Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34290146

RESUMEN

Many endogenous molecules, mostly proteins, purportedly activate the Toll-like receptor 4 (TLR4)-myeloid differentiation factor-2 (MD-2) complex, the innate immune receptor for lipopolysaccharide (LPS) derived from gram-negative bacteria. However, there is no structural evidence supporting direct TLR4-MD-2 activation by endogenous ligands. Sulfatides (3-O-sulfogalactosylceramides) are natural, abundant sulfated glycolipids that have variously been shown to initiate or suppress inflammatory responses. We show here that short fatty acid (FA) chain sulfatides directly activate mouse TLR4-MD-2 independent of CD14, trigger MyD88- and TRIF-dependent signaling, and stimulate tumor necrosis factor α (TNFα) and type I interferon (IFN) production in mouse macrophages. In contrast to the agonist activity toward the mouse receptor, the tested sulfatides antagonize TLR4-MD-2 activation by LPS in human macrophage-like cells. The agonistic and antagonistic activities of sulfatides require the presence of the sulfate group and are inversely related to the FA chain length. The crystal structure of mouse TLR4-MD-2 in complex with C16-sulfatide revealed that three C16-sulfatide molecules bound to the MD-2 hydrophobic pocket and induced an active dimer conformation of the receptor complex similar to that induced by LPS or lipid A. The three C16-sulfatide molecules partially mimicked the detailed interactions of lipid A to achieve receptor activation. Our results suggest that sulfatides may mediate sterile inflammation or suppress LPS-stimulated inflammation, and that additional endogenous negatively charged lipids with up to six lipid chains of limited length might also bind to TLR4-MD-2 and activate or inhibit this complex.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Antígeno 96 de los Linfocitos/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Sulfoglicoesfingolípidos/farmacología , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Línea Celular , Femenino , Humanos , Antígeno 96 de los Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación de Dinámica Molecular , Factor 88 de Diferenciación Mieloide/genética , Sulfoglicoesfingolípidos/química , Receptor Toll-Like 4/genética
2.
Nat Immunol ; 11(9): 814-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20711192

RESUMEN

Allergies to nickel (Ni(2+)) are the most frequent cause of contact hypersensitivity (CHS) in industrialized countries. The efficient development of CHS requires both a T lymphocyte-specific signal and a proinflammatory signal. Here we show that Ni(2+) triggered an inflammatory response by directly activating human Toll-like receptor 4 (TLR4). Ni(2+)-induced TLR4 activation was species-specific, as mouse TLR4 could not generate this response. Studies with mutant TLR4 proteins revealed that the non-conserved histidines 456 and 458 of human TLR4 are required for activation by Ni(2+) but not by the natural ligand lipopolysaccharide. Accordingly, transgenic expression of human TLR4 in TLR4-deficient mice allowed efficient sensitization to Ni(2+) and elicitation of CHS. Our data implicate site-specific human TLR4 inhibition as a potential strategy for therapeutic intervention in CHS that would not affect vital immune responses.


Asunto(s)
Dermatitis por Contacto , Níquel/inmunología , Receptor Toll-Like 4/inmunología , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Recombinantes/inmunología , Transducción de Señal , Receptor Toll-Like 4/genética
3.
PLoS Pathog ; 14(3): e1006914, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29522575

RESUMEN

Macrophages are a diverse group of phagocytic cells acting in host protection against stress, injury, and pathogens. Here, we show that the scavenger receptor SR-A6 is an entry receptor for human adenoviruses in murine alveolar macrophage-like MPI cells, and important for production of type I interferon. Scavenger receptors contribute to the clearance of endogenous proteins, lipoproteins and pathogens. Knockout of SR-A6 in MPI cells, anti-SR-A6 antibody or the soluble extracellular SR-A6 domain reduced adenovirus type-C5 (HAdV-C5) binding and transduction. Expression of murine SR-A6, and to a lower extent human SR-A6 boosted virion binding to human cells and transduction. Virion clustering by soluble SR-A6 and proximity localization with SR-A6 on MPI cells suggested direct adenovirus interaction with SR-A6. Deletion of the negatively charged hypervariable region 1 (HVR1) of hexon reduced HAdV-C5 binding and transduction, implying that the viral ligand for SR-A6 is hexon. SR-A6 facilitated macrophage entry of HAdV-B35 and HAdV-D26, two important vectors for transduction of hematopoietic cells and human vaccination. The study highlights the importance of scavenger receptors in innate immunity against human viruses.


Asunto(s)
Infecciones por Adenoviridae/virología , Pulmón/virología , Macrófagos Alveolares/virología , Macrófagos/virología , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/fisiología , Internalización del Virus , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/metabolismo , Adenovirus Humanos/inmunología , Animales , Humanos , Inmunidad Innata , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptores Inmunológicos/genética
4.
Immunity ; 29(5): 720-33, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-18951048

RESUMEN

Granzyme A (GzmA) is considered a major proapoptotic protease. We have discovered that GzmA-induced cell death involves rapid membrane damage that depends on the synergy between micromolar concentrations of GzmA and sublytic perforin (PFN). Ironically, GzmA and GzmB, independent of their catalytic activity, both mediated this swift necrosis. Even without PFN, lower concentrations of human GzmA stimulated monocytic cells to secrete proinflammatory cytokines (interleukin-1beta [IL-1beta], TNFalpha, and IL-6) that were blocked by a caspase-1 inhibitor. Moreover, murine GzmA and GzmA(+) cytotoxic T lymphocytes (CTLs) induce IL-1beta from primary mouse macrophages, and GzmA(-/-) mice resist lipopolysaccharide-induced toxicity. Thus, the granule secretory pathway plays an unexpected role in inflammation, with GzmA acting as an endogenous modulator.


Asunto(s)
Granzimas/inmunología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Leucocitos Mononucleares/inmunología , Perforina/inmunología , Linfocitos T Citotóxicos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Adenoviridae/inmunología , Animales , Adhesión Celular , Muerte Celular , Línea Celular Tumoral , Citotoxicidad Inmunológica , Técnicas de Silenciamiento del Gen , Granzimas/metabolismo , Células HeLa , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Células Jurkat , Macrófagos/inmunología , Ratones , Perforina/metabolismo , Linfocitos T Citotóxicos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células U937
5.
Immunol Cell Biol ; 94(4): 322-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26437769

RESUMEN

The adenylate cyclase toxin-hemolysin (CyaA) of Bordetella pertussis is a bi-functional leukotoxin. It penetrates myeloid phagocytes expressing the complement receptor 3 and delivers into their cytosol its N-terminal adenylate cyclase enzyme domain (~400 residues). In parallel, ~1300 residue-long RTX hemolysin moiety of CyaA forms cation-selective pores and permeabilizes target cell membrane for efflux of cytosolic potassium ions. The non-enzymatic CyaA-AC(-) toxoid, has repeatedly been successfully exploited as an antigen delivery tool for stimulation of adaptive T-cell immune responses. We show that the pore-forming activity confers on the CyaA-AC(-) toxoid a capacity to trigger Toll-like receptor and inflammasome signaling-independent maturation of CD11b-expressing dendritic cells (DC). The DC maturation-inducing potency of mutant toxoid variants in vitro reflected their specifically enhanced or reduced pore-forming activity and K(+) efflux. The toxoid-induced in vitro phenotypic maturation of DC involved the activity of mitogen activated protein kinases p38 and JNK and comprised increased expression of maturation markers, interleukin 6, chemokines KC and LIX and granulocyte-colony-stimulating factor secretion, prostaglandin E2 production and enhancement of chemotactic migration of DC. Moreover, i.v. injected toxoids induced maturation of splenic DC in function of their cell-permeabilizing capacity. Similarly, the capacity of DC to stimulate CD8(+) and CD4(+) T-cell responses in vitro and in vivo was dependent on the pore-forming activity of CyaA-AC(-). This reveals a novel self-adjuvanting capacity of the CyaA-AC(-) toxoid that is currently under clinical evaluation as a tool for delivery of immunotherapeutic anti-cancer CD8(+) T-cell vaccines into DC.


Asunto(s)
Toxina de Adenilato Ciclasa/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos , Proteínas Citotóxicas Formadoras de Poros/inmunología , Dominios Proteicos/inmunología , Toxina de Adenilato Ciclasa/genética , Adyuvantes Inmunológicos/genética , Animales , Vacunas contra el Cáncer/inmunología , Diferenciación Celular , Permeabilidad de la Membrana Celular , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/microbiología , Transporte Iónico , Ratones , Ratones Endogámicos C57BL , Proteínas Citotóxicas Formadoras de Poros/genética , Dominios Proteicos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(24): E2191-8, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23708119

RESUMEN

Macrophages are diverse cell types in the first line of antimicrobial defense. Only a limited number of primary mouse models exist to study their function. Bone marrow-derived, macrophage-CSF-induced cells with a limited life span are the most common source. We report here a simple method yielding self-renewing, nontransformed, GM-CSF/signal transducer and activator of transcription 5-dependent macrophages (Max Planck Institute cells) from mouse fetal liver, which reflect the innate immune characteristics of alveolar macrophages. Max Planck Institute cells are exquisitely sensitive to selected microbial agents, including bacterial LPS, lipopeptide, Mycobacterium tuberculosis, cord factor, and adenovirus and mount highly proinflammatory but no anti-inflammatory IL-10 responses. They show a unique pattern of innate responses not yet observed in other mononuclear phagocytes. This includes differential LPS sensing and an unprecedented regulation of IL-1α production upon LPS exposure, which likely plays a key role in lung inflammation in vivo. In conclusion, Max Planck Institute cells offer an useful tool to study macrophage biology and for biomedical science.


Asunto(s)
Células de la Médula Ósea/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Macrófagos Alveolares/inmunología , Macrófagos/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucina-1alfa/genética , Interleucina-1alfa/inmunología , Interleucina-1alfa/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mycobacterium tuberculosis/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fagocitosis/inmunología , Propionibacterium acnes/inmunología , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Factor de Transcripción STAT5/metabolismo , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Transcriptoma/inmunología
7.
BMC Pulm Med ; 15: 75, 2015 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-26204953

RESUMEN

BACKGROUND: Propionibacterium acnes was found in lungs and lymph nodes of patients with sarcoidosis and may induce hypersensitivity type granuloma formation. Data regarding the immune response to P. acnes of European sarcoid patients are scarce. METHODS: We assessed the total IgG and IgA amount and specific antibodies to P. acnes and to Staphylococcus aureus, serving as a control, in BAL fluid of 64 patients with sarcoidosis and of 21 healthy volunteers. In a subcohort of sarcoid patients and controls, TNF-α and GM-CSF production of BAL cells stimulated with heat-killed P. acnes were measured. RESULTS: In sarcoid patients, the total IgG and IgA levels in BAL fluid were significantly elevated compared to healthy volunteers. IgG and IgA titres against P. acnes and S. aureus were increased in sarcoid patients, yet based on the total amount of antibodies, only antibodies directed against P. acnes were relatively and significantly increased. Furthermore, BAL cells of sarcoid patients produced significantly more TNF-α and GM-CSF upon stimulation with heat-killed P. acnes compared to controls. CONCLUSIONS: Patients with sarcoidosis had elevated levels of specific antibodies against P. acnes which suggest contact with this bacterium in the past. Furthermore, BAL cells of sarcoid patients produced inflammatory cytokines (TNF-α and GM-CSF) upon stimulation with P. acnes indicating potential involvement of this pathogen in the pathogenesis of sarcoidosis in some patients.


Asunto(s)
Infecciones por Bacterias Grampositivas/complicaciones , Inmunidad Innata , Inmunoglobulinas/inmunología , Propionibacterium acnes/inmunología , Sarcoidosis Pulmonar/inmunología , Adulto , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Líquido del Lavado Bronquioalveolar/microbiología , Broncoscopía , Ensayo de Inmunoadsorción Enzimática , Femenino , Infecciones por Bacterias Grampositivas/inmunología , Humanos , Masculino , Persona de Mediana Edad , Propionibacterium acnes/aislamiento & purificación , Sarcoidosis Pulmonar/complicaciones , Sarcoidosis Pulmonar/microbiología
8.
J Immunol ; 189(1): 328-36, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22634614

RESUMEN

Recognition of foreign nucleic acids is important for the induction of an innate immune response against invading pathogens. Although the pathways involved in sensing bacterial DNA and viral RNA are now well established, only limited knowledge is available on mechanisms underlying recognition of bacterial RNA. It has been reported that intracellular delivery of Escherichia coli RNA activates the Nlrp3 inflammasome, but whether this is a general property of bacterial RNA remains unclear as are the pathways involved in pro-IL-1ß induction and caspase-1 activation by bacterial RNA. In this study, we report that bacterial RNA from both Gram-positive and Gram-negative bacteria induces activation of caspase-1 and secretion of IL-1ß by murine dendritic cells and bone-marrow derived macrophages. Stimulation was independent of the presence of 5'-triphosphate termini and occurred with whole RNA preparations from bacteria but not from eukaryotes. Induction of pro-IL-1ß as well as the priming for caspase-1 activation by bacterial RNA was dependent on UNC93B, an endoplasmic reticulum protein essential for delivery of TLRs to the endosome, whereas the established nucleic acid sensing endosomal TLRs 3, 7, and 9 were dispensable. Additionally, caspase-1 activation and IL-1ß production by transfected bacterial RNA were absent in MyD88-deficient cells but independent of TRIF. Thus, our data indicate the presence of a yet unidentified intracellular nucleic acid receptor involved in bacterial RNA-induced inflammasome activation and release of IL-1ß.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Caspasa 1/metabolismo , Interleucina-1beta/metabolismo , Glicoproteínas de Membrana/fisiología , Proteínas de Transporte de Membrana/fisiología , ARN Bacteriano/fisiología , Receptor Toll-Like 3/fisiología , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 9/fisiología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Animales , Línea Celular , Células Dendríticas/enzimología , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Activación Enzimática/genética , Macrófagos/enzimología , Macrófagos/metabolismo , Macrófagos/microbiología , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 7/deficiencia , Receptor Toll-Like 9/deficiencia
9.
J Innate Immun ; 16(1): 226-247, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38527452

RESUMEN

INTRODUCTION: While TLR ligands derived from microbial flora and pathogens are important activators of the innate immune system, a variety of factors such as intracellular bacteria, viruses, and parasites can induce a state of hyperreactivity, causing a dysregulated and potentially life-threatening cytokine over-response upon TLR ligand exposure. Type I interferon (IFN-αß) is a central mediator in the induction of hypersensitivity and is strongly expressed in splenic conventional dendritic cells (cDC) and marginal zone macrophages (MZM) when mice are infected with adenovirus. This study investigates the ability of adenoviral infection to influence the activation state of the immune system and underlines the importance of considering this state when planning the treatment of patients. METHODS: Infection with adenovirus-based vectors (Ad) or pretreatment with recombinant IFN-ß was used as a model to study hypersensitivity to lipopolysaccharide (LPS) in mice, murine macrophages, and human blood samples. The TNF-α, IL-6, IFN-αß, and IL-10 responses induced by LPS after pretreatment were measured. Mouse knockout models for MARCO, IFN-αßR, CD14, IRF3, and IRF7 were used to probe the mechanisms of the hypersensitive reaction. RESULTS: We show that, similar to TNF-α and IL-6 but not IL-10, the induction of IFN-αß by LPS increases strongly after Ad infection. This is true both in mice and in human blood samples ex vivo, suggesting that the regulatory mechanisms seen in the mouse are also present in humans. In mice, the scavenger receptor MARCO on IFN-αß-producing cDC and splenic marginal zone macrophages is important for Ad uptake and subsequent cytokine overproduction by LPS. Interestingly, not all IFN-αß-pretreated macrophage types exposed to LPS exhibit an enhanced TNF-α and IL-6 response. Pretreated alveolar macrophages and alveolar macrophage-like murine cell lines (MPI cells) show enhanced responses, while bone marrow-derived and peritoneal macrophages show a weaker response. This correlates with the respective absence or presence of the anti-inflammatory IL-10 response in these different macrophage types. In contrast, Ad or IFN-ß pretreatment enhances the subsequent induction of IFN-αß in all macrophage types. IRF3 is dispensable for the LPS-induced IFN-αß overproduction in infected MPI cells and partly dispensable in infected mice, while IRF7 is required. The expression of the LPS co-receptor CD14 is important but not absolutely required for the elicitation of a TNF-α over-response to LPS in Ad-infected mice. CONCLUSION: Viral infections or application of virus-based vaccines induces type I interferon and can tip the balance of the innate immune system in the direction of hyperreactivity to a subsequent exposure to TLR ligands. The adenoviral model presented here is one example of how multiple factors, both environmental and genetic, affect the physiological responses to pathogens. Being able to measure the current reactivity state of the immune system would have important benefits for infection-specific therapies and for the prevention of vaccination-elicited adverse effects.


Asunto(s)
Adenoviridae , Citocinas , Factor 3 Regulador del Interferón , Lipopolisacáridos , Macrófagos , Ratones Noqueados , Animales , Ratones , Lipopolisacáridos/inmunología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Macrófagos/inmunología , Citocinas/metabolismo , Ratones Endogámicos C57BL , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Vectores Genéticos , Infecciones por Adenoviridae/inmunología , Interferón Tipo I/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Células Cultivadas , Células Dendríticas/inmunología , Interferón beta/metabolismo
10.
EMBO Rep ; 12(1): 71-6, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21164516

RESUMEN

Group B streptococcus (GBS) is a leading cause of both neonatal sepsis and meningitis, two diseases that are characterized by inflammation. However, the manner in which GBS organisms are recognized by monocytes and macrophages is poorly understood. In this study, we report that the recognition of GBS and other Gram-positive bacteria by macrophages and monocytes relies on bacterial single-stranded RNA (ssRNA). ssRNA interacts with a signalling complex, which comprises the Toll-like receptor adaptors MyD88 and UNC-93B, but not the established MyD88-dependent ssRNA sensors. The role of ssRNA in the recognition of Gram-positive bacteria--leading to the induction of inflammatory cytokines--has potential implications for sepsis pathogenesis, diagnosis and treatment.


Asunto(s)
Interacciones Huésped-Patógeno , Macrófagos/microbiología , ARN Bacteriano/fisiología , Streptococcus agalactiae/fisiología , Animales , Secuencia de Bases , Citocinas/biosíntesis , Humanos , Recién Nacido , Macrófagos/inmunología , Proteínas de Transporte de Membrana/inmunología , Ratones , Datos de Secuencia Molecular , Factor 88 de Diferenciación Mieloide/inmunología , ARN Bacteriano/inmunología
11.
J Immunol ; 186(9): 5478-88, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21441453

RESUMEN

In macrophages, two signaling pathways, dependent on MyD88 or TIR domain-containing adaptor-inducing IFN-ß (TRIF) signaling, emanate from the LPS receptor TLR4/MD-2. In this study, we show that in murine bone marrow-derived mast cells (BMMCs), only the MyD88-dependent pathway is activated by LPS. The TRIF signaling branch leading both to NF-κB activation and enhanced proinflammatory cytokine production, as well as to IRF3 activation and subsequent IFN-ß production, is absent in LPS-stimulated BMMCs. IRF3 activation is also absent in peritoneal mast cells from LPS-injected mice. We observed strongly diminished TRAM expression in BMMCs, but overexpression of TRAM only moderately enhanced IL-6 and did not boost IFN-ß responses to LPS in these cells. A combination of very low levels of TRAM and TLR4/MD-2 with the known absence of membrane-bound CD14 are expected to contribute to the defective TRIF signaling in mast cells. We also show that, unlike in macrophages, in BMMCs the TRIF-dependent and -independent IFN-αß responses to other recognized IFN inducers (dsRNA, adenovirus, and B-DNA) are absent. These results show how the response to the same microbial ligand using the same receptor can be regulated in different cell types of the innate immune system.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Lipopolisacáridos/inmunología , Mastocitos/inmunología , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Western Blotting , Separación Celular , Inmunoprecipitación de Cromatina , Citocinas/biosíntesis , Citocinas/inmunología , Citometría de Flujo , Antígeno 96 de los Linfocitos , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 4 , Transfección
12.
J Immunol ; 184(10): 5809-18, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20385881

RESUMEN

Src family kinases are involved in a plethora of aspects of cellular signaling. We demonstrate in this study that the Src family kinase Lyn negatively regulates TLR signaling in murine bone marrow-derived macrophages (BMM Phis) and in vivo. LPS-stimulated Lyn(-/-) BMM Phis produced significantly more IL-6, TNF-alpha, and IFN-alpha/beta compared with wild type (WT) BMM Phis, suggesting that Lyn is able to control both MyD88- and TRIF-dependent signaling pathways downstream of TLR4. CD14 was not involved in this type of regulation. Moreover, Lyn attenuated proinflammatory cytokine production in BMM Phis in response to the TLR2 ligand FSL-1, but not to ligands for TLR3 (dsRNA) or TLR9 (CpG 1668). In agreement with these in vitro experiments, Lyn-deficient mice produced higher amounts of proinflammatory cytokines than did WT mice after i. v. injection of LPS or FSL-1. Although Lyn clearly acted as a negative regulator downstream of TLR4 and TLR2, it did not, different from what was proposed previously, prevent the induction of LPS tolerance. Stimulation with a low dose of LPS resulted in reduced production of proinflammatory cytokines after subsequent stimulation with a high dose of LPS in both WT and Lyn(-/-) BMM Phis, as well as in vivo. Mechanistically, Lyn interacted with PI3K; in correlation, PI3K inhibition resulted in increased LPS-triggered cytokine production. In this line, SHIP1(-/-) BMM Phis, exerting enhanced PI3K-pathway activation, produced fewer cytokines than did WT BMM Phis. The data suggest that the Lyn-mediated negative regulation of TLR signaling proceeds, at least in part, via PI3K.


Asunto(s)
Regulación hacia Abajo/inmunología , Activación de Macrófagos/inmunología , Fosfatidilinositol 3-Quinasas/fisiología , Monoéster Fosfórico Hidrolasas/fisiología , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/fisiología , Regulación hacia Arriba/inmunología , Familia-src Quinasas/fisiología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Regulación hacia Abajo/genética , Femenino , Tolerancia Inmunológica/genética , Inositol Polifosfato 5-Fosfatasas , Activación de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/deficiencia , Monoéster Fosfórico Hidrolasas/genética , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 4/antagonistas & inhibidores , Familia-src Quinasas/deficiencia , Familia-src Quinasas/genética
13.
Eur J Immunol ; 40(9): 2506-16, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20690177

RESUMEN

Propionibacterium acnes is a human commensal but also an opportunistic pathogen. In mice, P. acnes exerts strong immunomodulatory activities, including formation of intrahepatic granulomas and induction of LPS hypersensitivity. These activities are dependent on P. acnes recognition via TLR9 and subsequent IL-12-mediated IFN-gamma production. We show that P. acnes elicits IL-12p40 and p35 mRNA expression in macrophages, and IFN-gamma mRNA in liver CD4(+) T cells and NK cells. After priming with P. acnes, CD4(+) T cells serve as the major IFN-gamma mRNA source. In the absence of CD4(+) T cells, CD8(+) T cells (regardless of antigenic specificity) or NK cells can produce sufficient IFN-gamma to induce the P. acnes-driven immune effects. Moreover, in the absence of alpha beta T cells, gamma delta T cells also enable the development of strongly enhanced TNF-alpha and IFN-gamma responses to LPS and intrahepatic granuloma formation. Thus, under microbial pressure, different T-cell types, independent of their antigen specificity, exert NK-cell-like functions, which contribute decisively to the activation of the innate immune system.


Asunto(s)
Citocinas/biosíntesis , Células Asesinas Naturales/inmunología , Hígado/inmunología , Propionibacterium acnes/inmunología , Linfocitos T/inmunología , Animales , Antígenos Bacterianos/inmunología , Citocinas/genética , Granuloma/patología , Humanos , Inmunidad Innata , Inmunización , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/microbiología , Células Asesinas Naturales/patología , Hígado/microbiología , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones Oportunistas/inmunología , Infecciones Oportunistas/microbiología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/metabolismo , Linfocitos T/microbiología , Linfocitos T/patología
14.
Proc Natl Acad Sci U S A ; 105(13): 5177-82, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18362329

RESUMEN

The giant cytosolic protease tripeptidyl peptidase II (TPPII) has been implicated in the regulation of proliferation and survival of malignant cells, particularly lymphoma cells. To address its functions in normal cellular and systemic physiology we have generated TPPII-deficient mice. TPPII deficiency activates cell type-specific death programs, including proliferative apoptosis in several T lineage subsets and premature cellular senescence in fibroblasts and CD8(+) T cells. This coincides with up-regulation of p53 and dysregulation of NF-kappaB. Prominent degenerative alterations at the organismic level were a decreased lifespan and symptoms characteristic of immunohematopoietic senescence. These symptoms include accelerated thymic involution, lymphopenia, impaired proliferative T cell responses, extramedullary hematopoiesis, and inflammation. Thus, TPPII is important for maintaining normal cellular and systemic physiology, which may be relevant for potential therapeutic applications of TPPII inhibitors.


Asunto(s)
Envejecimiento/inmunología , Apoptosis/inmunología , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/metabolismo , Aminopeptidasas , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Fibroblastos , Eliminación de Gen , Linfopenia/enzimología , Linfopenia/genética , Linfopenia/patología , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Fenotipo , Serina Endopeptidasas/genética , Linfocitos T/citología , Linfocitos T/enzimología , Linfocitos T/inmunología , Timo/citología , Timo/enzimología , Timo/inmunología , Proteína p53 Supresora de Tumor/metabolismo
15.
Gut ; 59(8): 1079-87, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20639251

RESUMEN

BACKGROUND: The bacterial microflora aggravates graft-versus-host-disease (GvHD) after allogeneic stem cell transplantation, but the underlying mechanisms of manifestations of intestinal GvHD (iGvHD) in the gut remain poorly understood. AIM: To analyse the gut flora composition and the impact of bacterial sensing via Toll-like receptors (TLRs) in iGvHD. METHODS: By mimicking clinical low-intensity conditioning regimens used in humans, a novel irradiation independent, treosulfan and cyclophosphamide-based murine allogeneic transplantation model was established. A global survey of the intestinal microflora by cultural and molecular methods was performed, the intestinal immunopathology in TLR-deficient recipient mice with iGvHD investigated and finally, the impact of anti-TLR9 treatment on iGvHD development assessed. RESULTS: The inflammatory responses in iGvHD were accompanied by gut flora shifts towards enterobacteria, enterococci and Bacteroides/Prevotella spp. Analysis of iGvHD in MyD88(-/-), TRIF(-/-), TLR2/4(-/-), and TLR9(-/-) recipient mice showed that bacterial sensing via TLRs was essential for iGvHD development. Acute iGvHD was characterised by increasing numbers of apoptotic cells, proliferating cells, T cells and neutrophils within the colon. These responses were significantly reduced in MyD88(-/-), TLR2/4(-/-), TRIF(-/-) and TLR9(-/-) mice, as compared with wild-type controls. However, TRIF(-/-) and TLR2/4(-/-) mice were not protected from mortality, whereas TLR9(-/-) mice displayed increased survival rates. The important role of TLR9-mediated immunopathology was independently confirmed by significantly reduced macroscopic disease symptoms and colonic apoptosis as well as by reduced T-cell and neutrophil numbers within the colon after treatment with a synthetic inhibitory oligonucleotide. CONCLUSIONS: These results emphasise the critical role of gut microbiota, innate immunity and TLR9 in iGvHD and highlight anti-TLR9 strategies as novel therapeutic options.


Asunto(s)
Colitis/microbiología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/microbiología , Factor 88 de Diferenciación Mieloide/inmunología , Receptor Toll-Like 9/inmunología , Enfermedad Aguda , Animales , Apoptosis/inmunología , Trasplante de Médula Ósea , Proliferación Celular , Colitis/inmunología , Colitis/patología , Colitis/prevención & control , Modelos Animales de Enfermedad , Femenino , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/prevención & control , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Oligonucleótidos Antisentido/uso terapéutico , Bazo/trasplante , Receptor Toll-Like 9/deficiencia , Acondicionamiento Pretrasplante
16.
J Exp Med ; 195(1): 99-111, 2002 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-11781369

RESUMEN

Low molecular weight fragmentation products of the polysaccharide of Hyaluronic acid (sHA) produced during inflammation have been shown to be potent activators of immunocompetent cells such as dendritic cells (DCs) and macrophages. Here we report that sHA induces maturation of DCs via the Toll-like receptor (TLR)-4, a receptor complex associated with innate immunity and host defense against bacterial infection. Bone marrow-derived DCs from C3H/HeJ and C57BL/10ScCr mice carrying mutant TLR-4 alleles were nonresponsive to sHA-induced phenotypic and functional maturation. Conversely, DCs from TLR-2-deficient mice were still susceptible to sHA. In accordance, addition of an anti-TLR-4 mAb to human monocyte-derived DCs blocked sHA-induced tumor necrosis factor alpha production. Western blot analysis revealed that sHA treatment resulted in distinct phosphorylation of p38/p42/44 MAP-kinases and nuclear translocation of nuclear factor (NF)-kappa B, all components of the TLR-4 signaling pathway. Blockade of this pathway by specific inhibitors completely abrogated the sHA-induced DC maturation. Finally, intravenous injection of sHA-induced DC emigration from the skin and their phenotypic and functional maturation in the spleen, again depending on the expression of TLR-4. In conclusion, this is the first report that polysaccharide degradation products of the extracellular matrix produced during inflammation might serve as an endogenous ligand for the TLR-4 complex on DCs.


Asunto(s)
Células Dendríticas/inmunología , Proteínas de Drosophila , Ácido Hialurónico/inmunología , Glicoproteínas de Membrana/metabolismo , Oligosacáridos/inmunología , Receptores de Superficie Celular/metabolismo , Animales , Diferenciación Celular , Regulación hacia Abajo , Femenino , Humanos , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Receptores de Superficie Celular/deficiencia , Transducción de Señal , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like , Factor de Necrosis Tumoral alfa/metabolismo
17.
J Virol ; 83(18): 9411-22, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19570869

RESUMEN

Parapoxvirus ovis (PPVO) is a member of the Poxviridae family and belongs to the genus Parapoxvirus. It displays only limited homology with orthopoxviruses and has some molecular features such as an unusual high GC content distinct from orthopoxviruses. Inactivated PPVO (iPPVO) displays strong immunostimulatory capacities mediating antiviral activity in vivo. The role of dendritic cells (DC) and the pattern recognition receptors and signaling requirements responsible for immunostimulation by iPPVO are unknown. We demonstrate here that bone marrow-derived plasmacytoid DC (BM-pDC) and bone marrow-derived conventional DC (BM-cDC) secrete alpha/beta interferon (IFN-alpha/beta) in response to iPPVO. Furthermore, iPPVO induces tumor necrosis factor alpha (TNF-alpha) and interleukin-12/23p40 (IL-12/23p40) release and major histocompatibility complex class II (MHC-II), MHC-I, and CD86 upregulation by bone marrow-derived DC (BMDC). After engulfment, iPPVO is located in endosomal compartments and in the cytosol of BMDC. iPPVO elicits IFN-alpha/beta by Toll-like receptor (TLR)-independent pathways in BM-cDC, since IFN-alpha/beta release does not require myeloid differentiation primary response gene 88 (MyD88) or TIR-domain containing adaptor protein inducing interferon (TRIF). In contrast, iPPVO-induced TNF-alpha release and enhanced expression of MHC-I and CD86 but not of MHC-II by BMDC chiefly requires MyD88 but not TLR2 or TLR4. Induction of IFN-alpha by iPPVO in BM-cDC occurred in the absence of IFN regulatory factor 3 (IRF3) but required the presence of IRF7, whereas iPPVO-triggered IFN-beta production required the presence of either IRF7 or IRF3. These results provide the first evidence that iPPVO mediates its immunostimulatory properties by TLR-independent and TLR-dependent pathways and demonstrate an important role of cDC for IFN-alpha/beta production.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Parapoxvirus/inmunología , Receptores Toll-Like/metabolismo , Animales , Células de la Médula Ósea , Ratones , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Inactivación de Virus
18.
PLoS Pathog ; 4(11): e1000208, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19008951

RESUMEN

The early systemic production of interferon (IFN)-alphabeta is an essential component of the antiviral host defense mechanisms, but is also thought to contribute to the toxic side effects accompanying gene therapy with adenoviral vectors. Here we investigated the IFN-alphabeta response to human adenoviruses (Ads) in mice. By comparing the responses of normal, myeloid (m)DC- and plasmacytoid (p)DC-depleted mice and by measuring IFN-alphabeta mRNA expression in different organs and cells types, we show that in vivo, Ads elicit strong and rapid IFN-alphabeta production, almost exclusively in splenic mDCs. Using knockout mice, various strains of Ads (wild type, mutant and UV-inactivated) and MAP kinase inhibitors, we demonstrate that the Ad-induced IFN-alphabeta response does not require Toll-like receptors (TLR), known cytosolic sensors of RNA (RIG-I/MDA-5) and DNA (DAI) recognition and interferon regulatory factor (IRF)-3, but is dependent on viral endosomal escape, signaling via the MAP kinase SAPK/JNK and IRF-7. Furthermore, we show that Ads induce IFN-alphabeta and IL-6 in vivo by distinct pathways and confirm that IFN-alphabeta positively regulates the IL-6 response. Finally, by measuring TNF-alpha responses to LPS in Ad-infected wild type and IFN-alphabetaR(-/-) mice, we show that IFN-alphabeta is the key mediator of Ad-induced hypersensitivity to LPS. These findings indicate that, like endosomal TLR signaling in pDCs, TLR-independent virus recognition in splenic mDCs can also produce a robust early IFN-alphabeta response, which is responsible for the bulk of IFN-alphabeta production induced by adenovirus in vivo. The signaling requirements are different from known TLR-dependent or cytosolic IFN-alphabeta induction mechanisms and suggest a novel cytosolic viral induction pathway. The hypersensitivity to components of the microbial flora and invading pathogens may in part explain the toxic side effects of adenoviral gene therapy and contribute to the pathogenesis of adenoviral disease.


Asunto(s)
Adenoviridae/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Bazo/citología , Animales , Endosomas , Humanos , Interferón Tipo I/genética , Interleucina-6/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos , Proteínas Quinasas Activadas por Mitógenos , ARN Mensajero/análisis , Transducción de Señal , Bazo/inmunología , Distribución Tisular , Regulación hacia Arriba
19.
Microbes Infect ; 10(12-13): 1259-65, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18692153

RESUMEN

Plasmodium falciparum GPI contributes to malaria pathology by inducing cytokine release. It has been shown to be recognized through TLR2 and to a lesser extent TLR4 in vitro. However, previous findings on the role of TLRs in parasite clearance or pathology in vivo are conflicting. Thus, we analyzed the impact of TLR-signalling on protection using the P. yoelii infection model. Deficiency of single TLRs as well as triple TLR2/4/9-deficiency had no impact on parasitaemia. In contrast, mice deficient for the adaptor protein MyD88 were more susceptible to P. yoelii infection in that they exhibited an increased parasitaemia in the early phase of the infection and a higher lethality. This phenotype was caused mainly by impaired IL-18 signalling since parasitaemia in IL-18-deficient mice was also increased at early time points during P. yoelii infection compared to wild-type control mice. However, no lethality was observed in IL-18-deficient mice. Since parasitaemia in IL-1R-deficient mice was also slightly increased during P. yoelii infection, impaired IL-1R signalling contributed to the increased susceptibility of MyD88-deficient mice to a lesser extent. These findings correlated with a reduced IFN-gamma production in MyD88- and IL-18-deficient mice, but not in TLR2/4/9-deficient mice. We conclude that mainly IL-18/MyD88-dependent signalling but not TLR2/4/9-signalling is important for early parasite control in our model.


Asunto(s)
Interleucina-18/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Plasmodium yoelii/patogenicidad , Transducción de Señal , Animales , Femenino , Interleucina-18/genética , Malaria/inmunología , Malaria/parasitología , Malaria/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/genética , Parasitemia/inmunología , Parasitemia/parasitología , Parasitemia/patología , Receptores Toll-Like/deficiencia , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
20.
Immunobiology ; 213(3-4): 193-203, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18406367

RESUMEN

In this review, we summarize our investigations concerning the differential importance of CD14 and LBP in toll-like receptor 4 (TLR4)/myeloid differentiation protein-2 (MD-2)-mediated signaling by smooth and rough-form lipopolysaccharide (LPS) chemotypes and include the results obtained in studies with murine and human TLR4-transgenic mice. Furthermore, we present more recent data on the mechanisms involved in the induction of LPS hypersensitivity by bacterial and viral infections and on the reactivity of the hypersensitive host to non-LPS microbial ligands and endogenous mediators. Finally, the effects of pre-existing hypersensitivity on the course and outcome of a super-infection with Salmonella typhimurium or Listeria monocytogenes are summarized.


Asunto(s)
Infecciones por Bacterias Gramnegativas/metabolismo , Lipopolisacáridos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Humanos , Sistema Inmunológico , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Antígeno 96 de los Linfocitos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Transducción de Señal , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA