Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Immunol ; 197(6): 2421-33, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27521339

RESUMEN

A critical component of innate immune response to infection and tissue damage is the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome, and this pathway and its activation products have been implicated in the pathophysiology of a variety of diseases. NLRP3 inflammasome activation leads to the cleavage of pro-IL-1ß and pro-IL-18, as well as the subsequent release of biologically active IL-1ß, IL-18, and other soluble mediators of inflammation. In this study, we further define the pharmacology of the previously reported NLRP3 inflammasome-selective, IL-1ß processing inhibitor CP-456,773 (also known as MCC950), and we demonstrate its efficacy in two in vivo models of inflammation. Specifically, we show that in human and mouse innate immune cells CP-456,773 is an inhibitor of the cellular release of IL-1ß, IL-1α, and IL-18, that CP-456,773 prevents inflammasome activation induced by disease-relevant soluble and crystalline NLRP3 stimuli, and that CP-456,773 inhibits R848- and imiquimod-induced IL-1ß release. In mice, CP-456,773 demonstrates potent inhibition of the release of proinflammatory cytokines following acute i.p. challenge with LPS plus ATP in a manner that is proportional to the free/unbound concentrations of the drug, thereby establishing an in vivo pharmacokinetic/pharmacodynamic model for CP-456,773. Furthermore, CP-456,773 reduces ear swelling in an imiquimod cream-induced mouse model of skin inflammation, and it reduces airway inflammation in mice following acute challenge with house dust mite extract. These data implicate the NLRP3 inflammasome in the pathogenesis of dermal and airway inflammation, and they highlight the utility of CP-456,773 for interrogating the contribution of the NLRP3 inflammasome and its outputs in preclinical models of inflammation and disease.


Asunto(s)
Dermatitis/tratamiento farmacológico , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Inflamasomas/antagonistas & inhibidores , Inflamación/fisiopatología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Neumonía/tratamiento farmacológico , Neumonía/inmunología , Sulfonas/farmacología , Animales , Citocinas/antagonistas & inhibidores , Citocinas/inmunología , Dermatitis/inmunología , Dermatitis/fisiopatología , Modelos Animales de Enfermedad , Furanos , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Inmunidad Innata/efectos de los fármacos , Indenos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Interleucina-18/antagonistas & inhibidores , Interleucina-18/metabolismo , Interleucina-1alfa/antagonistas & inhibidores , Interleucina-1alfa/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/inmunología , Ratones , Neumonía/fisiopatología , Transducción de Señal , Sulfonamidas , Sulfonas/administración & dosificación , Sulfonas/uso terapéutico
2.
J Immunol ; 196(4): 1732-40, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26773149

RESUMEN

Neisseria gonorrhoeae, the causative agent of the sexually transmitted infection gonorrhea, has developed resistance to almost every conventional antibiotic. There is an urgent need to develop novel therapies against gonorrhea. Many pathogens, including N. gonorrhoeae, bind the complement inhibitor factor H (FH) to evade complement-dependent killing. Sialylation of gonococcal lipooligosaccharide, as occurs in vivo, augments binding of human FH through its domains 18-20 (FH18-20). We explored the use of fusing FH18-20 with IgG Fc (FH18-20/Fc) to create a novel anti-infective immunotherapeutic. FH18-20 also binds to select host glycosaminoglycans to limit unwanted complement activation on host cells. To identify mutation(s) in FH18-20 that eliminated complement activation on host cells, yet maintained binding to N. gonorrhoeae, we created four mutations in domains 19 or 20 described in atypical hemolytic uremic syndrome that prevented binding of mutated fH to human erythrocytes. One of the mutant proteins (D to G at position 1119 in domain 19; FHD1119G/Fc) facilitated complement-dependent killing of gonococci similar to unmodified FH18-20/Fc but, unlike FH18-20/Fc, did not lyse human erythrocytes. FHD1119G/Fc bound to all (100%) of 15 sialylated clinical N. gonorrhoeae isolates tested (including three contemporary ceftriaxone-resistant strains), mediated complement-dependent killing of 10 of 15 (67%) strains, and enhanced C3 deposition (≥10-fold above baseline levels) on each of the five isolates not directly killed by complement. FHD1119G/Fc facilitated opsonophagocytic killing of a serum-resistant strain by human polymorphonuclear neutrophils. FHD1119G/Fc administered intravaginally significantly reduced the duration and burden of gonococcal infection in the mouse vaginal colonization model. FHD1119G/Fc represents a novel immunotherapeutic against multidrug-resistant N. gonorrhoeae.


Asunto(s)
Factor H de Complemento/inmunología , Gonorrea/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoterapia/métodos , Proteínas Recombinantes de Fusión/inmunología , Animales , Factor H de Complemento/farmacología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Humanos , Fragmentos Fc de Inmunoglobulinas/farmacología , Ratones , Ratones Endogámicos BALB C , Neisseria gonorrhoeae/inmunología , Proteínas Recombinantes de Fusión/farmacología
3.
Nat Med ; 21(3): 248-55, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25686105

RESUMEN

The NOD-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3) inflammasome is a component of the inflammatory process, and its aberrant activation is pathogenic in inherited disorders such as cryopyrin-associated periodic syndrome (CAPS) and complex diseases such as multiple sclerosis, type 2 diabetes, Alzheimer's disease and atherosclerosis. We describe the development of MCC950, a potent, selective, small-molecule inhibitor of NLRP3. MCC950 blocked canonical and noncanonical NLRP3 activation at nanomolar concentrations. MCC950 specifically inhibited activation of NLRP3 but not the AIM2, NLRC4 or NLRP1 inflammasomes. MCC950 reduced interleukin-1ß (IL-1ß) production in vivo and attenuated the severity of experimental autoimmune encephalomyelitis (EAE), a disease model of multiple sclerosis. Furthermore, MCC950 treatment rescued neonatal lethality in a mouse model of CAPS and was active in ex vivo samples from individuals with Muckle-Wells syndrome. MCC950 is thus a potential therapeutic for NLRP3-associated syndromes, including autoinflammatory and autoimmune diseases, and a tool for further study of the NLRP3 inflammasome in human health and disease.


Asunto(s)
Proteínas Portadoras/antagonistas & inhibidores , Síndromes Periódicos Asociados a Criopirina/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Inflamasomas/antagonistas & inhibidores , Interleucina-1beta/efectos de los fármacos , Esclerosis Múltiple , Sulfonas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Furanos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Indenos , Inflamación , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Sulfonamidas , Sulfonas/farmacología
4.
Nat Immunol ; 15(8): 727-37, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24952505

RESUMEN

Microbes or danger signals trigger inflammasome sensors, which induce polymerization of the adaptor ASC and the assembly of ASC specks. ASC specks recruit and activate caspase-1, which induces maturation of the cytokine interleukin 1ß (IL-1ß) and pyroptotic cell death. Here we found that after pyroptosis, ASC specks accumulated in the extracellular space, where they promoted further maturation of IL-1ß. In addition, phagocytosis of ASC specks by macrophages induced lysosomal damage and nucleation of soluble ASC, as well as activation of IL-1ß in recipient cells. ASC specks appeared in bodily fluids from inflamed tissues, and autoantibodies to ASC specks developed in patients and mice with autoimmune pathologies. Together these findings reveal extracellular functions of ASC specks and a previously unknown form of cell-to-cell communication.


Asunto(s)
Apoptosis/inmunología , Caspasa 1/inmunología , Proteínas del Citoesqueleto/inmunología , Inflamación/inmunología , Interleucina-1beta/inmunología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Anticuerpos/inmunología , Proteínas Reguladoras de la Apoptosis , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Caspasa 1/genética , Inhibidores de Caspasas/farmacología , Comunicación Celular/inmunología , Proteínas del Citoesqueleto/genética , Humanos , Inflamasomas/inmunología , Lisosomas/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Fagocitosis/inmunología , Priones/química , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Transducción de Señal/inmunología
5.
Methods Mol Biol ; 1040: 91-101, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23852599

RESUMEN

All inflammasomes require the adapter protein apoptosis associated speck-like protein containing a CARD (ASC) for the activation of caspase-1. After inflammasome activation, ASC assembles into a large protein complex, which is termed "speck". ASC specks can be observed as they reach a size of around 1 µm and in most cells only one speck forms upon inflammasome activation. Hence, ASC speck formation can be used as a simple upstream readout for inflammasome activation. Here, we describe a method for analyzing inflammasome activation by ASC speck visualization. First, we describe the generation of a clonal inflammasome reporter macrophage cell line overexpressing fluorescently tagged ASC. We then discuss stimulation conditions and the microscopic evaluation of ASC speck formation.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Inflamasomas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Línea Celular , Proteínas del Citoesqueleto/genética , Expresión Génica , Vectores Genéticos/genética , Humanos , Inflamasomas/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Microscopía Confocal , Retroviridae/genética , Transducción Genética
6.
Nat Chem Biol ; 9(6): 398-405, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23603659

RESUMEN

Formation of the inflammasome, a scaffolding complex that activates caspase-1, is important in numerous diseases. Pyroptotic cell death induced by anthrax lethal toxin (LT) is a model for inflammasome-mediated caspase-1 activation. We discovered 7-desacetoxy-6,7-dehydrogedunin (7DG) in a phenotypic screen as a small molecule that protects macrophages from LT-induced death. Using chemical proteomics, we identified protein kinase R (PKR) as the target of 7DG and show that RNAi knockdown of PKR phenocopies treatment with 7DG. Further, we show that PKR's role in ASC assembly and caspase-1 activation induced by several different inflammasome stimuli is independent of PKR's kinase activity, demonstrating that PKR has a previously uncharacterized role in caspase-1 activation and pyroptosis that is distinct from its reported kinase-dependent roles in apoptosis and inflammasome formation in lipopolysaccharide-primed cells. Remarkably, PKR has different roles in two distinct cell death pathways and has a broad role in inflammasome function relevant in other diseases.


Asunto(s)
Muerte Celular , eIF-2 Quinasa/química , Animales , Bacillus anthracis/enzimología , Caspasa 1/metabolismo , Dominio Catalítico , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Proteínas HSP90 de Choque Térmico/metabolismo , Concentración de Iones de Hidrógeno , Inflamación , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Péptido Hidrolasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Conformación Proteica
7.
J Biol Chem ; 286(13): 11875-82, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21325272

RESUMEN

Myeloid differentiation protein 88 (MyD88) is a key signaling adapter in Toll-like receptor (TLR) signaling. MyD88 is also one of the most polymorphic adapter proteins. We screened the reported nonsynonymous coding mutations in MyD88 to identify variants with altered function. In reporter assays, a death domain variant, S34Y, was found to be inactive. Importantly, in reconstituted macrophage-like cell lines derived from knock-out mice, MyD88 S34Y was severely compromised in its ability to respond to all MyD88-dependent TLR ligands. Unlike wild-type MyD88, S34Y is unable to form distinct foci in the cells but is present diffused in the cytoplasm. We observed that IRAK4 co-localizes with MyD88 in these aggregates, and thus these foci appear to be "Myddosomes." The MyD88 S34Y loss-of-function mutant demonstrates how proper cellular localization of MyD88 to the Myddosome is a feature required for MyD88 function.


Asunto(s)
Sustitución de Aminoácidos , Citoplasma/metabolismo , Mutación Missense , Factor 88 de Diferenciación Mieloide/metabolismo , Animales , Citoplasma/genética , Células HEK293 , Humanos , Ratones , Factor 88 de Diferenciación Mieloide/genética , Transporte de Proteínas/fisiología , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
8.
J Biol Chem ; 285(36): 27935-43, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20592019

RESUMEN

Lipopolysaccharide (LPS) activates the innate immune response through the Toll-like receptor 4 (TLR4).MD-2 complex. A synthetic lipid A precursor, lipid IV(A), induces an innate immune response in mice but not in humans. Both TLR4 and MD-2 are required for the agonist activity of lipid IV(A) in mice, with TLR4 interacting through specific surface charges at the dimerization interface. In this study, we used site-directed mutagenesis to identify the MD-2 residues that determine lipid IV(A) species specificity. A single mutation of murine MD-2 at the hydrophobic pocket entrance, E122K, substantially reduced the response to lipid IV(A). Combining the murine MD-2 E122K with the murine TLR4 K367E/S386K/R434Q mutations completely abolished the response to lipid IV(A), effectively converting the murine cellular response to a human-like response. In human cells, however, simultaneous mutations of K122E, K125L, Y41F, and R69G on human MD-2 were required to promote a response to lipid IV(A). Combining the human MD-2 quadruple mutations with the human TLR4 E369K/Q436R mutations completely converted the human MD-2/human TLR4 receptor to a murine-like receptor. Because MD-2 residues 122 and 125 reside at the dimerization interface near the pocket entrance, surface charge differences here directly affect receptor dimerization. In comparison, residues 42 and 69 reside at the MD-2/TLR4 interaction surface opposite the dimerization interface. Surface charge differences there likely affect the binding angle and/or rigidity between MD-2 and TLR4, exerting an indirect influence on receptor dimerization and activation. Thus, surface charge differences at the two MD-2/TLR4 interfaces determine the species-specific activation of lipid IV(A).


Asunto(s)
Glucolípidos/metabolismo , Lípido A/análogos & derivados , Antígeno 96 de los Linfocitos/química , Antígeno 96 de los Linfocitos/metabolismo , Animales , Arginina , Ácido Aspártico , Línea Celular , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Leucina , Lípido A/metabolismo , Antígeno 96 de los Linfocitos/genética , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Fenotipo , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Especificidad de la Especie , Electricidad Estática , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Tirosina
9.
Nat Immunol ; 11(5): 395-402, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20351692

RESUMEN

Inflammasomes regulate the activity of caspase-1 and the maturation of interleukin 1beta (IL-1beta) and IL-18. AIM2 has been shown to bind DNA and engage the caspase-1-activating adaptor protein ASC to form a caspase-1-activating inflammasome. Using Aim2-deficient mice, we identify a central role for AIM2 in regulating caspase-1-dependent maturation of IL-1beta and IL-18, as well as pyroptosis, in response to synthetic double-stranded DNA. AIM2 was essential for inflammasome activation in response to Francisella tularensis, vaccinia virus and mouse cytomegalovirus and had a partial role in the sensing of Listeria monocytogenes. Moreover, production of IL-18 and natural killer cell-dependent production of interferon-gamma, events critical in the early control of virus replication, were dependent on AIM2 during mouse cytomegalovirus infection in vivo. Collectively, our observations demonstrate the importance of AIM2 in the sensing of both bacterial and viral pathogens and in triggering innate immunity.


Asunto(s)
Infecciones por Virus ADN/inmunología , Virus ADN/inmunología , Francisella tularensis/inmunología , Células Asesinas Naturales/metabolismo , Listeriosis/inmunología , Macrófagos/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Tularemia/inmunología , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Caspasa 1/genética , Caspasa 1/inmunología , Caspasa 1/metabolismo , Línea Celular , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Proteínas del Citoesqueleto/genética , ADN/inmunología , Infecciones por Virus ADN/genética , Infecciones por Virus ADN/metabolismo , Virus ADN/crecimiento & desarrollo , Virus ADN/patogenicidad , Proteínas de Unión al ADN , Francisella tularensis/patogenicidad , Humanos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/microbiología , Células Asesinas Naturales/patología , Células Asesinas Naturales/virología , Listeriosis/genética , Listeriosis/metabolismo , Activación de Linfocitos/genética , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo , Tularemia/genética , Tularemia/metabolismo , Carga Viral/genética , Carga Viral/inmunología
10.
J Immunol ; 184(3): 1168-79, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20042593

RESUMEN

Human metapneumoviruses (HMPVs) are recently identified Paramyxoviridae that contribute to respiratory tract infections in children. No effective treatments or vaccines are available. Successful defense against virus infection relies on early detection by germ line-encoded pattern recognition receptors and activation of cytokine and type I IFN genes. Recently, the RNA helicase retinoic acid-inducible gene I (RIG-I) has been shown to sense HMPV. In this study, we investigated the abilities of two prototype strains of HMPV (A1 [NL\1\00] and B1 [NL\1\99]) to activate RIG-I and induce type I IFNs. Despite the abilities of both HMPV-A1 and HMPV-B1 to infect and replicate in cell lines and primary cells, only the HMPV-A1 strain triggered RIG-I to induce IFNA/B gene transcription. The failure of the HMPV-B1 strain to elicit type I IFN production was dependent on the B1 phosphoprotein, which specifically prevented RIG-I-mediated sensing of HMPV viral 5' triphosphate RNA. In contrast to most cell types, plasmacytoid dendritic cells displayed a unique ability to sense both HMPV-A1 and HMPV-B1 and in this case sensing was via TLR7 rather than RIG-I. Collectively, these data reveal differential mechanisms of sensing for two closely related viruses, which operate in cell type-specific manners.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Metapneumovirus/inmunología , Fosfoproteínas/metabolismo , Receptor Toll-Like 7/metabolismo , Interferencia Viral/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/fisiología , Regulación Viral de la Expresión Génica/inmunología , Humanos , Inmunidad Innata , Interferón-alfa/biosíntesis , Interferón-alfa/genética , Interferón beta/biosíntesis , Interferón beta/genética , Ligandos , Metapneumovirus/genética , Metapneumovirus/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/metabolismo , Infecciones por Paramyxoviridae/virología , Fosfoproteínas/genética , ARN Viral/genética , Receptores Inmunológicos , Especificidad de la Especie , Receptor Toll-Like 7/deficiencia , Receptor Toll-Like 7/fisiología , Células Vero
11.
J Immunol ; 183(2): 787-91, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19570822

RESUMEN

The IL-1 family cytokines are regulated on transcriptional and posttranscriptional levels. Pattern recognition and cytokine receptors control pro-IL-1beta transcription whereas inflammasomes regulate the proteolytic processing of pro-IL-1beta. The NLRP3 inflammasome, however, assembles in response to extracellular ATP, pore-forming toxins, or crystals only in the presence of proinflammatory stimuli. How the activation of gene transcription by signaling receptors enables NLRP3 activation remains elusive and controversial. In this study, we show that cell priming through multiple signaling receptors induces NLRP3 expression, which we identified to be a critical checkpoint for NLRP3 activation. Signals provided by NF-kappaB activators are necessary but not sufficient for NLRP3 activation, and a second stimulus such as ATP or crystal-induced damage is required for NLRP3 activation.


Asunto(s)
Proteínas Portadoras/genética , Regulación de la Expresión Génica/inmunología , Inflamación/metabolismo , FN-kappa B/fisiología , Receptores de Citocinas/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Presentación de Antígeno , Proteínas Portadoras/metabolismo , Células Cultivadas , Humanos , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Noqueados , Monocitos/citología , Monocitos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Activación Transcripcional
12.
J Biol Chem ; 284(38): 25742-8, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19509286

RESUMEN

The adapter protein MyD88 adapter-like (Mal), encoded by TIR-domain containing adapter protein (Tirap) (MIM 606252), is the most polymorphic of the five adapter proteins involved in Toll-like receptor signaling, harboring eight non-synonymous single nucleotide polymorphisms in its coding region. We screened reported mutations of Mal for activity in reporter assays to test the hypothesis that variants of Mal existed with altered signaling potential. A TIR domain variant, Mal D96N (rs8177400), was found to be inactive. In reconstituted cell lines, Mal D96N acted as a hypomorphic mutation, with impaired cytokine production and NF-kappaB activation upon lipopolysaccharide or PAM2CSK4 stimulation. Moreover, co-immunoprecipitation studies revealed that Mal D96N is unable to interact with MyD88, a prerequisite for downstream signaling to occur. Computer modeling data suggested that residue 96 resides in the MyD88 binding site, further supporting these findings. Genotyping of Mal D96N in three different cohorts suggested that it is a rare mutation. We, thus, describe a rare variant in Mal that exerts its effect via its inability to bind MyD88.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Sustitución de Aminoácidos , Sitios de Unión/fisiología , Línea Celular , Estudios de Cohortes , Simulación por Computador , Femenino , Humanos , Lipopéptidos/farmacología , Lipopolisacáridos/farmacología , Masculino , Glicoproteínas de Membrana/genética , Modelos Moleculares , Mutación Missense , Factor 88 de Diferenciación Mieloide/genética , Polimorfismo de Nucleótido Simple , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Receptores de Interleucina-1/genética , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética
13.
Nat Immunol ; 9(8): 857-65, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18604209

RESUMEN

The fibrillar peptide amyloid-beta (A beta) has a chief function in the pathogenesis of Alzheimer's disease. Interleukin 1 beta (IL-1 beta) is a key cytokine in the inflammatory response to A beta. Insoluble materials such as crystals activate the inflammasome formed by the cytoplasmic receptor NALP3, which results in the release of IL-1 beta. Here we identify the NALP3 inflammasome as a sensor of A beta in a process involving the phagocytosis of A beta and subsequent lysosomal damage and release of cathepsin B. Furthermore, the IL-1 beta pathway was essential for the microglial synthesis of proinflammatory and neurotoxic factors, and the inflammasome, caspase-1 and IL-1 beta were critical for the recruitment of microglia to exogenous A beta in the brain. Our findings suggest that activation of the NALP3 inflammasome is important for inflammation and tissue damage in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Inmunidad Innata/inmunología , Inflamación/metabolismo , Proteínas Portadoras/metabolismo , Inflamación/genética , Inflamación/inmunología , Mediadores de Inflamación/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR
14.
Nat Immunol ; 8(7): 772-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17572678

RESUMEN

Microbial and synthetic DNA rich in CpG dinucleotides stimulates Toll-like receptor 9 (TLR9), whereas DNA lacking CpG either is inert or can inhibit TLR9 activation. The molecular mechanisms by which TLR9 becomes activated or is inhibited are not well understood. Here we show that TLR9 bound to stimulatory and inhibitory DNA; however, only stimulatory DNA led to substantial conformational changes in the TLR9 ectodomain. In the steady state, 'inactive' TLR9 homodimers formed in an inactivated conformation. Binding of DNA containing CpG, but not of DNA lacking CpG, to TLR9 dimers resulted in allosteric changes in the TLR9 cytoplasmic signaling domains. In endosomes, conformational changes induced by DNA containing CpG resulted in close apposition of the cytoplasmic signaling domains, a change that is probably required for the recruitment of signaling adaptor molecules. Our results indicate that the formation of TLR9 dimers is not sufficient for its activation but instead that TLR9 activation is regulated by conformational changes induced by DNA containing CpG.


Asunto(s)
Receptor Toll-Like 9/química , Receptor Toll-Like 9/metabolismo , Regulación Alostérica , Línea Celular , Islas de CpG/inmunología , Humanos , Ligandos , Oligodesoxirribonucleótidos/metabolismo , Unión Proteica , Conformación Proteica
15.
Proc Natl Acad Sci U S A ; 104(6): 1919-24, 2007 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-17261807

RESUMEN

Hemozoin (HZ) is an insoluble crystal formed in the food vacuole of malaria parasites. HZ has been reported to induce inflammation by directly engaging Toll-like receptor (TLR) 9, an endosomal receptor. "Synthetic" HZ (beta-hematin), typically generated from partially purified extracts of bovine hemin, is structurally identical to natural HZ. When HPLC-purified hemin was used to synthesize the crystal, beta-hematin had no inflammatory activity. In contrast, natural HZ from Plasmodium falciparum cultures was a potent TLR9 inducer. Natural HZ bound recombinant TLR9 ectodomain, but not TLR2. Both TLR9 stimulation and TLR9 binding of HZ were abolished by nuclease treatment. PCR analysis demonstrated that natural HZ is coated with malarial but not human DNA. Purified malarial DNA activated TLR9 but only when DNA was targeted directly to the endosome with a transfection reagent. Stimulatory quantities of natural HZ contain <1 microg of malarial DNA; its potency in activating immune responses was even greater than transfecting malarial DNA. Thus, although the malarial genome is extremely AT-rich, its DNA is highly proinflammatory, with the potential to induce cytokinemia and fever during disease. However, its activity depends on being bound to HZ, which we propose amplifies the biological responses to malaria DNA by targeting it to a TLR9(+) intracellular compartment.


Asunto(s)
Presentación de Antígeno , ADN Protozoario/metabolismo , Hemoproteínas/fisiología , Inmunidad Innata , Plasmodium falciparum/genética , Receptor Toll-Like 9/metabolismo , Animales , ADN Protozoario/inmunología , Humanos , Activación de Linfocitos/inmunología , Melanoma Experimental , Ratones , Plasmodium falciparum/inmunología , Receptor Toll-Like 9/inmunología
16.
J Leukoc Biol ; 80(6): 1584-92, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16946018

RESUMEN

The cell surface receptor complex formed by TLR4 and myeloid differentiation 2 (MD-2) is engaged when cells are exposed to LPS. Recent studies suggested that surface localization of functional mouse TLR4 (mTLR4) depends on the simultaneous expression of MD-2. As we did not observe a similar requirement, we conducted a comparative study of human TLR4 and mTLR4 surface expression in immune cells derived from the MD-2 knockout mouse and LPS-responsive cell lines and in cells that ectopically express TLR4. Our results indicate that in the human and mouse models, neither TLR4 function nor TLR4 surface targeting requires MD-2 coexpression. Accordingly, we report on one human cell line, which constitutively expresses functional TLR4 on the cell surface in the absence of MD-2 expression.


Asunto(s)
Regulación de la Expresión Génica , Antígeno 96 de los Linfocitos/biosíntesis , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Humanos , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/deficiencia , Antígeno 96 de los Linfocitos/inmunología , Ratones , Ratones Noqueados , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/inmunología
17.
Immunobiology ; 211(6-8): 437-47, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16920483

RESUMEN

Toll-like receptors (TLRs) are a small family of type-I glycoproteins that bind to and are activated by conserved non-self molecular signatures carried by microorganisms. Toll-like receptor 4 is triggered by most lipopolysaccharides (LPS). LPS is a complex amphipathic saccharolipidic glycan derived from Gram-negative bacteria. Unique among TLRs, TLR4 activity and interaction with its natural ligand(s) strictly depends on the presence of the extracellular adaptor MD-2. MD-2 is a small secreted glycoprotein that binds with cytokine-like affinities to both the hydrophobic portion of LPS and to the extracellular domain of TLR4. The interaction between MD-2 and LPS induces a triggering event on TLR4, which involves the molecular rearrangement of the receptor complex and its homotypic aggregation. In silico analysis suggests that MD-2 and MD-1 are paralogs derived from a common predecessor at the level of early vertebrates. In this review, we summarize the current state of knowledge concerning MD-2.


Asunto(s)
Antígeno 96 de los Linfocitos/fisiología , Secuencia de Aminoácidos , Animales , Bacterias/inmunología , Humanos , Inmunidad Innata/fisiología , Inflamación/inmunología , Lipopolisacáridos/inmunología , Antígeno 96 de los Linfocitos/genética , Datos de Secuencia Molecular , Receptor Toll-Like 4/fisiología
18.
Proc Natl Acad Sci U S A ; 103(16): 6299-304, 2006 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-16603631

RESUMEN

TRIF-related adaptor molecule (TRAM) is the fourth Toll/IL-1 resistance domain-containing adaptor to be described that participates in Toll-like receptor (TLR) signaling. TRAM functions exclusively in the TLR4 pathway. Here we show by confocal microscopy that TRAM is localized in the plasma membrane and the Golgi apparatus, where it colocalizes with TLR4. Membrane localization of TRAM is the result of myristoylation because mutation of a predicted myristoylation site in TRAM (TRAM-G2A) brought about dissociation of TRAM from the membrane and its relocation to the cytosol. Further, TRAM, but not TRAM-G2A, was radiolabeled with [3H]myristate in vivo. Unlike wild-type TRAM, overexpression of TRAM-G2A failed to elicit either IFN regulatory factor 3 or NF-kappaB signaling. Moreover, TRAM-G2A was unable to reconstitute LPS responses in bone marrow-derived macrophages from TRAM-deficient mice. These observations provide clear evidence that the myristoylation of TRAM targets it to the plasma membrane, where it is essential for LPS responses through the TLR4 signal transduction pathway, and suggest a hitherto unappreciated manner in which LPS responses can be regulated.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Macrófagos/inmunología , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/análisis , Proteínas Adaptadoras del Transporte Vesicular/genética , Secuencia de Aminoácidos , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Membrana Celular/química , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Mutación , Ácido Mirístico/metabolismo , Transducción de Señal , Receptor Toll-Like 4/análisis
19.
J Immunol ; 175(10): 6465-72, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16272300

RESUMEN

The detection of Gram-negative LPS depends upon the proper function of the TLR4-MD-2 receptor complex in immune cells. TLR4 is the signal transduction component of the LPS receptor, whereas MD-2 is the endotoxin-binding unit. MD-2 appears to activate TLR4 when bound to TLR4 and ligated by LPS. Only the monomeric form of MD-2 was found to bind LPS and only monomeric MD-2 interacts with TLR4. Monomeric MD-2 binds TLR4 with an apparent Kd of 12 nM; this binding avidity was unaltered in the presence of endotoxin. E5564, an LPS antagonist, appears to inhibit cellular activation by competitively preventing the binding of LPS to MD-2. Depletion of endogenous soluble MD-2 from human serum, with an immobilized TLR4 fusion protein, abrogated TLR4-mediated LPS responses. By determining the concentration of added-back MD-2 that restored normal LPS responsiveness, the concentration of MD-2 was estimated to be approximately 50 nM. Similarly, purified TLR4-Fc fusion protein, when added to the supernatants of TLR4-expressing cells in culture, inhibited the interaction of MD-2 with TLR4, thus preventing LPS stimulation. The ability to inhibit the effects of LPS as a result of the binding of TLR4-Fc or E5564 to MD-2 highlights MD-2 as the logical target for drug therapies designed to pharmacologically intervene against endotoxin-induced disease.


Asunto(s)
Lipopolisacáridos/toxicidad , Antígeno 96 de los Linfocitos/metabolismo , Receptor Toll-Like 4/metabolismo , Línea Celular , Humanos , Cinética , Lípido A/análogos & derivados , Lípido A/farmacología , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/metabolismo , Antígeno 96 de los Linfocitos/sangre , Antígeno 96 de los Linfocitos/química , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal , Solubilidad , Receptor Toll-Like 4/química
20.
J Immunol ; 175(8): 5260-8, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16210631

RESUMEN

The paramyxovirus Sendai (SV), is a well-established inducer of IFN-alphabeta gene expression. In this study we show that SV induces IFN-alphabeta gene expression normally in cells from mice with targeted deletions of the Toll-IL-1 resistance domain containing adapters MyD88, Mal, Toll/IL-1R domain-containing adaptor inducing IFN-beta (TRIF), and TRIF-related adaptor molecule TLR3, or the E3 ubiquitin ligase, TNFR-associated factor 6. This TLR-independent induction of IFN-alphabeta after SV infection is replication dependent and mediated by the RNA helicase, retinoic acid-inducible gene-I (RIG-I) and not the related family member, melanoma differentiation-associated gene 5. Furthermore, we characterize a RIG-I-like RNA helicase, Lgp2. In contrast to RIG-I or melanoma differentiation-associated gene 5, Lgp2 lacks signaling caspase recruitment and activation domains. Overexpression of Lgp2 inhibits SV and Newcastle disease virus signaling to IFN-stimulated regulatory element- and NF-kappaB-dependent pathways. Importantly, Lgp2 does not prevent TLR3 signaling. Like RIG-I, Lgp2 binds double-stranded, but not single-stranded, RNA. Quantitative PCR analysis demonstrates that Lgp2 is present in unstimulated cells at a lower level than RIG-I, although both helicases are induced to similar levels after virus infection. We propose that Lgp2 acts as a negative feedback regulator of antiviral signaling by sequestering dsRNA from RIG-I.


Asunto(s)
ARN Helicasas/fisiología , Receptores Toll-Like/fisiología , Transactivadores/fisiología , Replicación Viral/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Antígenos de Diferenciación/genética , Línea Celular , Humanos , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Noqueados , Proteínas de la Mielina/genética , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito , Factor 88 de Diferenciación Mieloide , Proteolípidos/genética , ARN Bicatenario/metabolismo , Receptores Inmunológicos/genética , Virus Sendai/fisiología , Transducción de Señal/fisiología , Factor 6 Asociado a Receptor de TNF/genética , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Transactivadores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA