión Proteica , Transducción de Señal
19.
Immunol Cell Biol ; 94(5): 520-4, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26833024

RESUMEN

Inflammasomes are molecular complexes activated by infection and cellular stress, leading to caspase-1 activation and subsequent interleukin-1ß (IL-1ß) processing and cell death. The autoimmune NZB mouse strain does not express NLRP3, a key inflammasome initiator mediating responses to a wide variety of stimuli including endogenous danger signals, environmental irritants and a range of bacterial, fungal and viral pathogens. We have previously identified an intronic point mutation in the Nlrp3 gene from NZB mice that generates a splice acceptor site. This leads to inclusion of a pseudoexon that introduces an early termination codon and is proposed to be the cause of NLRP3 inflammasome deficiency in NZB cells. Here we have used exon skipping antisense oligonucleotides (AONs) to prevent aberrant splicing of Nlrp3 in NZB macrophages, and this restored both NLRP3 protein expression and NLRP3 inflammasome activity. Thus, the single point mutation leading to aberrant splicing is the sole cause of NLRP3 inflammasome deficiency in NZB macrophages. The NZB mouse provides a model for addressing a splicing defect in macrophages and could be used to further investigate AON design and delivery of AONs to macrophages in vivo.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Exones/genética , Inflamasomas/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Oligonucleótidos Antisentido/farmacología , Empalme Alternativo/genética , Animales , Secuencia de Bases , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Endogámicos NZB , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
20.
EMBO Rep ; 15(9): 982-90, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24990442

RESUMEN

A current paradigm proposes that mitochondrial damage is a critical determinant of NLRP3 inflammasome activation. Here, we genetically assess whether mitochondrial signalling represents a unified mechanism to explain how NLRP3 is activated by divergent stimuli. Neither co-deletion of the essential executioners of mitochondrial apoptosis BAK and BAX, nor removal of the mitochondrial permeability transition pore component cyclophilin D, nor loss of the mitophagy regulator Parkin, nor deficiency in MAVS affects NLRP3 inflammasome function. In contrast, caspase-8, a caspase essential for death-receptor-mediated apoptosis, is required for efficient Toll-like-receptor-induced inflammasome priming and cytokine production. Collectively, these results demonstrate that mitochondrial apoptosis is not required for NLRP3 activation, and highlight an important non-apoptotic role for caspase-8 in regulating inflammasome activation and pro-inflammatory cytokine levels.


Asunto(s)
Proteínas Portadoras/biosíntesis , Caspasa 8/biosíntesis , Inflamasomas/metabolismo , Mitocondrias/metabolismo , Apoptosis/genética , Autofagia/genética , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Proteínas Portadoras/genética , Caspasa 8/genética , Células Cultivadas , Peptidil-Prolil Isomerasa F , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/genética , Humanos , Interleucina-1beta/biosíntesis , Mitocondrias/patología , Mitofagia/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores Toll-Like/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA