Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Annu Rev Immunol ; 41: 453-481, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-36750319

RESUMEN

The innate immune system detects pathogens via germline-encoded receptors that bind to conserved pathogen ligands called pathogen-associated molecular patterns (PAMPs). Here we consider an additional strategy of pathogen sensing called effector-triggered immunity (ETI). ETI involves detection of pathogen-encoded virulence factors, also called effectors. Pathogens produce effectors to manipulate hosts to create a replicative niche and/or block host immunity. Unlike PAMPs, effectors are often diverse and rapidly evolving and can thus be unsuitable targets for direct detection by germline-encoded receptors. Effectors are instead often sensed indirectly via detection of their virulence activities. ETI is a viable strategy for pathogen sensing and is used across diverse phyla, including plants, but the molecular mechanisms of ETI are complex compared to simple receptor/ligand-based PAMP detection. Here we survey the mechanisms and functions of ETI, with a particular focus on emerging insights from animal studies. We suggest that many examples of ETI may remain to be discovered, hiding in plain sight throughout immunology.


Asunto(s)
Reconocimiento de Inmunidad Innata , Moléculas de Patrón Molecular Asociado a Patógenos , Humanos , Animales , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Virulencia
2.
Cell ; 179(6): 1264-1275.e13, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31778653

RESUMEN

TLR8 is among the highest-expressed pattern-recognition receptors in the human myeloid compartment, yet its mode of action is poorly understood. TLR8 engages two distinct ligand binding sites to sense RNA degradation products, although it remains unclear how these ligands are formed in cellulo in the context of complex RNA molecule sensing. Here, we identified the lysosomal endoribonuclease RNase T2 as a non-redundant upstream component of TLR8-dependent RNA recognition. RNase T2 activity is required for rendering complex single-stranded, exogenous RNA molecules detectable for TLR8. This is due to RNase T2's preferential cleavage of single-stranded RNA molecules between purine and uridine residues, which critically contributes to the supply of catabolic uridine and the generation of purine-2',3'-cyclophosphate-terminated oligoribonucleotides. Thus-generated molecules constitute agonistic ligands for the first and second binding pocket of TLR8. Together, these results establish the identity and origin of the RNA-derived molecular pattern sensed by TLR8.


Asunto(s)
Endorribonucleasas/metabolismo , Proteolisis , Receptor Toll-Like 8/metabolismo , Secuencias de Aminoácidos , Secuencia de Bases , Línea Celular , Endorribonucleasas/deficiencia , Humanos , Modelos Moleculares , Monocitos/metabolismo , Células Mieloides/metabolismo , Isótopos de Nitrógeno , Oligonucleótidos/metabolismo , Purinas/metabolismo , ARN/metabolismo , Staphylococcus aureus/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/agonistas , Receptor Toll-Like 8/química , Uridina/metabolismo
3.
Cell ; 171(5): 1110-1124.e18, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29033128

RESUMEN

Detection of cytosolic DNA constitutes a central event in the context of numerous infectious and sterile inflammatory conditions. Recent studies have uncovered a bipartite mode of cytosolic DNA recognition, in which the cGAS-STING axis triggers antiviral immunity, whereas AIM2 triggers inflammasome activation. Here, we show that AIM2 is dispensable for DNA-mediated inflammasome activation in human myeloid cells. Instead, detection of cytosolic DNA by the cGAS-STING axis induces a cell death program initiating potassium efflux upstream of NLRP3. Forward genetics identified regulators of lysosomal trafficking to modulate this cell death program, and subsequent studies revealed that activated STING traffics to the lysosome, where it triggers membrane permeabilization and thus lysosomal cell death (LCD). Importantly, the cGAS-STING-NLRP3 pathway constitutes the default inflammasome response during viral and bacterial infections in human myeloid cells. We conclude that targeting the cGAS-STING-LCD-NLRP3 pathway will ameliorate pathology in inflammatory conditions that are associated with cytosolic DNA sensing.


Asunto(s)
Muerte Celular , Inflamasomas/metabolismo , Monocitos/citología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ADN/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Monocitos/metabolismo , Transducción de Señal
4.
Immunity ; 55(12): 2271-2284.e7, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36384135

RESUMEN

The NLRP3 inflammasome plays a central role in antimicrobial defense as well as in the context of sterile inflammatory conditions. NLRP3 activity is governed by two independent signals: the first signal primes NLRP3, rendering it responsive to the second signal, which then triggers inflammasome formation. Our understanding of how NLRP3 priming contributes to inflammasome activation remains limited. Here, we show that IKKß, a kinase activated during priming, induces recruitment of NLRP3 to phosphatidylinositol-4-phosphate (PI4P), a phospholipid enriched on the trans-Golgi network. NEK7, a mitotic spindle kinase that had previously been thought to be indispensable for NLRP3 activation, was redundant for inflammasome formation when IKKß recruited NLRP3 to PI4P. Studying iPSC-derived human macrophages revealed that the IKKß-mediated NEK7-independent pathway constitutes the predominant NLRP3 priming mechanism in human myeloid cells. Our results suggest that PI4P binding represents a primed state into which NLRP3 is brought by IKKß activity.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Quinasa I-kappa B , Inflamasomas/metabolismo , Ratones Endogámicos C57BL , Quinasas Relacionadas con NIMA/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Red trans-Golgi/metabolismo
5.
Nature ; 600(7887): 138-142, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34759314

RESUMEN

Pathogens use virulence factors to inhibit the immune system1. The guard hypothesis2,3 postulates that hosts monitor (or 'guard') critical innate immune pathways such that their disruption by virulence factors provokes a secondary immune response1. Here we describe a 'self-guarded' immune pathway in human monocytes, in which guarding and guarded functions are combined in one protein. We find that this pathway is triggered by ICP0, a key virulence factor of herpes simplex virus type 1, resulting in robust induction of anti-viral type I interferon (IFN). Notably, induction of IFN by ICP0 is independent of canonical immune pathways and the IRF3 and IRF7 transcription factors. A CRISPR screen identified the ICP0 target MORC34 as an essential negative regulator of IFN. Loss of MORC3 recapitulates the IRF3- and IRF7-independent IFN response induced by ICP0. Mechanistically, ICP0 degrades MORC3, which leads to de-repression of a MORC3-regulated DNA element (MRE) adjacent to the IFNB1 locus. The MRE is required in cis for IFNB1 induction by the MORC3 pathway, but is not required for canonical IFN-inducing pathways. As well as repressing the MRE to regulate IFNB1, MORC3 is also a direct restriction factor of HSV-15. Our results thus suggest a model in which the primary anti-viral function of MORC3 is self-guarded by its secondary IFN-repressing function-thus, a virus that degrades MORC3 to avoid its primary anti-viral function will unleash the secondary anti-viral IFN response.


Asunto(s)
Adenosina Trifosfatasas/inmunología , Proteínas de Unión al ADN/inmunología , Modelos Inmunológicos , Factores de Virulencia/inmunología , Adenosina Trifosfatasas/deficiencia , Adenosina Trifosfatasas/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Edición Génica , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Humanos , Proteínas Inmediatas-Precoces/inmunología , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Monocitos/inmunología , Receptor de Interferón alfa y beta , Proteínas Represoras/deficiencia , Proteínas Represoras/inmunología , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Ubiquitina-Proteína Ligasas/inmunología
6.
Immunity ; 44(4): 833-46, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27037191

RESUMEN

Interleukin-1ß (IL-1ß) is a cytokine whose bioactivity is controlled by activation of the inflammasome. However, in response to lipopolysaccharide, human monocytes secrete IL-1ß independently of classical inflammasome stimuli. Here, we report that this constituted a species-specific response that is not observed in the murine system. Indeed, in human monocytes, lipopolysaccharide triggered an "alternative inflammasome" that relied on NLRP3-ASC-caspase-1 signaling, yet was devoid of any classical inflammasome characteristics including pyroptosome formation, pyroptosis induction, and K(+) efflux dependency. Genetic dissection of the underlying signaling pathway in a monocyte transdifferentiation system revealed that alternative inflammasome activation was propagated by TLR4-TRIF-RIPK1-FADD-CASP8 signaling upstream of NLRP3. Importantly, involvement of this signaling cascade was limited to alternative inflammasome activation and did not extend to classical NLRP3 activation. Because alternative inflammasome activation embraces both sensitivity and promiscuity of TLR4, we propose a pivotal role for this signaling cascade in TLR4-driven, IL-1ß-mediated immune responses and immunopathology in humans.


Asunto(s)
Proteínas Portadoras/inmunología , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Monocitos/inmunología , Receptor Toll-Like 4/inmunología , Animales , Caspasa 1/inmunología , Línea Celular , Transdiferenciación Celular/inmunología , Humanos , Interleucina-1beta/metabolismo , Lipopolisacáridos , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Potasio/metabolismo , Canales de Potasio/inmunología , Piroptosis/inmunología , Transducción de Señal/inmunología
7.
Nature ; 549(7672): 394-398, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28902841

RESUMEN

Cytosolic DNA arising from intracellular pathogens triggers a powerful innate immune response. It is sensed by cyclic GMP-AMP synthase (cGAS), which elicits the production of type I interferons by generating the second messenger 2'3'-cyclic-GMP-AMP (cGAMP). Endogenous nuclear or mitochondrial DNA can also be sensed by cGAS under certain conditions, resulting in sterile inflammation. The cGAS dimer binds two DNA ligands shorter than 20 base pairs side-by-side, but 20-base-pair DNA fails to activate cGAS in vivo and is a poor activator in vitro. Here we show that cGAS is activated in a strongly DNA length-dependent manner both in vitro and in human cells. We also show that cGAS dimers form ladder-like networks with DNA, leading to cooperative sensing of DNA length: assembly of the pioneering cGAS dimer between two DNA molecules is ineffective; but, once formed, it prearranges the flanking DNA to promote binding of subsequent cGAS dimers. Remarkably, bacterial and mitochondrial nucleoid proteins HU and mitochondrial transcription factor A (TFAM), as well as high-mobility group box 1 protein (HMGB1), can strongly stimulate long DNA sensing by cGAS. U-turns and bends in DNA induced by these proteins pre-structure DNA to nucleate cGAS dimers. Our results suggest a nucleation-cooperativity-based mechanism for sensitive detection of mitochondrial DNA and pathogen genomes, and identify HMGB/TFAM proteins as DNA-structuring host factors. They provide an explanation for the peculiar cGAS dimer structure and suggest that cGAS preferentially binds incomplete nucleoid-like structures or bent DNA.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ADN/química , ADN/metabolismo , Proteínas HMGB/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas Mitocondriales/metabolismo , Nucleotidiltransferasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Femenino , Humanos , Ratones , Modelos Moleculares , Conformación de Ácido Nucleico , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/química , Multimerización de Proteína
8.
Proc Natl Acad Sci U S A ; 116(3): 970-975, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30591564

RESUMEN

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a critical regulator of cell death and inflammation, but its relevance for human disease pathogenesis remains elusive. Studies of monogenic disorders might provide critical insights into disease mechanisms and therapeutic targeting of RIPK1 for common diseases. Here, we report on eight patients from six unrelated pedigrees with biallelic loss-of-function mutations in RIPK1 presenting with primary immunodeficiency and/or intestinal inflammation. Mutations in RIPK1 were associated with reduced NF-κB activity, defective differentiation of T and B cells, increased inflammasome activity, and impaired response to TNFR1-mediated cell death in intestinal epithelial cells. The characterization of RIPK1-deficient patients highlights the essential role of RIPK1 in controlling human immune and intestinal homeostasis, and might have critical implications for therapies targeting RIPK1.


Asunto(s)
Diferenciación Celular , Inmunidad Mucosa/genética , Enfermedades Inflamatorias del Intestino , Mucosa Intestinal , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Inmunodeficiencia Combinada Grave , Linfocitos B/inmunología , Linfocitos B/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Epiteliales/inmunología , Células Epiteliales/patología , Femenino , Células HCT116 , Células HEK293 , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Masculino , Mutación , FN-kappa B/genética , FN-kappa B/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/patología , Linfocitos T/inmunología , Linfocitos T/patología
9.
J Biol Chem ; 295(52): 18065-18075, 2020 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-33082141

RESUMEN

TNF is a highly pro-inflammatory cytokine that contributes not only to the regulation of immune responses but also to the development of severe inflammatory diseases. TNF is synthesized as a transmembrane protein, which is further matured via proteolytic cleavage by metalloproteases such as ADAM17, a process known as shedding. At present, TNF is mainly detected by measuring the precursor or the mature cytokine of bulk cell populations by techniques such as ELISA or immunoblotting. However, these methods do not provide information on the exact timing and extent of TNF cleavage at single-cell resolution and they do not allow the live visualization of shedding events. Here, we generated C-tag TNF as a genetically encoded reporter to study TNF shedding at the single-cell level. The functionality of the C-tag TNF reporter is based on the exposure of a cryptic epitope on the C terminus of the transmembrane portion of pro-TNF on cleavage. In both denatured and nondenatured samples, this epitope can be detected by a nanobody in a highly sensitive and specific manner only upon TNF shedding. As such, C-tag TNF can successfully be used for the detection of TNF cleavage in flow cytometry and live-cell imaging applications. We furthermore demonstrate its applicability in a forward genetic screen geared toward the identification of genetic regulators of TNF maturation. In summary, the C-tag TNF reporter can be employed to gain novel insights into the complex regulation of ADAM-dependent TNF shedding.


Asunto(s)
Proteínas ADAM/metabolismo , Genes Reporteros , Procesamiento de Imagen Asistido por Computador/métodos , Imagen Molecular/métodos , Proteína Quinasa C/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas ADAM/genética , Células HEK293 , Humanos , Proteína Quinasa C/genética , Proteolisis , Factor de Necrosis Tumoral alfa/genética
10.
EMBO J ; 33(24): 2937-46, 2014 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-25425575

RESUMEN

Intracellular recognition of non-self and also self-nucleic acids can result in the initiation of potent pro-inflammatory and antiviral cytokine responses. Most recently, cGAS was shown to be critical for the recognition of cytoplasmic dsDNA. Binding of dsDNA to cGAS results in the synthesis of cGAMP(2'-5'), which then binds to the endoplasmic reticulum resident protein STING. This initiates a signaling cascade that triggers the induction of an antiviral immune response. While most studies on intracellular nucleic acids have focused on dsRNA or dsDNA, it has remained unexplored whether cytosolic RNA:DNA hybrids are also sensed by the innate immune system. Studying synthetic RNA:DNA hybrids, we indeed observed a strong type I interferon response upon cytosolic delivery of this class of molecule. Studies in THP-1 knockout cells revealed that the recognition of RNA:DNA hybrids is completely attributable to the cGAS-STING pathway. Moreover, in vitro studies showed that recombinant cGAS produced cGAMP upon RNA:DNA hybrid recognition. Altogether, our results introduce RNA:DNA hybrids as a novel class of intracellular PAMP molecules and describe an alternative cGAS ligand next to dsDNA.


Asunto(s)
ADN/genética , Proteínas de la Membrana/metabolismo , Hibridación de Ácido Nucleico , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/metabolismo , ARN/genética , Virus/inmunología , Mordeduras y Picaduras , Línea Celular , ADN/metabolismo , Humanos , Interferón Tipo I/metabolismo , Monocitos/inmunología , Unión Proteica , ARN/metabolismo
11.
Genome Res ; 24(10): 1719-23, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25186908

RESUMEN

The application of designer nucleases allows the induction of DNA double-strand breaks (DSBs) at user-defined genomic loci. Due to imperfect DNA repair mechanisms, DSBs can lead to alterations in the genomic architecture, such as the disruption of the reading frame of a critical exon. This can be exploited to generate somatic knockout cell lines. While high genome editing activities can be achieved in various cellular systems, obtaining cell clones that contain all-allelic frameshift mutations at the target locus of interest remains a laborious task. To this end, we have developed an easy-to-follow deep sequencing workflow and the evaluation tool OutKnocker (www.OutKnocker.org), which allows convenient, reliable, and cost-effective identification of knockout cell lines.


Asunto(s)
Endonucleasas/metabolismo , Técnicas de Genotipaje/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Línea Celular Tumoral , Mutación del Sistema de Lectura , Técnicas de Inactivación de Genes , Técnicas de Genotipaje/economía , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Humanos , Datos de Secuencia Molecular , Edición de ARN , Análisis de Secuencia de ADN/economía , Navegador Web
12.
Eur J Immunol ; 45(10): 2911-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26174085

RESUMEN

Inflammasome activation culminates in activation of caspase-1, which leads to the maturation and subsequent release of cytokines of the interleukin 1 (IL-1) family and results in a particular form of cell death known as pyroptosis. In addition, in the murine system, a so-called non-canonical inflammasome involving caspase-11 has been described that directly responds to cytosolic LPS. Here, we show that the human monocytic cell line THP1 activates the inflammasome in response to cytosolic LPS in a TLR4-independent fashion. This response is mediated by caspase-4 and accompanied by caspase-1 activation, pyroptosis, and IL-1ß maturation. In addition to caspase-4, efficient IL-1ß conversion upon intracellular LPS delivery relies on potassium efflux, NLRP3, ASC, and caspase-1, indicating that although caspase-4 activation alone is sufficient to induce pyroptosis, this process depends on the NLRP3 inflammasome activation to drive IL-1ß maturation. Altogether, this study provides evidence for the presence of a non-canonical inflammasome in humans and its dependence on caspase-4.


Asunto(s)
Proteínas Portadoras/inmunología , Caspasas Iniciadoras/inmunología , Inflamasomas/inmunología , Células Mieloides/inmunología , Proteínas Portadoras/genética , Caspasa 1/genética , Caspasa 1/inmunología , Caspasas Iniciadoras/genética , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Lipopolisacáridos/farmacología , Células Mieloides/citología , Proteína con Dominio Pirina 3 de la Familia NLR
13.
EMBO J ; 35(20): 2167-2169, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27572465
15.
Front Immunol ; 13: 1074440, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36578489

RESUMEN

Necroptosis is a form of regulated cell death that can occur downstream of several immune pathways. While previous studies have shown that dysregulated necroptosis can lead to strong inflammatory responses, little is known about the identity of the endogenous molecules that trigger these responses. Using a reductionist in vitro model, we found that soluble TNF is strongly released in the context of necroptosis. On the one hand, necroptosis promotes TNF translation by inhibiting negative regulatory mechanisms acting at the post-transcriptional level. On the other hand, necroptosis markedly enhances TNF release by activating ADAM proteases. In studying TNF release at single-cell resolution, we found that TNF release triggered by necroptosis is activated in a switch-like manner that exceeds steady-state TNF processing in magnitude and speed. Although this shedding response precedes massive membrane damage, it is closely associated with lytic cell death. Further, we found that lytic cell death induction using a pore-forming toxin also triggers TNF shedding, indicating that the activation of ADAM proteases is not strictly related to the necroptotic pathway but likely associated with biophysical changes of the cell membrane upon lytic cell death. These results demonstrate that lytic cell death, particularly necroptosis, is a critical trigger for TNF release and thus qualify TNF as a necroptosis-associated alarmin.


Asunto(s)
Alarminas , Apoptosis , Humanos , Necrosis , Necroptosis , Factor de Necrosis Tumoral alfa/metabolismo , Péptido Hidrolasas
16.
Elife ; 102021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34151776

RESUMEN

Type I interferons (IFNs) are essential for anti-viral immunity, but often impair protective immune responses during bacterial infections. An important question is how type I IFNs are strongly induced during viral infections, and yet are appropriately restrained during bacterial infections. The Super susceptibility to tuberculosis 1 (Sst1) locus in mice confers resistance to diverse bacterial infections. Here we provide evidence that Sp140 is a gene encoded within the Sst1 locus that represses type I IFN transcription during bacterial infections. We generated Sp140-/- mice and found that they are susceptible to infection by Legionella pneumophila and Mycobacterium tuberculosis. Susceptibility of Sp140-/- mice to bacterial infection was rescued by crosses to mice lacking the type I IFN receptor (Ifnar-/-). Our results implicate Sp140 as an important negative regulator of type I IFNs that is essential for resistance to bacterial infections.


Asunto(s)
Infecciones Bacterianas/inmunología , Interferón Tipo I/metabolismo , Factores de Transcripción/metabolismo , Alelos , Animales , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Interferón Tipo I/genética , Macrófagos/fisiología , Masculino , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Mycobacterium tuberculosis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Organismos Libres de Patógenos Específicos , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/farmacología
17.
J Mol Biol ; 430(2): 133-141, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29203171

RESUMEN

NLRP3 is the most studied inflammasome sensor due to its crucial involvement in sterile and infection-triggered inflammation. Although its molecular mode of activation remains to be defined, it is well established that low intracellular potassium concentrations result in its activation. This functionality allows the classical NLRP3 pathway to serve as a highly sensitive, but non-specific surveillance mechanism responding to any type of perturbation that breaches plasma membrane integrity and the associated potassium gradient across the membrane. Here, we review our current knowledge on potassium efflux-dependent NLRP3 activation, with a special focus on how major cell death programs are rendered pro-inflammatory by secondary NLRP3 activation. Apart from the "alternative inflammasome" as the major exception to the rule, this connection explains the fundamental importance of NLRP3 in cell death-associated inflammation and firmly establishes NLRP3 as a principal surveillance mechanism of cellular integrity.


Asunto(s)
Inflamasomas/inmunología , Inflamación/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Animales , Muerte Celular , Humanos , Potasio/inmunología
18.
Methods Mol Biol ; 1714: 57-66, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29177855

RESUMEN

Monocytes and macrophages play a pivotal role in the induction and shaping of immune responses. Expressing a broad array of pattern recognition receptors (PRRs), monocytes and macrophages constitute an integral component of the innate branch of the immune system. Traditionally, the majority of innate immune sensing and signaling pathways have been studied in macrophages of the murine system. This is largely due to the fact that genetic loss-of-function studies are amenable in this species. On the other hand, human cell lines of the monocyte-macrophage cell lineage have been widely used to study myeloid cells in vitro. However, commonly utilized models (e.g., THP-1 cells) only mimic a limited spectrum of the immunobiology of primary human myeloid cells. Recently, we have explored the possibility to fill this gap with a human trans-differentiation cell culture system, in which lineage conversion from malignant B-lineage cells to monocytes/macrophages is caused by the inducible nuclear translocation of a C/EBPα transgene, BLaER1 cells. Using this model, we were able to characterize a novel inflammasome signaling entity that could not have been uncovered in the murine system or THP-1 cells. Here, we describe the handling of BLaER1 cells, providing a detailed protocol for their induced trans-differentiation. We also provide data to demonstrate the applicability of the BLaER1 monocyte/macrophage system to study phagocytosis and various PRR cascades in human cells.


Asunto(s)
Diferenciación Celular , Macrófagos/citología , Modelos Biológicos , Monocitos/citología , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Línea Celular , Humanos , Inmunidad Innata , Macrófagos/metabolismo , Monocitos/metabolismo , Células Mieloides/citología , Células Mieloides/metabolismo , Transducción de Señal
19.
Cell Rep ; 25(9): 2354-2368.e5, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30485805

RESUMEN

IL-1ß is a cytokine of pivotal importance to the orchestration of inflammatory responses. Synthesized as an inactive pro-cytokine, IL-1ß requires proteolytic maturation to gain biological activity. Here, we identify intrinsic apoptosis as a non-canonical trigger of IL-1ß maturation. Guided by the discovery of the immunomodulatory activity of vioprolides, cyclic peptides isolated from myxobacteria, we observe IL-1ß maturation independent of canonical inflammasome pathways, yet dependent on intrinsic apoptosis. Mechanistically, vioprolides inhibit MCL-1 and BCL2, which in turn triggers BAX/BAK-dependent mitochondrial outer membrane permeabilization (MOMP). Induction of MOMP results in the release of pro-apoptotic factors initiating intrinsic apoptosis, as well as the depletion of IAPs (inhibitors of apoptosis proteins). IAP depletion, in turn, operates upstream of ripoptosome complex formation, subsequently resulting in caspase-8-dependent IL-1ß maturation. These results establish the ripoptosome/caspase-8 complex as a pro-inflammatory checkpoint that senses the perturbation of mitochondrial integrity.


Asunto(s)
Apoptosis , Caspasa 8/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Caspasa 1/metabolismo , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , L-Lactato Deshidrogenasa/metabolismo , Ratones , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Péptidos Cíclicos/farmacología , Permeabilidad , Inhibidores de la Síntesis de la Proteína/farmacología
20.
Curr Opin Immunol ; 44: 7-13, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27842238

RESUMEN

Classical modes of NLRP3 activation entail a priming step that enables its activation (signal 1) and a potassium efflux-dependent activation signal (signal 2) that triggers pyroptosome formation and pyroptosis, a lytic cell death necessary for IL-1ß release. Opposing to that, human monocytes engage an alternative NLRP3 inflammasome pathway in response to LPS that proceeds in the absence of signal 2 activation and enables IL-1ß secretion without pyroptosis. This specifically relies on Caspase-8 to propagate signaling to NLRP3, leading to inflammasome activation in absence of pyroptosome formation. Here, we summarize the current knowledge about alternative inflammasome activation, discuss potential extensions of this signaling entity beyond LPS-dependent activation, speculate about its role in tissue homeostasis and sterile inflammation and highlight the implications of pyroptosis-independent IL-1ß secretion.


Asunto(s)
Inflamasomas/metabolismo , Inflamación/inmunología , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Caspasa 8/metabolismo , Homeostasis , Humanos , Lipopolisacáridos/inmunología , Antiportadores de Potasio-Hidrógeno/metabolismo , Piroptosis , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA