Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 8(27): eabq0084, 2022 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-35857476

RESUMEN

Toll-like receptors (TLRs) recognize pathogen- and host-derived factors and control immune responses via the adaptor protein MyD88 and members of the interferon regulatory transcription factor (IRF) family. IRFs orchestrate key effector functions, including cytokine release, cell differentiation, and, under certain circumstances, inflammation pathology. Here, we show that IRF activity is generically controlled by the Src kinase family member LYN, which phosphorylates all TLR-induced IRFs at a conserved tyrosine residue, resulting in K48-linked polyubiquitination and proteasomal degradation of IRFs. We further show that LYN activity is controlled by the upstream kinase C-terminal Src kinase (CSK), whose activity, in turn, is controlled by the adaptor protein SPOP, which serves as molecular bridge to recruit CSK into the TLR signaling complex and to activate CSK catalytic activity. Consistently, deletion of SPOP or CSK results in increased LYN activity, LYN-directed IRF degradation, and inhibition of IRF transcriptional activity. Together, the data reveal a key regulatory mechanism for IRF family members controlling TLR biology.

2.
Sci Rep ; 11(1): 24507, 2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-34969960

RESUMEN

Diagnostic tests that detect antibodies (AB) against SARS-CoV-2 for evaluation of seroprevalence and guidance of health care measures are important tools for managing the COVID-19 pandemic. Current tests have certain limitations with regard to turnaround time, costs and availability, particularly in point-of-care (POC) settings. We established a hemagglutination-based AB test that is based on bi-specific proteins which contain a dromedary-derived antibody (nanobody) binding red blood cells (RBD) and a SARS-CoV-2-derived antigen, such as the receptor-binding domain of the Spike protein (Spike-RBD). While the nanobody mediates swift binding to RBC, the antigen moiety directs instantaneous, visually apparent hemagglutination in the presence of SARS-CoV-2-specific AB generated in COVID-19 patients or vaccinated individuals. Method comparison studies with assays cleared by emergency use authorization demonstrate high specificity and sensitivity. To further increase objectivity of test interpretation, we developed an image analysis tool based on digital image acquisition (via a cell phone) and a machine learning algorithm based on defined sample-training and -validation datasets. Preliminary data, including a small clinical study, provides proof of principle for test performance in a POC setting. Together, the data support the interpretation that this AB test format, which we refer to as 'NanoSpot.ai', is suitable for POC testing, can be manufactured at very low costs and, based on its generic mode of action, can likely be adapted to a variety of other pathogens.


Asunto(s)
Anticuerpos Antivirales/sangre , Prueba Serológica para COVID-19/métodos , Pruebas Diagnósticas de Rutina/métodos , Pruebas de Hemaglutinación/métodos , Pruebas en el Punto de Atención , Glicoproteína de la Espiga del Coronavirus/inmunología , Humanos , Prueba de Estudio Conceptual
3.
Res Sq ; 2021 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-34312614

RESUMEN

Diagnostic tests that detect antibodies (AB) against SARS-CoV-2 for evaluation of seroprevalence and guidance of health care measures are important tools for managing the COVID-19 pandemic. Current tests have certain limitations with regard to turnaround time, costs and availability, particularly in point-of-care (POC) settings. We established a hemagglutination-based AB test (HAT) that is based on bi-specific proteins which contain a dromedary-derived antibody (nanobody) binding red blood cells (RBD) and a SARS-CoV-2-derived antigen, such as the receptor-binding domain of the Spike protein (Spike-RBD). While the nanobody mediates swift binding to RBC, the antigen moiety directs instantaneous, visually apparent hemagglutination in the presence of SARS-CoV-2-specific AB generated in COVID-19 patients or vaccinated individuals. Method comparison studies with assays cleared by emergency use authorization (EUA) demonstrate high specificity and sensitivity. To further increase objectivity of test interpretation, we developed an image analysis tool based on digital image acquisition (via a cell phone) and a machine learning algorithm based on defined sample-training and -validation datasets. Preliminary data, including a small clinical study, provides proof of principle for test performance in a POC setting. Together, the data support the interpretation that this AB test format, which we refer to as 'NanoSpot.ai', is suitable for POC testing, can be manufactured at very low costs and, based on its generic mode of action, can likely be adapted to a variety of other pathogens.

4.
J Clin Invest ; 129(6): 2251-2265, 2019 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-31033479

RESUMEN

Systemic lupus erythematosus (SLE) is a complex autoimmune disease with genetic and environmental contributions. Hallmarks of the disease are the appearance of immune complexes (IC) containing autoreactive Abs and TLR-activating nucleic acids, whose deposition in kidney glomeruli is suspected to promote tissue injury and glomerulonephritis (GN). Here, using a mouse model based on the human SLE susceptibility locus TNFAIP3-interacting protein 1 (TNIP1, also known as ABIN1), we investigated the pathogenesis of GN. We found that GN was driven by TLRs but, remarkably, proceeded independently of ICs. Rather, disease in 3 different mouse models and patients with SLE was characterized by glomerular accumulation of patrolling monocytes (PMos), a cell type with an emerging key function in vascular inflammation. Consistent with such function in GN, monocyte-specific deletion of ABIN1 promoted kidney disease, whereas selective elimination of PMos provided protection. In contrast to GN, PMo elimination did not protect from reduced survival or disease symptoms such as IC generation and splenomegaly, suggesting that GN and other inflammatory processes are governed by distinct pathogenic mechanisms. These data identify TLR-activated PMos as the principal component of an intravascular process that contributes to glomerular inflammation and kidney injury.


Asunto(s)
Nefritis Lúpica/inmunología , Monocitos/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Nefritis Lúpica/genética , Nefritis Lúpica/patología , Ratones , Ratones Noqueados , Monocitos/patología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
5.
ChemMedChem ; 13(20): 2208-2216, 2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30117269

RESUMEN

The immune system uses members of the toll-like receptor (TLR) family to recognize a variety of pathogen- and host-derived molecules in order to initiate immune responses. Although TLR-mediated, pro-inflammatory immune responses are essential for host defense, prolonged and exaggerated activation can result in inflammation pathology that manifests in a variety of diseases. Therefore, small-molecule inhibitors of the TLR signaling pathway might have promise as anti-inflammatory drugs. We previously identified a class of triaryl pyrazole compounds that inhibit TLR signaling by modulation of the protein-protein interactions essential to the pathway. We have now systematically examined the structural features essential for inhibition of this pathway, revealing characteristics of compounds that inhibited all TLRs tested (pan-TLR signaling inhibitors) as well as compounds that selectively inhibited certain TLRs. These findings reveal interesting classes of compounds that could be optimized for particular inflammatory diseases governed by different TLRs.


Asunto(s)
Pirazoles/química , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/metabolismo , Animales , Humanos , Ratones , Estructura Molecular , Células RAW 264.7 , Receptores de Estrógenos/metabolismo , Relación Estructura-Actividad
6.
Sci Signal ; 11(543)2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30108181

RESUMEN

Toll-like receptors (TLRs) recognize various pathogen- and host tissue-derived molecules and initiate inflammatory immune responses. Exaggerated or prolonged TLR activation, however, can lead to etiologically diverse diseases, such as bacterial sepsis, metabolic and autoimmune diseases, or stroke. Despite the apparent medical need, no small-molecule drugs against TLR pathways are clinically available. This may be because of the complex signaling mechanisms of TLRs, which are governed by a series of protein-protein interactions initiated by Toll/interleukin-1 receptor homology domains (TIR) found in TLRs and the cytoplasmic adaptor proteins TIRAP and MyD88. Oligomerization of TLRs with MyD88 or TIRAP leads to the recruitment of members of the IRAK family of kinases and the E3 ubiquitin ligase TRAF6. We developed a phenotypic drug screening system based on the inducible homodimerization of either TIRAP, MyD88, or TRAF6, that ranked hits according to their hierarchy of action. From a bioactive compound library, we identified methyl-piperidino-pyrazole (MPP) as a TLR-specific inhibitor. Structure-activity relationship analysis, quantitative proteomics, protein-protein interaction assays, and cellular thermal shift assays suggested that MPP targets the TIR domain of MyD88. Chemical evolution of the original MPP scaffold generated compounds with selectivity for distinct TLRs that interfered with specific TIR interactions. Administration of an MPP analog to mice protected them from TLR4-dependent inflammation. These results validate this phenotypic screening approach and suggest that the MPP scaffold could serve as a starting point for the development of anti-inflammatory drugs.


Asunto(s)
Piperidinas/farmacología , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Receptores Toll-Like/antagonistas & inhibidores , Animales , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Células HEK293 , Humanos , Concentración 50 Inhibidora , Ratones , Unión Proteica/efectos de los fármacos , Mapas de Interacción de Proteínas/efectos de los fármacos , Células RAW 264.7 , Receptores Toll-Like/metabolismo
7.
Proc Natl Acad Sci U S A ; 113(41): E6162-E6171, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27671649

RESUMEN

Psoriasis is a chronic inflammatory skin disease with a clear genetic contribution, characterized by keratinocyte proliferation and immune cell infiltration. Various closely interacting cell types, including innate immune cells, T cells, and keratinocytes, are known to contribute to inflammation. Innate immune cells most likely initiate the inflammatory process by secretion of IL-23. IL-23 mediates expansion of T helper 17 (Th17) cells, whose effector functions, including IL-17A, activate keratinocytes. Keratinocyte activation in turn results in cell proliferation and chemokine expression, the latter of which fuels the inflammatory process through further immune cell recruitment. One question that remains largely unanswered is how genetic susceptibility contributes to this process and, specifically, which cell type causes disease due to psoriasis-specific genetic alterations. Here we describe a mouse model based on the human psoriasis susceptibility locus TNIP1, also referred to as ABIN1, whose gene product is a negative regulator of various inflammatory signaling pathways, including the Toll-like receptor pathway in innate immune cells. We find that Tnip1-deficient mice recapitulate major features of psoriasis on pathological, genomic, and therapeutic levels. Different genetic approaches, including tissue-specific gene deletion and the use of various inflammatory triggers, reveal that Tnip1 controls not only immune cells, but also keratinocyte biology. Loss of Tnip1 in keratinocytes leads to deregulation of IL-17-induced gene expression and exaggerated chemokine production in vitro and overt psoriasis-like inflammation in vivo. Together, the data establish Tnip1 as a critical regulator of IL-17 biology and reveal a causal role of keratinocytes in the pathogenesis of psoriasis.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Susceptibilidad a Enfermedades , Queratinocitos/metabolismo , Psoriasis/etiología , Psoriasis/metabolismo , Aminoquinolinas/efectos adversos , Animales , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Imiquimod , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Ratones , Ratones Noqueados , Psoriasis/patología , Transcriptoma
8.
Immunology ; 148(2): 216-26, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26931177

RESUMEN

SHARPIN is an essential component of the linear ubiquitin chain assembly complex (LUBAC) complex that controls signalling pathways of various receptors, including the tumour necrosis factor receptor (TNFR), Toll-like receptor (TLR) and antigen receptor, in part by synthesis of linear, non-degrading ubiquitin chains. Consistent with SHARPIN's function in different receptor pathways, the phenotype of SHARPIN-deficient mice is complex, including the development of inflammatory systemic and skin diseases, the latter of which depend on TNFR signal transduction. Given the established function of SHARPIN in primary and malignant B cells, we hypothesized that SHARPIN might also regulate T-cell receptor (TCR) signalling and thereby control T-cell biology. Here, we focus primarily on the role of SHARPIN in T cells, specifically regulatory T (Treg) cells. We found that SHARPIN-deficient (Sharpin(cpdm/cpdm) ) mice have significantly reduced numbers of FOXP3(+) Treg cells in lymphoid organs and the peripheral blood. Competitive reconstitution of irradiated mice with mixed bone marrow from wild-type and SHARPIN-deficient mice revealed an overall reduced thymus population with SHARPIN-deficient cells with almost complete loss of thymic Treg development. Consistent with this cell-intrinsic function of SHARPIN in Treg development, TCR stimulation of SHARPIN-deficient thymocytes revealed reduced activation of nuclear factor-κB and c-Jun N-terminal kinase, establishing a function of SHARPIN in TCR signalling, which may explain the defective Treg development. In turn, in vitro generation and suppressive activity of mature SHARPIN-deficient Treg cells were comparable to wild-type cells, suggesting that maturation, but not function, of SHARPIN-deficient Treg cells is impaired. Taken together, these findings show that SHARPIN controls TCR signalling and is required for efficient generation of Treg cells in vivo, whereas the inhibitory function of mature Treg cells appears to be independent of SHARPIN.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Linfocitos T Reguladores/fisiología , Timocitos/fisiología , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Factores de Transcripción Forkhead/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/genética , Transducción de Señal/genética
9.
J Virol ; 88(16): 9038-48, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899174

RESUMEN

UNLABELLED: Influenza A virus (IAV) replication depends on the interaction of virus proteins with host factors. The viral nonstructural protein 1 (NS1) is essential in this process by targeting diverse cellular functions, including mRNA splicing and translation, cell survival, and immune defense, in particular the type I interferon (IFN-I) response. In order to identify host proteins targeted by NS1, we established a replication-competent recombinant IAV that expresses epitope-tagged forms of NS1 and NS2, which are encoded by the same gene segment, allowing purification of NS proteins during natural cell infection and analysis of interacting proteins by quantitative mass spectrometry. We identified known NS1- and NS2-interacting proteins but also uncharacterized proteins, including PACT, an important cofactor for the IFN-I response triggered by the viral RNA-sensor RIG-I. We show here that NS1 binds PACT during virus replication and blocks PACT/RIG-I-mediated activation of IFN-I, which represents a critical event for the host defense. Protein interaction and interference with IFN-I activation depended on the functional integrity of the highly conserved RNA binding domain of NS1. A mutant virus with deletion of NS1 induced high levels of IFN-I in control cells, as expected; in contrast, shRNA-mediated knockdown of PACT compromised IFN-I activation by the mutant virus, but not wild-type virus, a finding consistent with the interpretation that PACT (i) is essential for IAV recognition and (ii) is functionally compromised by NS1. Together, our data describe a novel approach to identify virus-host protein interactions and demonstrate that NS1 interferes with PACT, whose function is critical for robust IFN-I production. IMPORTANCE: Influenza A virus (IAV) is an important human pathogen that is responsible for annual epidemics and occasional devastating pandemics. Viral replication and pathogenicity depends on the interference of viral factors with components of the host defense system, particularly the type I interferon (IFN-I) response. The viral NS1 protein is known to counteract virus recognition and IFN-I production, but the molecular mechanism is only partially defined. We used a novel proteomic approach to identify host proteins that are bound by NS1 during virus replication and identified the protein PACT, which had previously been shown to be involved in virus-mediated IFN-I activation. We find that NS1 prevents PACT from interacting with an essential component of the virus recognition pathway, RIG-I, thereby disabling efficient IFN-I production. These observations provide an important piece of information on how IAV efficiently counteracts the host immune defense.


Asunto(s)
Antivirales/metabolismo , Virus de la Influenza A/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Perros , Células HEK293 , Humanos , Virus de la Influenza A/genética , Interferón beta/genética , Interferón beta/metabolismo , Células de Riñón Canino Madin Darby , Regiones Promotoras Genéticas/genética , Proteómica/métodos , ARN Viral/genética , Proteínas de Unión al ARN/genética , Receptores Inmunológicos , Proteínas no Estructurales Virales/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/genética
10.
PLoS Pathog ; 9(11): e1003727, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244159

RESUMEN

Streptococcus pneumoniae infection is a leading cause of bacterial pneumonia, sepsis and meningitis and is associated with high morbidity and mortality. Type I interferon (IFN-I), whose contribution to antiviral and intracellular bacterial immunity is well established, is also elicited during pneumococcal infection, yet its functional significance is not well defined. Here, we show that IFN-I plays an important role in the host defense against pneumococci by counteracting the transmigration of bacteria from the lung to the blood. Mice that lack the type I interferon receptor (Ifnar1 (-/-)) or mice that were treated with a neutralizing antibody against the type I interferon receptor, exhibited enhanced development of bacteremia following intranasal pneumococcal infection, while maintaining comparable bacterial numbers in the lung. In turn, treatment of mice with IFNß or IFN-I-inducing synthetic double stranded RNA (poly(I:C)), dramatically reduced the development of bacteremia following intranasal infection with S. pneumoniae. IFNß treatment led to upregulation of tight junction proteins and downregulation of the pneumococcal uptake receptor, platelet activating factor receptor (PAF receptor). In accordance with these findings, IFN-I reduced pneumococcal cell invasion and transmigration across epithelial and endothelial layers, and Ifnar1 (-/-) mice showed overall enhanced lung permeability. As such, our data identify IFN-I as an important component of the host immune defense that regulates two possible mechanisms involved in pneumococcal invasion, i.e. PAF receptor-mediated transcytosis and tight junction-dependent pericellular migration, ultimately limiting progression from a site-restricted lung infection to invasive, lethal disease.


Asunto(s)
Barrera Alveolocapilar/metabolismo , Barrera Hematoencefálica/metabolismo , Interferón Tipo I/metabolismo , Neumonía Neumocócica/metabolismo , Streptococcus pneumoniae/patogenicidad , Animales , Bacteriemia/genética , Bacteriemia/metabolismo , Barrera Alveolocapilar/microbiología , Barrera Alveolocapilar/patología , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/patología , Interferón Tipo I/genética , Ratones , Ratones Noqueados , Neumonía Neumocócica/genética , Neumonía Neumocócica/patología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Streptococcus pneumoniae/metabolismo
11.
Nat Methods ; 10(8): 795-803, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23749299

RESUMEN

Investigation of immune-cell differentiation and function is limited by shortcomings of suitable and scalable experimental systems. Here we show that retroviral delivery of an estrogen-regulated form of Hoxb8 into mouse bone marrow cells can be used along with Flt3 ligand to conditionally immortalize early hematopoietic progenitor cells (Hoxb8-FL cells). Hoxb8-FL cells have lost self-renewal capacity and potential to differentiate into megakaryocytes and erythrocytes but retain the potential to differentiate into myeloid and lymphoid cells. They differentiate in vitro and in vivo into macrophages, granulocytes, dendritic cells, B lymphocytes and T lymphocytes that are phenotypically and functionally indistinguishable from their primary counterparts. Quantitative in vitro assays indicate that myeloid and B-cell potential of Hoxb8-FL cells is comparable to that of primary lymphoid-primed multipotent progenitors, whereas T-cell potential is diminished. The simplicity of this system and the unlimited proliferative capacity of Hoxb8-FL cells will enable studies of immune-cell differentiation and function.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/metabolismo , Linfocitos/citología , Células Mieloides/citología , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Femenino , Citometría de Flujo , Linfocitos/ultraestructura , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/ultraestructura , Análisis de Componente Principal , Proteómica
12.
J Immunol ; 188(9): 4602-10, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22474019

RESUMEN

Immune cell-mediated tissue injury is a common feature of different inflammatory diseases, yet the pathogenetic mechanisms and cell types involved vary significantly. Hypereosinophilic syndrome (HES) represents a group of inflammatory diseases that is characterized by increased numbers of pathogenic eosinophilic granulocytes in the peripheral blood and diverse organs. On the basis of clinical and laboratory findings, various forms of HES have been defined, yet the molecular mechanism and potential signaling pathways that drive eosinophil expansion remain largely unknown. In this study, we show that mice deficient of the serine/threonine-specific protein kinase NF-κB-inducing kinase (NIK) develop a HES-like disease, reflected by progressive blood and tissue eosinophilia, tissue injury, and premature death at around 25-30 wk of age. Similar to the lymphocytic form of HES, CD4(+) T cells from NIK-deficient mice express increased levels of Th2-associated cytokines, and eosinophilia and survival of NIK-deficient mice could be prevented completely by genetic ablation of CD4(+) T cells. Experiments based on bone marrow chimeric mice, however, demonstrated that inflammation in NIK-deficient mice depended on radiation-resistant tissues, implicating that NIK-deficient immune cells mediate inflammation in a nonautonomous manner. Surprisingly, disease development was independent of NIK's known function as an IκB kinase α (IKKα) kinase, because mice carrying a mutation in the activation loop of IKKα, which is phosphorylated by NIK, did not develop inflammatory disease. Our data show that NIK activity in nonhematopoietic cells controls Th2 cell development and prevents eosinophil-driven inflammatory disease, most likely using a signaling pathway that operates independent of the known NIK substrate IKKα.


Asunto(s)
Síndrome Hipereosinofílico/inmunología , Quinasa I-kappa B/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Animales , Activación Enzimática/genética , Activación Enzimática/inmunología , Síndrome Hipereosinofílico/genética , Síndrome Hipereosinofílico/metabolismo , Síndrome Hipereosinofílico/patología , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Noqueados , Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Secundaria de Proteína , Células Th2/inmunología , Células Th2/metabolismo , Células Th2/patología , Quinasa de Factor Nuclear kappa B
13.
Proc Natl Acad Sci U S A ; 108(44): E998-1006, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22011580

RESUMEN

Toll-like receptors (TLRs) are expressed on innate immune cells and trigger inflammation upon detection of pathogens and host tissue injury. TLR-mediated proinflammatory-signaling pathways are counteracted by partially characterized anti-inflammatory mechanisms that prevent exaggerated inflammation and host tissue damage as manifested in inflammatory diseases. We biochemically identified a component of TLR-signaling pathways, A20-binding inhibitor of NF-κB (ABIN1), which recently has been linked by genome-wide association studies to the inflammatory diseases systemic lupus erythematosus and psoriasis. We generated ABIN1-deficient mice to study the function of ABIN1 in vivo and during TLR activation. Here we show that ABIN1-deficient mice develop a progressive, lupus-like inflammatory disease characterized by expansion of myeloid cells, leukocyte infiltrations in different parenchymatous organs, activated T and B lymphocytes, elevated serum Ig levels, and the appearance of autoreactive antibodies. Kidneys develop glomerulonephritis and proteinuria, reflecting tissue injury. Surprisingly, ABIN1-deficient macrophages exhibit normal regulation of major proinflammatory signaling pathways and mediators but show selective deregulation of the transcription factor CCAAT/enhancer binding protein ß (C/EBPß) and its target genes, such as colony-stimulating factor 3 (Csf3), nitric oxide synthase, inducible (Nos2), and S100 calcium-binding protein A8 (S100a8). Their gene products, which are intimately linked to innate immune cell expansion (granulocyte colony-stimulating factor), cytotoxicity (inducible nitric oxide synthase), and host factor-derived inflammation (S100A8), may explain, at least in part, the inflammatory phenotype observed. Together, our data reveal ABIN1 as an essential anti-inflammatory component of TLR-signaling pathways that controls C/EBPß activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Lupus Eritematoso Sistémico/prevención & control , FN-kappa B/antagonistas & inhibidores , Psoriasis/prevención & control , Receptores Toll-Like/fisiología , Animales , Secuencia de Bases , Células de la Médula Ósea/fisiología , Cartilla de ADN , Muerte Fetal , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
14.
J Biol Chem ; 286(43): 37147-57, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21896478

RESUMEN

B-cell CLL/lymphoma 10 (BCL10) is crucial for the activation of NF-κB in numerous immune receptor signaling pathways, including the T-cell receptor (TCR) and B-cell receptor signaling pathways. However, the molecular mechanisms that lead to signal transduction from BCL10 to downstream NF-κB effector kinases, such as TAK1 and components of the IKK complex, are not entirely understood. Here we used a proteomic approach and identified the E3 ligase MIB2 as a novel component of the activated BCL10 complex. In vitro translation and pulldown assays suggest direct interaction between BCL10 and MIB2. Overexpression experiments show that MIB2 controls BCL10-mediated activation of NF-κB by promoting autoubiquitination and ubiquitination of IKKγ/NEMO, as well as recruitment and activation of TAK1. Knockdown of MIB2 inhibited BCL10-dependent NF-κB activation. Together, our results identify MIB2 as a novel component of the activated BCL10 signaling complex and a missing link in the BCL10-dependent NF-κB signaling pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína 10 de la LLC-Linfoma de Células B , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Jurkat , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , FN-kappa B/genética , Proteómica , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/fisiología
15.
Infect Immun ; 78(12): 5287-94, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20855509

RESUMEN

Infections caused by Streptococcus pneumoniae are major causes of morbidity and mortality, which are in part mediated by immune cell-dependent mechanisms. Yet, the specific contributions of individual cell types to immunopathology are only partially understood. T cells are well characterized with respect to their function in protective humoral immune responses; however, their roles during early stages of infection and invasive pneumococcal disease (IPD) are less well defined. Using a mouse model of pneumococcal sepsis, we found that CD4(+) T cells were recruited to the lung as early as 12 h after intranasal infection. Recruitment was accompanied by upregulation of CD69 and B7-H1, reflecting T-cell activation. Unexpectedly, major histocompatibility complex (MHC) class II-deficient mice, which lack CD4(+) T cells, displayed an increased survival despite comparable bacterial titers in the blood, spleen, and lung. The higher survival correlated with a lower cytokine and chemokine response upon S. pneumoniae challenge in MHC class II-deficient mice, suggesting that inflammation may contribute to the mortality of IPD. Comparable to the case for MHC class II-deficient mice, antibody-mediated depletion of CD4(+) T cells and drug-induced inhibition of T-cell function with cyclosporine, or interference with T-cell activation using CTLA4-immunoglobulin (Abatacept), led to significant increases in survival during IPD. Our results reveal an important and adverse role of CD4(+) T cells in the pathogenesis of IPD and suggest that modulation of T-cell activation during early phases of S. pneumoniae invasive infection may provide a therapeutic option.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Activación de Linfocitos/inmunología , Infecciones Neumocócicas/inmunología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Quimiocinas/sangre , Quimiocinas/fisiología , Ciclosporina/farmacología , Citocinas/sangre , Citocinas/fisiología , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/inmunología , Pulmón/inmunología , Pulmón/microbiología , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Bazo/inmunología , Bazo/microbiología , Streptococcus pneumoniae/inmunología
17.
Immunity ; 24(4): 475-87, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16618605

RESUMEN

TGFbeta presented and activated by integrin alphavbeta6 expressed on alveolar epithelial cells (AEC) continuously inhibits the functionality of alveolar macrophages (AM). Despite this inhibition, AM retain their ability to respond to inhaled microorganisms. Herein we describe a homeostatic circuit through which the effects of microbial products on macrophages transiently circumvent this inhibition by repressing alphavbeta6 expression on AEC. Subsequent production of matrix metalloproteinases by activated AM activates latent TGFbeta, reinduces alphavbeta6 expression, and thereby reinstates tonic inhibition of AM function. Our results reveal how AM can be activated while minimizing their potential to inflict collateral damage to the adjacent lung tissue and indicate that tissue-specific microenvironmental factors shape organ-specific defense strategies against microbial invasion.


Asunto(s)
Infecciones Bacterianas/inmunología , Comunicación Celular/inmunología , Homeostasis/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Western Blotting , Femenino , Integrinas/inmunología , Integrinas/metabolismo , Pulmón/citología , Pulmón/microbiología , Metaloproteinasas de la Matriz/inmunología , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptor Cross-Talk/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
18.
Thromb Haemost ; 95(1): 151-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16543974

RESUMEN

Chlamydia (C.) pneumoniae are thought to infect monocytes and use them as vectors into the vessel wall, where they may accelerate atherosclerosis. We investigated the effects of C. pneumoniae on monocytic matrix metalloproteinase (MMP) activation with focus on the role of the extracellular matrix metalloproteinase inducer EMMPRIN. Human monocytes or monocytic MonoMac6 cells were infected with C. pneumoniae. Infection enhanced mRNA- and surface expression of EMMPRIN and Membrane-type-1 Matrix Metalloproteinase (MT1-MMP), plus the secretion of MMP-7, MMP-9 and the urokinase receptor (uPAR). Chlamydial heat shock protein 60 was identified to be partially responsible for EMMPRIN and MMP-9 induction, while C. trachomatis-infection had no stimulatory effect, indicating a C. pneumoniae-specific activation pathway. Suppression of EMMPRIN by gene silencing almost completely hindered the induction of MT1-MMP and MMP-9 by C. pneumoniae, suggesting a predominant regulatory role for EMMPRIN. Moreover, C. pneumoniae-infected monocytes exhibited increased MMP- and plasmin-dependent migration through "matrigel". Additionally, incubation of SMCs with supernatants of C. pneumoniae-infected monocytes induced MMP-2 activation, which was inhibited by IL1-Receptor antagonist or anti-IL-6-mAb, indicating paracrine intercellular activation pathways. In conclusion, C.pneumoniae induce MMP activity directly in monocytes through an EMMPRIN-dependent pathway and indirectly in SMCs via monocyte-derived cytokines.


Asunto(s)
Aterosclerosis/enzimología , Basigina/metabolismo , Chlamydophila pneumoniae/aislamiento & purificación , Monocitos/enzimología , Aterosclerosis/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/farmacología , Basigina/genética , Línea Celular , Movimiento Celular , Chaperonina 60/genética , Chaperonina 60/farmacología , Colágeno , Combinación de Medicamentos , Matriz Extracelular/metabolismo , Humanos , Laminina , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz Asociadas a la Membrana , Monocitos/efectos de los fármacos , Monocitos/microbiología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Comunicación Paracrina , Proteoglicanos , Interferencia de ARN , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Rotura Espontánea/enzimología , Rotura Espontánea/microbiología
19.
Nature ; 439(7073): 204-7, 2006 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-16306937

RESUMEN

Toll-like receptors (TLRs) are activated by pathogen-associated molecular patterns to induce innate immune responses and production of pro-inflammatory cytokines, interferons and anti-inflammatory cytokines. TLRs activate downstream effectors through adaptors that contain Toll/interleukin-1 receptor (TIR) domains, but the mechanisms accounting for diversification of TLR effector functions are unclear. To dissect biochemically TLR signalling, we established a system for isolating signalling complexes assembled by dimerized adaptors. Using MyD88 as a prototypical adaptor, we identified TNF receptor-associated factor 3 (TRAF3) as a new component of TIR signalling complexes that is recruited along with TRAF6. Using myeloid cells from TRAF3- and TRAF6-deficient mice, we show that TRAF3 is essential for the induction of type I interferons (IFN) and the anti-inflammatory cytokine interleukin-10 (IL-10), but is dispensable for expression of pro-inflammatory cytokines. In fact, TRAF3-deficient cells overproduce pro-inflammatory cytokines owing to defective IL-10 production. Despite their structural similarity, the functions of TRAF3 and TRAF6 are largely distinct. TRAF3 is also recruited to the adaptor TRIF (Toll/IL-1 receptor domain-containing adaptor-inducing IFN-beta) and is required for marshalling the protein kinase TBK1 (also called NAK) into TIR signalling complexes, thereby explaining its unique role in activation of the IFN response.


Asunto(s)
Transducción de Señal , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antígenos de Diferenciación/química , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Línea Celular , Dimerización , Regulación de la Expresión Génica , Inmunidad Innata , Interferones/biosíntesis , Interleucina-10/biosíntesis , Ratones , Células Mieloides/metabolismo , Factor 88 de Diferenciación Mieloide , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Especificidad por Sustrato , Factor 6 Asociado a Receptor de TNF/deficiencia , Factor 6 Asociado a Receptor de TNF/genética
20.
J Immunol ; 174(9): 5864-73, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15843591

RESUMEN

The interactions between genetic and environmental factors play a major role in the development of childhood asthma. We hypothesized that a pre-existing Th2/asthmatic response can promote Th2 responses to newly encountered Ags (i.e., phenotype spread). To test this hypothesis, we developed a mouse model in which the requirements for the induction and inhibition of phenotype spread to a clinically relevant neo-allergen (i.e., ragweed) were investigated. Our results indicate that 1) phenotype spread to the neo-allergen can be induced only within the first 8 h after a bronchial challenge with the first Ag (OVA); 2) Th2 differentiation of naive CD4(+) T cells occurs in bronchial lymph nodes; 3) trafficking of naive CD4(+) T cells to local lymph nodes and IL-4 produced by OVA-activated Th2 cells play essential roles in the differentiation of naive CD4(+) T cells to Th2 cells; and 4) suppression of the production of chemokines involved in the homing of naive CD4(+) T and Th2 cells to bronchial lymph nodes by a TLR9 agonist inhibited phenotype spread and abrogated the consequent development of experimental asthma. These findings provide a mechanistic insight into Th2 phenotype spread and offer an animal model for testing relevant immunomodulatory interventions.


Asunto(s)
Adyuvantes Inmunológicos/fisiología , Asma/inmunología , Diferenciación Celular/inmunología , Inhibidores de Crecimiento/fisiología , Inmunofenotipificación , Células Th2/inmunología , Células Th2/metabolismo , Adyuvantes Inmunológicos/uso terapéutico , Traslado Adoptivo , Ambrosia/inmunología , Animales , Asma/patología , Asma/prevención & control , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Diferenciación Celular/genética , Línea Celular , Movimiento Celular/genética , Movimiento Celular/inmunología , Niño , Inhibidores de Crecimiento/uso terapéutico , Humanos , Interleucina-4/biosíntesis , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones SCID , Ratones Transgénicos , Oligodesoxirribonucleótidos/uso terapéutico , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Fase de Descanso del Ciclo Celular/genética , Fase de Descanso del Ciclo Celular/inmunología , Células Th2/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA