Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 295(51): 17842-17851, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33454018

RESUMEN

Animals can sense the presence of microbes in their tissues and mobilize their own defenses by recognizing and responding to conserved microbial structures (often called microbe-associated molecular patterns (MAMPs)). Successful host defenses may kill the invaders, yet the host animal may fail to restore homeostasis if the stimulatory microbial structures are not silenced. Although mice have many mechanisms for limiting their responses to lipopolysaccharide (LPS), a major Gram-negative bacterial MAMP, a highly conserved host lipase is required to extinguish LPS sensing in tissues and restore homeostasis. We review recent progress in understanding how this enzyme, acyloxyacyl hydrolase (AOAH), transforms LPS from stimulus to inhibitor, reduces tissue injury and death from infection, prevents prolonged post-infection immunosuppression, and keeps stimulatory LPS from entering the bloodstream. We also discuss how AOAH may increase sensitivity to pulmonary allergens. Better appreciation of how host enzymes modify LPS and other MAMPs may help prevent tissue injury and hasten recovery from infection.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Bacterias Gramnegativas/metabolismo , Lipopolisacáridos/metabolismo , Animales , Hidrolasas de Éster Carboxílico/química , Hidrolasas de Éster Carboxílico/genética , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Antígeno 96 de los Linfocitos/química , Antígeno 96 de los Linfocitos/metabolismo , Neutrófilos/metabolismo , Receptor Toll-Like 4/química , Receptor Toll-Like 4/metabolismo
2.
J Leukoc Biol ; 106(1): 127-132, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30694581

RESUMEN

Proinflammatory immune responses to Gram-negative bacterial lipopolysaccharides (LPS) are crucial to innate host defenses but can also contribute to pathology. How host cells sensitively detect structural features of LPS was a mystery for years, especially given that a portion of the molecule essential for its potent proinflammatory properties-lipid A-is buried in the bacterial membrane. Studies of responses to extracellular and vacuolar LPS revealed a crucial role for accessory proteins that specifically bind LPS-rich membranes and extract LPS monomers to generate a complex of LPS, MD-2, and TLR4. These insights provided means to understand better both the remarkable host sensitivity to LPS and the means whereby specific LPS structural features are discerned. More recently, the noncanonical inflammasome, consisting of caspases-4/5 in humans and caspase-11 in mice, has been demonstrated to mediate responses to LPS that has reached the host cytosol. Precisely how LPS gains access to cytosolic caspases-and in what form-is not well characterized, and understanding this process will provide crucial insights into how the noncanonical inflammasome is regulated during infection. Herein, we briefly review what is known about LPS detection by cytosolic caspases-4/5/11, focusing on lessons derived from studies of the better-characterized TLR4 system that might direct future mechanistic questions.


Asunto(s)
Citosol/química , Lipopolisacáridos/análisis , Antígeno 96 de los Linfocitos/fisiología , Receptor Toll-Like 4/fisiología , Animales , Caspasas/fisiología , Humanos , Inflamasomas/fisiología , Lipopolisacáridos/química , Lipopolisacáridos/farmacología
3.
J Immunol ; 200(9): 3047-3052, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29602772

RESUMEN

The NLRP3 inflammasome is activated in response to microbial and danger signals, resulting in caspase-1-dependent secretion of the proinflammatory cytokines IL-1ß and IL-18. Canonical NLRP3 inflammasome activation is a two-step process requiring both priming and activation signals. During inflammasome activation, NLRP3 associates with mitochondria; however, the role for this interaction is unclear. In this article, we show that mouse NLRP3 and caspase-1 independently interact with the mitochondrial lipid cardiolipin, which is externalized to the outer mitochondrial membrane at priming in response to reactive oxygen species. An NLRP3 activation signal is then required for the calcium-dependent association of the adaptor molecule ASC with NLRP3 on the mitochondrial surface, resulting in inflammasome complex assembly and activation. These findings demonstrate a novel lipid interaction for caspase-1 and identify a role for mitochondria as supramolecular organizing centers in the assembly and activation of the NLRP3 inflammasome.


Asunto(s)
Cardiolipinas/metabolismo , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Cardiolipinas/inmunología , Caspasa 1/inmunología , Inflamasomas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología
4.
Innate Immun ; 23(3): 307-318, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28359219

RESUMEN

The pro-inflammatory potency and causal relationship with asthma of inhaled endotoxins have underscored the importance of accurately assessing the endotoxin content of organic dusts. The Limulus amebocyte lysate (LAL) assay has emerged as the preferred assay, but its ability to measure endotoxin in intact bacteria and organic dusts with similar sensitivity as purified endotoxin is unknown. We used metabolically radiolabeled Neisseria meningitidis and both rough and smooth Escherichia coli to compare dose-dependent activation in the LAL with purified endotoxin from these bacteria and shed outer membrane (OM) blebs. Labeled [14C]-3-OH-fatty acids were used to quantify the endotoxin content of the samples. Purified meningococcal and E. coli endotoxins and OM blebs displayed similar specific activity in the LAL assay to the purified LPS standard. In contrast, intact bacteria exhibited fivefold lower specific activity in the LAL assay but showed similar MD-2-dependent potency as purified endotoxin in inducing acute airway inflammation in mice. Pre-treatment of intact bacteria and organic dusts with 0.1 M Tris-HCl/10 mM EDTA increased by fivefold the release of endotoxin. These findings demonstrate that house dust and other organic dusts should be extracted with Tris/EDTA to more accurately assess the endotoxin content and pro-inflammatory potential of these environmental samples.


Asunto(s)
Endotoxinas/metabolismo , Escherichia coli/inmunología , Prueba de Limulus/métodos , Neisseria meningitidis/inmunología , Neumonía/inmunología , Animales , Radioisótopos de Carbono , Errores Diagnósticos/prevención & control , Polvo/análisis , Endotoxinas/inmunología , Humanos , Antígeno 96 de los Linfocitos/genética , Antígeno 96 de los Linfocitos/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Sensibilidad y Especificidad
5.
Innate Immun ; 23(4): 336-344, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28409545

RESUMEN

Caspases of the non-canonical inflammasome (caspases -4, -5, and -11) directly bind endotoxin (LOS/LPS) and can be activated in the absence of any co-factors. Models of LPS-induced caspase activation have postulated that 1:1 binding of endotoxin monomers to caspase trigger caspase oligomerization and activation, analogous to that established for endotoxin-induced activation of MD-2/TLR4. However, using metabolically radiolabeled LOS and LPS, we now show high affinity and selective binding of caspase-4 to high molecular mass aggregates of purified endotoxin and to endotoxin-rich outer membrane vesicles without formation of 1:1 endotoxin:caspase complexes. Thus, our findings demonstrate markedly different endotoxin recognition properties of caspase-4 from that of MD-2/TLR4 and strongly suggest that activation of caspase-4 (and presumably caspase-5 and caspase-11) are mediated by interactions with activating endotoxin-rich membrane interfaces rather than by endotoxin monomers.


Asunto(s)
Caspasas Iniciadoras/metabolismo , Vesículas Citoplasmáticas/metabolismo , Lipopolisacáridos/metabolismo , Membranas Mitocondriales/metabolismo , Neisseria meningitidis/inmunología , Protoplastos/metabolismo , Staphylococcus aureus/inmunología , Caspasas Iniciadoras/genética , Pared Celular/metabolismo , Humanos , Unión Proteica , Multimerización de Proteína , Proteínas Recombinantes/genética
6.
PLoS One ; 11(6): e0157842, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27326857

RESUMEN

Francisella tularensis, the Gram-negative bacterium that causes tularemia, produces a high molecular weight capsule that is immunologically distinct from Francisella lipopolysaccharide but contains the same O-antigen tetrasaccharide. To pursue the possibility that the capsule of Francisella live vaccine strain (LVS) has a structurally unique lipid anchor, we have metabolically labeled Francisella with [14C]acetate to facilitate highly sensitive compositional analysis of capsule-associated lipids. Capsule was purified by two independent methods and yielded similar results. Autoradiographic and immunologic analysis confirmed that this purified material was largely devoid of low molecular weight LPS and of the copious amounts of free lipid A that the Francisellae accumulate. Chemical hydrolysis yielded [14C]-labeled free fatty acids characteristic of Francisella lipid A but with a different molar ratio of 3-OH C18:0 to 3-OH C16:0 and different composition of non-hydroxylated fatty acids (mainly C14:0 rather than C16:0) than that of free Francisella lipid A. Mild acid hydrolysis to induce selective cleavage of KDO-lipid A linkage yielded a [14C]-labeled product that partitioned during Bligh/Dyer extraction and migrated during thin-layer chromatography like lipid A. These findings suggest that the O-antigen capsule of Francisella contains a covalently linked and structurally distinct lipid A species. The presence of a discrete lipid A-like molecule associated with capsule raises the possibility that Francisella selectively exploits lipid A structural heterogeneity to regulate synthesis, transport, and stable bacterial surface association of the O-antigen capsular layer.


Asunto(s)
Cápsulas Bacterianas/química , Francisella tularensis/inmunología , Lípido A/química , Antígenos O/química , Ácido Desoxicólico , Electroforesis en Gel de Poliacrilamida , Ácidos Grasos/análisis , Concentración de Iones de Hidrógeno , Immunoblotting , Lipopolisacáridos/química , Modelos Biológicos , Peso Molecular , Antígenos O/aislamiento & purificación
7.
J Immunol ; 196(5): 2309-18, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26826249

RESUMEN

Myeloid differentiation factor 2 (MD-2) is an extracellular protein, associated with the ectodomain of TLR4, that plays a critical role in the recognition of bacterial LPS. Despite high overall structural and functional similarity, human (h) and murine (m) MD-2 exhibit several species-related differences. hMD-2 is capable of binding LPS in the absence of TLR4, whereas mMD-2 supports LPS responsiveness only when mMD-2 and mTLR4 are coexpressed in the same cell. Previously, charged residues at the edge of the LPS binding pocket have been attributed to this difference. In this study, site-directed mutagenesis was used to explore the hydrophobic residues within the MD-2 binding pocket as the source of functional differences between hMD-2 and mMD-2. Whereas decreased hydrophobicity of residues 61 and 63 in the hMD-2 binding pocket retained the characteristics of wild-type hMD-2, a relatively minor change of valine to alanine at position 135 completely abolished the binding of LPS to the hMD-2 mutant. The mutant, however, retained the LPS binding in complex with TLR4 and also cell activation, resulting in a murine-like phenotype. These results were supported by the molecular dynamics simulation. We propose that the residue at position 135 of MD-2 governs the dynamics of the binding pocket and its ability to accommodate lipid A, which is allosterically affected by bound TLR4.


Asunto(s)
Antígeno 96 de los Linfocitos/genética , Antígeno 96 de los Linfocitos/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Transporte Biológico , Línea Celular , Expresión Génica , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/metabolismo , Antígeno 96 de los Linfocitos/química , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Alineación de Secuencia , Relación Estructura-Actividad , Receptor Toll-Like 4/metabolismo
8.
mBio ; 6(5): e01193-15, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26463160

RESUMEN

UNLABELLED: Mammalian lipopolysaccharide (LPS) binding proteins (LBPs) occur mainly in extracellular fluids and promote LPS delivery to specific host cell receptors. The function of LBPs has been studied principally in the context of host defense; the possible role of LBPs in nonpathogenic host-microbe interactions has not been well characterized. Using the Euprymna scolopes-Vibrio fischeri model, we analyzed the structure and function of an LBP family protein, E. scolopes LBP1 (EsLBP1), and provide evidence for its role in triggering a symbiont-induced host developmental program. Previous studies showed that, during initial host colonization, the LPS of V. fischeri synergizes with peptidoglycan (PGN) monomer to induce morphogenesis of epithelial tissues of the host animal. Computationally modeled EsLBP1 shares some but not all structural features of mammalian LBPs that are thought important for LPS binding. Similar to human LBP, recombinant EsLBP1 expressed in insect cells bound V. fischeri LPS and Neisseria meningitidis lipooligosaccharide (LOS) with nanomolar or greater affinity but bound Francisella tularensis LPS only weakly and did not bind PGN monomer. Unlike human LBP, EsLBP1 did not bind N. meningitidis LOS:CD14 complexes. The eslbp1 transcript was upregulated ~22-fold by V. fischeri at 24 h postinoculation. Surprisingly, this upregulation was not induced by exposure to LPS but, rather, to the PGN monomer alone. Hybridization chain reaction-fluorescent in situ hybridization (HCR-FISH) and immunocytochemistry (ICC) localized eslbp1 transcript and protein in crypt epithelia, where V. fischeri induces morphogenesis. The data presented here provide a window into the evolution of LBPs and the scope of their roles in animal symbioses. IMPORTANCE: Mammalian lipopolysaccharide (LPS)-binding protein (LBP) is implicated in conveying LPS to host cells and potentiating its signaling activity. In certain disease states, such as obesity, the overproduction of this protein has been a reliable biomarker of chronic inflammation. Here, we describe a symbiosis-induced invertebrate LBP whose tertiary structure and LPS-binding characteristics are similar to those of mammalian LBPs; however, the primary structure of this distantly related squid protein (EsLBP1) differs in key residues previously believed to be essential for LPS binding, suggesting that an alternative strategy exists. Surprisingly, symbiotic expression of eslbp1 is induced by peptidoglycan derivatives, not LPS, a pattern converse to that of RegIIIγ, an important mammalian immunity protein that binds peptidoglycan but whose gene expression is induced by LPS. Finally, EsLBP1 occurs along the apical surfaces of all the host's epithelia, suggesting that it was recruited from a general defensive role to one that mediates specific interactions with its symbiont.


Asunto(s)
Proteínas de Fase Aguda/química , Proteínas de Fase Aguda/metabolismo , Aliivibrio fischeri/fisiología , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Decapodiformes/crecimiento & desarrollo , Decapodiformes/microbiología , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Simbiosis , Proteínas de Fase Aguda/genética , Aliivibrio fischeri/química , Animales , Proteínas Portadoras/genética , Decapodiformes/fisiología , Francisella tularensis/química , Perfilación de la Expresión Génica , Lipopolisacáridos/metabolismo , Glicoproteínas de Membrana/genética , Neisseria meningitidis/química , Unión Proteica , Transcripción Genética
9.
Biochim Biophys Acta ; 1848(11 Pt B): 3072-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26079797

RESUMEN

Group IIA secretory phospholipase A2 (sPLA(2)-IIA) of mammalian species is unique among the many structurally and functionally related mammalian sPLA(2) in their high net positive charge and potent (nM) antibacterial activity. Toward the Gram-positive bacteria tested thus far, the global cationic properties of sPLA(2)-IIA are necessary for optimal binding to intact bacteria and penetration of the multi-layered thick cell wall, but not for the degradation of membrane phospholipids that is essential for bacterial killing. Various Gram-positive bacterial species can differ as much as 1000-fold in sPLA(2)-IIA sensitivity despite similar intrinsic enzymatic activity of sPLA(2)-IIA toward the membrane phospholipids of various bacteria. d-alanylation of wall- and lipo-teichoic acids in Staphylococcus aureus and sortase function in Streptococcus pyogenes increase bacterial resistance to sPLA(2)-IIA by up to 100-fold apparently by affecting translocation of bound sPLA(2)-IIA to the cell membrane. Action of the sPLA(2)-IIA and other related sPLA(2) against Gram-negative bacteria is more dependent on cationic properties of the enzyme near the amino-terminus of the protein and collaboration with other host defense proteins that produce alterations of the unique Gram-negative bacterial outer membrane that normally represents a barrier to sPLA(2)-IIA action. This article is part of a Special Issue entitled: Bacterial Resistance to Antimicrobial Peptides.


Asunto(s)
Bacterias Gramnegativas/metabolismo , Infecciones por Bacterias Gramnegativas/enzimología , Bacterias Grampositivas/metabolismo , Infecciones por Bacterias Grampositivas/enzimología , Fosfolipasas A2 Grupo II/metabolismo , Fosfolípidos/metabolismo , Animales , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/patogenicidad , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/prevención & control , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/patogenicidad , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/prevención & control , Fosfolipasas A2 Grupo II/uso terapéutico , Interacciones Huésped-Patógeno , Humanos , Lipólisis , Viabilidad Microbiana , Transducción de Señal , Especificidad por Sustrato
10.
J Immunol ; 192(4): 1887-95, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24420921

RESUMEN

LPS exerts potent immunostimulatory effects through activation of the TLR4/MD-2 receptor complex. The hexaacylated lipid A is an agonist of mouse (mTLR4) and human TLR4/MD-2, whereas the tetraacylated lipid IVa and paclitaxel activate only mTLR4/MD-2 and antagonize activation of the human receptor complex. Hydrophobic mutants of TLR4 or MD-2 were used to investigate activation of human embryonic kidney 293 cells by different TLR4 agonists. We show that each of the hydrophobic residues F438 and F461, which are located on the convex face of leucine-rich repeats 16 and 17 of the mTLR4 ectodomain, are essential for activation of with lipid IVa and paclitaxel, which, although not a structural analog of LPS, activates cells expressing mTLR4/MD-2. Both TLR4 mutants were inactive when stimulated with lipid IVa or paclitaxel, but retained significant activation when stimulated with LPS or hexaacylated lipid A. We show that the phenylalanine residue at position 126 of mouse MD-2 is indispensable only for activation with paclitaxel. Its replacement with leucine or valine completely abolished activation with paclitaxel while preserving the responsiveness to lipid IVa and lipid A. This suggests specific interaction of paclitaxel with F126 because its replacement with leucine even augmented activation by lipid A. These results provide an insight into the molecular mechanism of TLR4 activation by two structurally very different agonists.


Asunto(s)
Glucolípidos/inmunología , Lípido A/análogos & derivados , Antígeno 96 de los Linfocitos/inmunología , Paclitaxel/farmacología , Receptor Toll-Like 4/inmunología , Moduladores de Tubulina/farmacología , Acilación , Animales , Sitios de Unión , Línea Celular , Activación Enzimática , Glucolípidos/química , Glucolípidos/farmacología , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Lípido A/química , Lípido A/inmunología , Lípido A/farmacología , Antígeno 96 de los Linfocitos/química , Ratones , Paclitaxel/química , Fenilalanina/química , Unión Proteica , Estructura Terciaria de Proteína , Receptor Toll-Like 4/química
11.
Innate Immun ; 20(1): 88-103, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23729477

RESUMEN

A hallmark of Francisella tularensis, a highly virulent Gram-negative bacterium, is an unusual LPS that possesses both structural heterogeneity and characteristics that may contribute to innate immune evasion. However, none of the methods yet employed has been sufficient to determine the overall LPS composition of Francisella. We now demonstrate that metabolic labeling of francisellae with [(14)C]acetate, combined with fractionation of [(14)C]acetate-labeled lipids by ethanol precipitation rather than hot phenol-water extraction, permits a more sensitive and quantitative appraisal of overall compositional heterogeneity in lipid A and LPS. The majority of lipid A of different francisellae strains grown in diverse bacteriologic media and within human phagocytes accumulated as very hydrophobic species, including free lipid A, with <10% of the lipid A molecules substituted with O-Ag polysaccharides. The spectrum of lipid A and LPS species varied in a medium- and strain-dependent fashion, and growth in THP-1 cells yielded lipid A species that were not present in the same bacteria grown in brain heart infusion broth. In summary, metabolic labeling with [(14)C]acetate greatly facilitates assessment of the effect of genotypic and/or environmental variables on the synthesis and accumulation of lipid A and LPS by Francisella, including during growth within the cytosol of infected host cells.


Asunto(s)
Francisella tularensis/fisiología , Lípido A/metabolismo , Lipopolisacáridos/química , Monocitos/inmunología , Fagocitos/inmunología , Radioisótopos de Carbono/química , Línea Celular , Proliferación Celular , Células Cultivadas , Fraccionamiento Químico , Precipitación Química , Medios de Cultivo , Etanol , Francisella tularensis/patogenicidad , Humanos , Evasión Inmune , Inmunidad Innata , Lípido A/química , Metabolismo , Monocitos/virología , Virulencia
12.
Innate Immun ; 19(5): 545-60, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23439691

RESUMEN

A purified complex of metabolically labeled [(3)H]lipooligosaccharide (LOS) and recombinant human myeloid differentiation factor 2 (MD-2), [(3)H]LOS·MD-2, has been used to demonstrate pM affinity binding interactions with soluble TLR4 ectodomain (TLR4ecd). For measurement of the binding parameters of membrane-bound TLR4, we took advantage of the stability of endotoxin·MD-2 and tyrosine(s) present on the surface of MD-2 to radioiodinate LOS·MD-2. Radioiodinated LOS·MD-2 generated a reagent with an estimated 1:1 molar ratio of [(125)I] to sMD-2 with 20-fold higher specific radioactivity and TLR4-activating properties comparable to metabolically-labeled LOS·MD-2. LOS·MD-2[(125)I] and [(3)H]LOS·MD-2 have similar affinities for soluble (FLAG) TLR4ecd and for membrane-bound TLR4 in HEK293T/TLR4 cells. In a similar dose-dependent manner, sMD-2 and LOS·MD-2 inhibit LOS·MD-2[(125)I] binding to TLR4 indicating the pM affinity binding of LOS·MD-2[(125)I] is agonist-independent. LOS·MD-2[(125)I] allowed measurement of low levels of cell-surface human or murine TLR4 expressed by stable cell lines (2000-3000 sites/cell) and quantitatively measures low levels of 'MD-2-free' TLR4 (est. 250 molecules/cell) in cells co-expressing TLR4 and MD-2. Occupation of 50-100 TLR4/cell by LOS·MD-2 is sufficient to trigger measurable TLR4-dependent cell activation. LOS·MD-2[(125)I] provides a powerful reagent to measure quantitatively functional human and murine cell-surface TLR4, including in cells where surface TLR4 is potentially functionally significant but not detectable by other methods.


Asunto(s)
Radioisótopos de Yodo/metabolismo , Lipopolisacáridos/metabolismo , Antígeno 96 de los Linfocitos/metabolismo , Complejos Multiproteicos/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Estudios de Factibilidad , Células HEK293 , Humanos , Inmunidad Innata , Ratones , Complejos Multiproteicos/química , Unión Proteica , Ensayo de Unión Radioligante , Sensibilidad y Especificidad , Receptor Toll-Like 4/genética , Transgenes/genética
13.
J Immunol ; 189(8): 4088-103, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22972924

RESUMEN

APOBEC3 (A3) proteins are virus-restriction factors that provide intrinsic immunity against infections by viruses like HIV-1 and mouse mammary tumor virus. A3 proteins are inducible by inflammatory stimuli, such as LPS and IFN-α, via mechanisms that are not fully defined. Using genetic and pharmacological studies on C57BL/6 mice and cells, we show that IFN-α and LPS induce A3 via different pathways, independently of each other. IFN-α positively regulates mouse APOBEC3 (mA3) mRNA expression through IFN-αR/PKC/STAT1 and negatively regulates mA3 mRNA expression via IFN-αR/MAPKs-signaling pathways. Interestingly, LPS shows some variation in its regulatory behavior. Although LPS-mediated positive regulation of mA3 mRNA occurs through TLR4/TRIF/IRF3/PKC, it negatively modulates mA3 mRNA via TLR4/MyD88/MAPK-signaling pathways. Additional studies on human peripheral blood mononuclear cells reveal that PKC differentially regulates IFN-α and LPS induction of human A3A, A3F, and A3G mRNA expression. In summary, we identified important signaling targets downstream of IFN-αR and TLR4 that mediate A3 mRNA induction by both LPS and IFN-α. Our results provide new insights into the signaling targets that could be manipulated to enhance the intracellular store of A3 and potentially enhance A3 antiviral function in the host.


Asunto(s)
Citidina Desaminasa/biosíntesis , Interferón-alfa/fisiología , Lipopolisacáridos/fisiología , ARN Mensajero/biosíntesis , Transducción de Señal/inmunología , Regulación hacia Arriba/inmunología , Animales , Línea Celular , Línea Celular Transformada , Citidina Desaminasa/genética , Células Dendríticas/inmunología , Células Dendríticas/patología , Células Dendríticas/virología , VIH-1/inmunología , Humanos , Mediadores de Inflamación/fisiología , Líquido Intracelular/inmunología , Líquido Intracelular/virología , Macrófagos/inmunología , Macrófagos/patología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Regulación hacia Arriba/genética
14.
mBio ; 3(4)2012.
Artículo en Inglés | MEDLINE | ID: mdl-22872782

RESUMEN

UNLABELLED: Respiratory syncytial virus (RSV) is a leading cause of infant mortality worldwide. Toll-like receptor 4 (TLR4), a signaling receptor for structurally diverse microbe-associated molecular patterns, is activated by the RSV fusion (F) protein and by bacterial lipopolysaccharide (LPS) in a CD14-dependent manner. TLR4 signaling by LPS also requires the presence of an additional protein, MD-2. Thus, it is possible that F protein-mediated TLR4 activation relies on MD-2 as well, although this hypothesis has not been formally tested. LPS-free RSV F protein was found to activate NF-κB in HEK293T transfectants that express wild-type (WT) TLR4 and CD14, but only when MD-2 was coexpressed. These findings were confirmed by measuring F-protein-induced interleukin 1ß (IL-1ß) mRNA in WT versus MD-2(-/-) macrophages, where MD-2(-/-) macrophages failed to show IL-1ß expression upon F-protein treatment, in contrast to the WT. Both Rhodobacter sphaeroides LPS and synthetic E5564 (eritoran), LPS antagonists that inhibit TLR4 signaling by binding a hydrophobic pocket in MD-2, significantly reduced RSV F-protein-mediated TLR4 activity in HEK293T-TLR4-CD14-MD-2 transfectants in a dose-dependent manner, while TLR4-independent NF-κB activation by tumor necrosis factor alpha (TNF-α) was unaffected. In vitro coimmunoprecipitation studies confirmed a physical interaction between native RSV F protein and MD-2. Further, we demonstrated that the N-terminal domain of the F1 segment of RSV F protein interacts with MD-2. These data provide new insights into the importance of MD-2 in RSV F-protein-mediated TLR4 activation. Thus, targeting the interaction between MD-2 and RSV F protein may potentially lead to novel therapeutic approaches to help control RSV-induced inflammation and pathology. IMPORTANCE: This study shows for the first time that the fusion (F) protein of respiratory syncytial virus (RSV), a major cause of bronchiolitis and death, particularly in infants and young children, physically interacts with the Toll-like receptor 4 (TLR4) coreceptor, MD-2, through its N-terminal domain. We show that F protein-induced TLR4 activation can be blocked by lipid A analog antagonists. This observation provides a strong experimental rationale for testing such antagonists in animal models of RSV infection for potential use in people.


Asunto(s)
Regulación hacia Abajo , Lípido A/análogos & derivados , Antígeno 96 de los Linfocitos/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitiales Respiratorios/metabolismo , Transducción de Señal , Receptor Toll-Like 4/inmunología , Proteínas Virales de Fusión/metabolismo , Animales , Línea Celular , Humanos , Lípido A/metabolismo , Lipopolisacáridos/metabolismo , Antígeno 96 de los Linfocitos/genética , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/microbiología , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/inmunología , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Proteínas Virales de Fusión/genética
15.
J Biol Chem ; 287(20): 16346-55, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22433852

RESUMEN

Host response to invasion by many gram-negative bacteria depends upon activation of Toll-like receptor 4 (TLR4) by endotoxin presented as a monomer bound to myeloid differentiation factor 2 (MD-2). Metabolic labeling of hexaacylated endotoxin (LOS) from Neisseria meningitidis with [(13)C]acetate allowed the use of NMR to examine structural properties of the fatty acyl chains of LOS present in TLR4-agonistic and -antagonistic binary and ternary complexes with, respectively, wild-type or mutant (F126A) MD-2 ± TLR4 ectodomain. Chemical shift perturbation indicates that Phe(126) affects the environment and/or position of each of the bound fatty acyl chains both in the binary LOS·MD-2 complex and in the ternary LOS·MD-2·TLR4 ectodomain complex. In both wild-type and mutant LOS·MD-2 complexes, one of the six fatty acyl chains of LOS is more susceptible to paramagnetic attenuation, suggesting protrusion of that fatty acyl chain from the hydrophobic pocket of MD-2, independent of association with TLR4. These findings indicate that re-orientation of the aromatic side chain of Phe(126) is induced by binding of hexaacylated E, preceding interaction with TLR4. This re-arrangement of Phe(126) may act as a "hydrophobic switch," driving agonist-dependent contacts needed for TLR4 dimerization and activation.


Asunto(s)
Endotoxinas/química , Antígeno 96 de los Linfocitos/química , Multimerización de Proteína , Receptor Toll-Like 4/química , Acetilación , Sustitución de Aminoácidos , Endotoxinas/genética , Endotoxinas/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Antígeno 96 de los Linfocitos/genética , Antígeno 96 de los Linfocitos/metabolismo , Mutación Missense , Neisseria meningitidis/química , Resonancia Magnética Nuclear Biomolecular/métodos , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
16.
Innate Immun ; 18(3): 478-91, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21994253

RESUMEN

Response to Gram-negative bacteria (GNB) is partially mediated by the recognition of GNB-derived endotoxin by host cells. Potent host response to endotoxin depends on the sequential interaction of endotoxin with lipopolysaccharide binding protein (LBP), CD14, MD-2 and TLR4. While CD14 facilitates the efficient transfer of endotoxin monomers to MD-2 and MD-2·TLR4, activation of MD-2·TLR4 can occur in the absence of CD14 through an unknown mechanism. Here, we show that incubation of purified endotoxin (E) aggregates (E(agg), M ( r ) ≥ 20 million) in PBS with ≥ 0.1% albumin in the absence of divalent cations Ca(2+) and Mg(2+), yields E·albumin complexes (M ( r ) ∼70,000). E·albumin transfers E monomers to sMD-2 or sMD-2·TLR4 ectodomain (TLR4(ecd)) with a 'K (d)' of ∼4 nM and induces MD-2·TLR4-dependent, CD14-independent cell activation with a potency only 10-fold less than that of monomeric E·CD14 complexes. Our findings demonstrate, for the first time, a mechanistic basis for delivery of endotoxin monomers to MD-2 and for activation of TLR4 that is independent of CD14.


Asunto(s)
Endotoxinas/metabolismo , Infecciones por Bacterias Grampositivas/inmunología , Antígeno 96 de los Linfocitos/metabolismo , Macrófagos/inmunología , Receptor Toll-Like 4/metabolismo , Albúminas/metabolismo , Animales , Línea Celular , Interacciones Huésped-Patógeno , Humanos , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Polimerizacion , Ingeniería de Proteínas , Transporte de Proteínas
17.
J Exp Med ; 209(1): 157-71, 2012 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-22184635

RESUMEN

Alarmins are endogenous mediators capable of promoting the recruitment and activation of antigen-presenting cells (APCs), including dendritic cells (DCs), that can potentially alert host defense against danger signals. However, the relevance of alarmins to the induction of adaptive immune responses remains to be demonstrated. In this study, we report the identification of HMGN1 (high-mobility group nucleosome-binding protein 1) as a novel alarmin and demonstrate that it contributes to the induction of antigen-specific immune responses. HMGN1 induced DC maturation via TLR4 (Toll-like receptor 4), recruitment of APCs at sites of injection, and activation of NF-κB and multiple mitogen-activated protein kinases in DCs. HMGN1 promoted antigen-specific immune response upon co-administration with antigens, and Hmgn1(-/-) mice developed greatly reduced antigen-specific antibody and T cell responses when immunized with antigens in the presence of lipopolysaccharide (LPS). The impaired ability of Hmgn1(-/-) mice to mount antigen-specific immune responses was accompanied by both deficient DC recruitment at sites of immunization and reduced production of inflammatory cytokines. Bone marrow chimera experiments revealed that HMGN1 derived from nonleukocytes was critical for the induction of antigen-specific antibody and T cell responses. Thus, extracellular HMGN1 acts as a novel alarmin critical for LPS-induced development of innate and adaptive immune responses.


Asunto(s)
Proteína HMGN1/metabolismo , Inmunidad , Lipopolisacáridos/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antígenos/inmunología , Diferenciación Celular , Línea Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Células HEK293 , Proteína HMGN1/genética , Proteína HMGN1/inmunología , Humanos , Inmunidad/genética , Inmunidad Innata/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Fenotipo , Transducción de Señal , Receptor Toll-Like 4/metabolismo
18.
Sci Transl Med ; 3(110): 110ra118, 2011 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-22116933

RESUMEN

Identification of safe, effective treatments to mitigate toxicity after extensive radiation exposure has proven challenging. Only a limited number of candidate approaches have emerged, and the U.S. Food and Drug Administration has yet to approve any agent for a mass-casualty radiation disaster. Because patients undergoing hematopoietic stem cell transplantation undergo radiation treatment that produces toxicities similar to radiation-disaster exposure, we studied patients early after such treatment to identify new approaches to this problem. Patients rapidly developed endotoxemia and reduced plasma bactericidal/permeability-increasing protein (BPI), a potent endotoxin-neutralizing protein, in association with neutropenia. We hypothesized that a treatment supplying similar endotoxin-neutralizing activity might replace the BPI deficit and mitigate radiation toxicity and tested this idea in mice. A single 7-Gy radiation dose, which killed 95% of the mice by 30 days, was followed 24 hours later by twice-daily, subcutaneous injections of the recombinant BPI fragment rBPI21 or vehicle alone for 14 or 30 days, with or without an oral fluoroquinolone antibiotic with broad-spectrum antibacterial activity, including that against endotoxin-bearing Gram-negative bacteria. Compared to either fluoroquinolone alone or vehicle plus fluoroquinolone, the combined rBPI21 plus fluoroquinolone treatment improved survival, accelerated hematopoietic recovery, and promoted expansion of stem and progenitor cells. The observed efficacy of rBPI21 plus fluoroquinolone initiated 24 hours after lethal irradiation, combined with their established favorable bioactivity and safety profiles in critically ill humans, suggests the potential clinical use of this radiation mitigation strategy and supports its further evaluation.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/uso terapéutico , Proteínas Sanguíneas/uso terapéutico , Médula Ósea/patología , Fluoroquinolonas/uso terapéutico , Traumatismos por Radiación/tratamiento farmacológico , Técnicas de Ablación , Animales , Péptidos Catiónicos Antimicrobianos/administración & dosificación , Péptidos Catiónicos Antimicrobianos/farmacología , Recuento de Células Sanguíneas , Proteínas Sanguíneas/administración & dosificación , Proteínas Sanguíneas/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Citocinas/sangre , Endotoxemia/sangre , Endotoxemia/complicaciones , Endotoxinas/metabolismo , Enrofloxacina , Fluoroquinolonas/administración & dosificación , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Trasplante de Células Madre Hematopoyéticas , Humanos , Mediadores de Inflamación/sangre , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos BALB C , Neutropenia/sangre , Neutropenia/complicaciones , Tamaño de los Órganos/efectos de los fármacos , Tamaño de los Órganos/efectos de la radiación , Traumatismos por Radiación/sangre , Traumatismos por Radiación/complicaciones , Análisis de Supervivencia , Irradiación Corporal Total
19.
Biochem Soc Trans ; 39(4): 1039-44, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21787344

RESUMEN

LBP [LPS (lipopolysaccharide)-binding protein] and BPI (bactericidal/permeability-increasing protein) are components of the immune system that have been principally studied in mammals for their involvement in defence against bacterial pathogens. These proteins share a basic architecture and residues involved in LPS binding. Putative orthologues, i.e. proteins encoded by similar genes that diverged from a common ancestor, have been found in a number of non-mammalian vertebrate species and several non-vertebrates. Similar to other aspects of immunity, such as the activity of Toll-like receptors and NOD (nucleotide-binding oligomerization domain) proteins, analysis of the conservation of LBPs and BPIs in the invertebrates promises to provide insight into features essential to the form and function of these molecules. This review considers state-of-the-art knowledge in the diversity of the LBP/BPI proteins across the eukaryotes and also considers their role in mutualistic symbioses. Recent studies of the LBPs and BPIs in an invertebrate model of beneficial associations, the Hawaiian bobtail squid Euprymna scolopes' alliance with the marine luminous bacterium Vibrio fischeri, are discussed as an example of the use of non-vertebrate models for the study of LBPs and BPIs.


Asunto(s)
Proteínas de Fase Aguda/genética , Péptidos Catiónicos Antimicrobianos/genética , Proteínas Sanguíneas/genética , Proteínas Portadoras/genética , Secuencia Conservada , Evolución Molecular , Glicoproteínas de Membrana/genética , Proteínas de Fase Aguda/metabolismo , Secuencia de Aminoácidos , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas Sanguíneas/metabolismo , Proteínas Portadoras/metabolismo , Humanos , Metabolismo de los Lípidos , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Unión Proteica , Homología de Secuencia de Aminoácido , Simbiosis/fisiología
20.
J Leukoc Biol ; 90(3): 459-70, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21653234

RESUMEN

Detection and clearance of bacterial infection require balanced effector and resolution signals to avoid chronic inflammation. Detection of GNB LPS by TLR4 on m induces inflammatory responses, contributing to chronic inflammation and tissue injury. LXs and Rvs are endogenous lipid mediators that enhance resolution of inflammation, and their actions on primary human m responses toward GNB are largely uncharacterized. Here, we report that LXA(4), LXB(4), and RvD1, tested at 0.1-1 µM, inhibited LPS-induced TNF production from primary human m, with ATL and 17(R)-RvD1, demonstrating potent inhibition at 0.1 µM. In addition, 17(R)-RvD1 inhibited LPS-induced primary human m production of IL-7, IL-12p70, GM-CSF, IL-8, CCL2, and MIP-1α without reducing that of IL-6 or IL-10. Remarkably, when stimulated with live Escherichia coli, m treated with 17(R)-RvD1 demonstrated increased TNF production and enhanced internalization and killing of the bacteria. 17(R)-RvD1-enhanced TNF, internalization, and killing were not evident for an lpxM mutant of E. coli expressing hypoacylated LPS with reduced inflammatory activity. Furthermore, 17(R)-RvD1-enhanced, E. coli-induced TNF production was evident in WT but not TLR4-deficient murine m. Thus, Rvs differentially modulate primary human m responses to E. coli in an LPS- and TLR4-dependent manner, such that this Rv could promote resolution of GNB/LPS-driven inflammation by reducing m proinflammatory responses to isolated LPS and increasing m responses important for clearance of infection.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Escherichia coli/patogenicidad , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Western Blotting , Moléculas de Adhesión Celular/genética , Diferenciación Celular , Proliferación Celular , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Humanos , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Mutación/genética , Fosfoproteínas/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...