Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
EMBO J ; 41(8): e109700, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35274759

RESUMEN

HOIL-1, a component of the linear ubiquitin chain assembly complex (LUBAC), ubiquitylates serine and threonine residues in proteins by esterification. Here, we report that mice expressing an E3 ligase-inactive HOIL-1[C458S] mutant accumulate polyglucosan in brain, heart and other organs, indicating that HOIL-1's E3 ligase activity is essential to prevent these toxic polysaccharide deposits from accumulating. We found that HOIL-1 monoubiquitylates glycogen and α1:4-linked maltoheptaose in vitro and identify the C6 hydroxyl moiety of glucose as the site of ester-linked ubiquitylation. The monoubiquitylation of maltoheptaose was accelerated > 100-fold by the interaction of Met1-linked or Lys63-linked ubiquitin oligomers with the RBR domain of HOIL-1. HOIL-1 also transferred pre-formed ubiquitin oligomers to maltoheptaose en bloc, producing polyubiquitylated maltoheptaose in one catalytic step. The Sharpin and HOIP components of LUBAC, but not HOIL-1, bound to unbranched and infrequently branched glucose polymers in vitro, but not to highly branched mammalian glycogen, suggesting a potential function in targeting HOIL-1 to unbranched glucosaccharides in cells. We suggest that monoubiquitylation of unbranched glucosaccharides may initiate their removal from cells, preventing precipitation as polyglucosan.


Asunto(s)
Ubiquitina-Proteína Ligasas , Ubiquitina , Animales , Glucanos , Glucosa , Glucógeno/metabolismo , Mamíferos , Ratones , FN-kappa B/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
2.
EMBO Rep ; 22(10): e50743, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34403206

RESUMEN

The rapid formation and activation of the NLRP3 inflammasome is induced by co-stimulation with LPS and nigericin. It requires the LPS-stimulated activation of IKKß, which exerts its effects independently of de novo gene transcription, protein translation and other protein kinases activated by IKKß. IKKß is not required for the nigericin-induced dispersion of the trans-Golgi network (TGN), but to bring NLRP3 in proximity with TGN38. The nigericin-induced dispersion of the Golgi is enhanced by co-stimulation with LPS, and this enhancement is IKKß-dependent. Prolonged stimulation with LPS to increase the expression of NLRP3, followed by stimulation with nigericin, produced larger TGN38-positive puncta, and the ensuing activation of the NLRP3 inflammasome was also suppressed by IKKß inhibitors added prior to stimulation with nigericin. IKKß therefore has a key role in recruiting NLRP3 to the dispersed TGN, leading to the formation and activation of the NLRP3 inflammasome.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Quinasa I-kappa B , Inflamasomas/genética , Interleucina-1beta , Lipopolisacáridos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Nigericina , Red trans-Golgi
3.
J Immunol ; 201(11): 3373-3382, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30355787

RESUMEN

The A20-binding inhibitor of NF-κB 2 (ABIN2) interacts with Met1-linked ubiquitin chains and is an integral component of the tumor progression locus 2 (Tpl2) kinase complex. We generated a knock-in mouse expressing the ubiquitin-binding-defective mutant ABIN2[D310N]. The expression of Tpl2 and its activation by TLR agonists in macrophages or by IL-1ß in fibroblasts from these mice was unimpaired, indicating that the interaction of ABIN2 with ubiquitin oligomers is not required for the stability or activation of Tpl2. The ABIN2[D310N] mice displayed intestinal inflammation and hypersensitivity to dextran sodium sulfate-induced colitis, an effect that was mediated by radiation-resistant cells rather than by hematopioetic cells. The IL-1ß-dependent induction of cyclooxygenase 2 (COX2) and the secretion of PGE2 was reduced in mouse embryonic fibroblasts and intestinal myofibroblasts (IMFs) from ABIN2[D310N] mice. These observations are similar to those reported for the Tpl2 knockout (KO) mice (Roulis et al. 2014. Proc. Natl. Acad. Sci. USA 111: E4658-E4667), but the IL-1ß-dependent production of COX2 and PGE2 in mouse embryonic fibroblasts or IMFs was unaffected by pharmacological inhibition of Tpl2 in wild-type mice. The expression of ABIN2 is decreased drastically in Tpl2 KO mice. These and other lines of evidence suggest that the hypersensitivity of Tpl2 KO mice to dextran sodium sulfate-induced colitis is not caused by the loss of Tpl2 catalytic activity but by the loss of ABIN2, which impairs COX2 and PGE2 production in IMFs by a Tpl2 kinase-independent pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Colitis/inmunología , Quinasas Quinasa Quinasa PAM/metabolismo , Macrófagos/inmunología , Miofibroblastos/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Colitis/inducido químicamente , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Sulfato de Dextran , Dinoprostona/metabolismo , Técnicas de Sustitución del Gen , Interleucina-1beta/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación/genética , Unión Proteica/genética , Proteínas Proto-Oncogénicas/genética , Ribonucleasa Pancreática/metabolismo , Transducción de Señal , Ubiquitinas/metabolismo
4.
J Immunol ; 197(11): 4266-4273, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27807192

RESUMEN

Polymorphisms in the TNIP1 gene encoding A20-binding inhibitor of NF-κB1 (ABIN1) predispose to lupus and other autoimmune diseases in at least eight human populations. We found previously that knock-in mice expressing a ubiquitin-binding-defective mutant of ABIN1 (ABIN1[D485N]) develop autoimmunity as they age and succumb to a disease resembling lupus nephritis in humans. In this article, we report that Flt3-derived dendritic cells from these mice overproduced type 1 IFNs upon stimulation with ligands that activate TLR7 or TLR9. However, crossing ABIN1[D485N] mice to IFNAR1-knockout mice that do not express the α-subunit of the type 1 IFNR did not prevent splenomegaly, the appearance of high serum levels of autoantibodies and other Igs, or liver inflammation and only reduced kidney inflammation modestly. In contrast, crossing ABIN1[D485N] mice to knock-in mice expressing catalytically inactive mutants of IRAK1 or IRAK4 prevented splenomegaly, autoimmunity, and liver and kidney inflammation. Our results support the notion that IRAK1 and/or IRAK4 are attractive targets for the development of drugs to prevent, and perhaps treat, lupus nephritis and other autoinflammatory diseases caused by the decreased ability of ABIN1 or other proteins to restrict the strength of MyD88 signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Regulación de la Expresión Génica/inmunología , Interferón Tipo I/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Nefritis Lúpica/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Sustitución de Aminoácidos , Animales , Cruzamientos Genéticos , Regulación de la Expresión Génica/genética , Técnicas de Sustitución del Gen , Interferón Tipo I/genética , Quinasas Asociadas a Receptores de Interleucina-1/genética , Riñón , Nefritis Lúpica/genética , Nefritis Lúpica/patología , Nefritis Lúpica/prevención & control , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Noqueados , Mutación Missense , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
5.
J Immunol ; 191(5): 2717-30, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23918981

RESUMEN

The roles of IL-1R-associated kinase (IRAK)2 and IRAK1 in cytokine production were investigated using immune cells from knock-in mice expressing the TNFR-associated factor 6 (TRAF6) binding-defective mutant IRAK2[E525A] or the catalytically inactive IRAK1[D359A] mutant. In bone marrow-derived macrophages (BMDMs), the IRAK2-TRAF6 interaction was required for the late (2-8 h) but not the early phase (0-2 h) of il6 and tnfa mRNA production, and hence for IL-6 and TNF-α secretion by TLR agonists that signal via MyD88. Loss of the IRAK2-TRAF6 interaction had little effect on the MyD88-dependent production of anti-inflammatory molecules produced during the early phase, such as Dual Specificity Phosphatase 1, and a modest effect on IL-10 secretion. The LPS/TLR4-stimulated production of il6 and tnfa mRNA and IL-6 and TNF-α secretion was hardly affected, because the Toll/IL-1R domain-containing adapter-inducing IFN-ß (TRIF) signaling pathway was used instead of the IRAK2-TRAF6 interaction to sustain late-phase mRNA production. IRAK1 catalytic activity was not rate limiting for il6, tnfa, or il10 mRNA production or the secretion of these cytokines by BMDMs, but IFN-ß mRNA induction by TLR7 and TLR9 agonists was greatly delayed in plasmacytoid dendritic cells (pDCs) from IRAK1[D359A] mice. In contrast, IFN-ß mRNA production was little affected in pDCs from IRAK2[E525A] mice, but subsequent IFN-α mRNA production and IFN-α secretion were reduced. IFN-ß and IFN-α production were abolished in pDCs from IRAK1[D359A] × IRAK2[E525A] double knock-in mice. Our results establish that the IRAK2-TRAF6 interaction is rate limiting for the late, but not the early phase of cytokine production in BMDM and pDCs, and that the IRAK2-TRAF6 interaction is needed to sustain IκB-inducing kinase ß activity during prolonged activation of the MyD88 signaling network. [corrected]


Asunto(s)
Citocinas/biosíntesis , Células Dendríticas/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Macrófagos/metabolismo , Transducción de Señal/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Dendríticas/inmunología , Técnicas de Sustitución del Gen , Quinasa I-kappa B/inmunología , Quinasa I-kappa B/metabolismo , Immunoblotting , Inmunoprecipitación , Inflamación/inmunología , Inflamación/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Macrófagos/inmunología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor 6 Asociado a Receptor de TNF/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo , Transfección
6.
J Gen Virol ; 95(Pt 1): 44-51, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24158397

RESUMEN

The V proteins of paramyxoviruses are composed of two evolutionarily distinct domains, the N-terminal 75 % being common to the viral P, V and W proteins, and not highly conserved between viruses, whilst the remaining 25 % consists of a cysteine-rich V-specific domain, which is conserved across almost all paramyxoviruses. There is evidence supporting a number of different functions of the V proteins of morbilliviruses in blocking the signalling pathways of type I and II IFNs, but it is not clear which domains of V are responsible for which activities and whether all these activities are required for effective blockade of IFN signalling. We have shown here that the two domains of rinderpest virus V protein have distinct functions: the N-terminal domain acted to bind STAT1, whilst the C-terminal V-specific domain interacted with the IFN receptor-associated kinases Jak1 and Tyk2. Effective blockade of IFN signalling required the intact V protein.


Asunto(s)
Interferones/metabolismo , Virus de la Peste Bovina/metabolismo , Peste Bovina/metabolismo , Transducción de Señal , Proteínas Virales/química , Proteínas Virales/metabolismo , Animales , Línea Celular , Humanos , Interferones/genética , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Peste Bovina/enzimología , Peste Bovina/genética , Peste Bovina/virología , Virus de la Peste Bovina/química , Virus de la Peste Bovina/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Proteínas Virales/genética
7.
J Am Soc Nephrol ; 24(11): 1743-54, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23970121

RESUMEN

The genetic factors underlying the pathogenesis of lupus nephritis associated with systemic lupus erythematosus are largely unknown, although animal studies indicate that nuclear factor (NF)-κB is involved. We reported previously that a knockin mouse expressing an inactive form of ABIN1 (ABIN1[D485N]) develops lupus-like autoimmune disease and demonstrates enhanced activation of NF-κB and mitogen-activated protein kinases in immune cells after toll-like receptor stimulation. In the current study, we show that ABIN1[D485N] mice develop progressive GN similar to class III and IV lupus nephritis in humans. To investigate the clinical relevance of ABIN1 dysfunction, we genotyped five single-nucleotide polymorphisms in the gene encoding ABIN1, TNIP1, in samples from European-American, African American, Asian, Gullah, and Hispanic participants in the Large Lupus Association Study 2. Comparing cases of systemic lupus erythematosus with nephritis and cases of systemic lupus erythematosus without nephritis revealed strong associations with lupus nephritis at rs7708392 in European Americans and rs4958881 in African Americans. Comparing cases of systemic lupus erythematosus with nephritis and healthy controls revealed a stronger association at rs7708392 in European Americans but not at rs4958881 in African Americans. Our data suggest that variants in the TNIP1 gene are associated with the risk for lupus nephritis and could be mechanistically involved in disease development via aberrant regulation of NF-κB and mitogen-activated protein kinase activity.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Nefritis Lúpica/genética , Animales , Proteínas de Unión al ADN/genética , Técnica del Anticuerpo Fluorescente , Humanos , Riñón/patología , Riñón/fisiopatología , Nefritis Lúpica/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/fisiología , Polimorfismo de Nucleótido Simple
8.
Lupus Sci Med ; 10(1)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36822800

RESUMEN

OBJECTIVE: The aim of the study was to investigate whether the IRAK1/JAK2/Flt3 inhibitor pacritinib prevents disease development in the lupus-prone ABIN1[D485N] knock-in mouse. METHODS: ABIN1[D485N] knock-in mice aged 8 weeks were fed for 10 weeks on a diet containing pacritinib. Body weight was monitored, and serum collected at the end to measure pacritinib, autoantibody and immunoglobulin levels. Splenic immune cell populations were analysed, and the kidney, liver and lungs examined for pathology. RESULTS: Pacritinib prevented multiple facets of the lupus phenotype in ABIN1[D485N] knock-in mice, including splenomegaly, expansion of splenic germinal centre B cells, follicular T helper cells, and neutrophils, elevated serum levels of double-stranded DNA antibodies and immunoglobulins, glomerular IgA and lung inflammation. CONCLUSIONS: Pacritinib may be useful for the treatment of multiorgan inflammation in patients with lupus.


Asunto(s)
Lupus Eritematoso Sistémico , Animales , Ratones , Inflamación , Riñón/patología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/patología , Fenotipo
9.
Front Cell Dev Biol ; 10: 887533, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35800898

RESUMEN

In this review, we have summarized classical post-translational modifications (PTMs) such as phosphorylation, ubiquitylation, and SUMOylation of the different components of one of the most studied NLRP3, and other emerging inflammasomes. We will highlight how the discovery of these modifications have provided mechanistic insight into the biology, function, and regulation of these multiprotein complexes not only in the context of the innate immune system but also in adaptive immunity, hematopoiesis, bone marrow transplantation, as well and their role in human diseases. We have also collected available information concerning less-studied modifications such as acetylation, ADP-ribosylation, nitrosylation, prenylation, citrullination, and emphasized their relevance in the regulation of inflammasome complex formation. We have described disease-associated mutations affecting PTMs of inflammasome components. Finally, we have discussed how a deeper understanding of different PTMs can help the development of biomarkers and identification of novel drug targets to treat diseases caused by the malfunctioning of inflammasomes.

10.
Lupus Sci Med ; 8(1)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34836923

RESUMEN

OBJECTIVE: We have reported previously that the IRAK4 inhibitor PF06426779 given to ubiquitin-binding-defective ABIN1[D485N] mice at 6 weeks of age prevents the major facets of lupus that develop 10 weeks later. The present study was undertaken to investigate whether PF06426779 could reverse the lupus phenotype when administered to 13-week-old ABIN1[D485N] mice that had already developed symptoms of lupus. METHODS: Splenomegaly, the number of splenic neutrophils, TFH and Germinal Centre B (GCB) cells, serum levels of immunoglobulins, the extent of kidney, liver and lung pathology, and glomerular IgA and IgM were measured after feeding 13-week-old ABIN1[D485N] and wild-type mice for another 10 weeks with R&M3 diet with and without PF06426779 (4 g/kg). RESULTS: Following drug treatment, spleen size and weight, splenic neutrophil numbers, and serum IgA and glomerular IgA levels of ABIN1[D485N] mice returned to those seen in wild-type mice. The rise in splenic TFH and GCB numbers, the increase in kidney and liver pathology, and the concentrations of serum IgG1, IgG2A and IgE between 13 and 23 weeks were suppressed. There was no reduction in the level of anti-self double-stranded DNA, anti-self nuclear antigens or IgM during the drug treatment. CONCLUSIONS: The results demonstrate the therapeutic potential of IRAK4 inhibitors for the treatment of lupus and raise the possibility of monitoring efficacy by measuring decreases in the serum levels of IgA. Our results support the view that there may be a closer connection between lupus and IgA nephropathy than realised previously.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1 , Lupus Eritematoso Sistémico , Animales , Humanos , Riñón/patología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/patología , Ratones , Fenotipo
11.
FEBS J ; 288(20): 5909-5924, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33932090

RESUMEN

The atypical E3 ligase HOIL-1 forms ester bonds between ubiquitin and serine/threonine residues in proteins, but the physiological roles of this unusual modification are unknown. We now report that IL-18 signalling leading to the production of interferon γ (IFNγ) and granulocyte-macrophage colony-stimulating factor (GM-CSF) is enhanced in cytotoxic T cells from knock-in mice expressing the E3 ligase-inactive HOIL-1[C458S] mutant, demonstrating that the formation of HOIL-1-catalysed ester-linked ubiquitin bonds restricts the activation of this pathway. We show that the interaction of IRAK2 with TRAF6 is required for IL-18-stimulated IFN-γ and GM-CSF production, and that the increased production of these cytokines in cytotoxic T cells from HOIL-1[C458S] mice correlates with an increase in both the number and size of the Lys63/Met1-linked hybrid ubiquitin chains attached to IRAK2 in these cells. In contrast, the secretion of IL-12 and IL-6 and the formation of il-12 and il-6 mRNA induced in bone marrow-derived macrophages (BMDMs) by prolonged stimulation with TLR-activating ligands that signal via myddosomes, which also requires the interaction of IRAK2 with TRAF6, were not increased but modestly reduced in HOIL-1[C458S] BMDM. The decreased production of these cytokines correlated with reduced ubiquitylation of IRAK2. Our results establish that changes in HOIL-1-catalysed ester-linked ubiquitylation can promote or reduce cytokine production depending on the ligand, receptor and immune cell and may be explained by differences in the ubiquitylation of IRAK2.


Asunto(s)
Ésteres/química , Interleucina-18/metabolismo , Macrófagos/inmunología , Linfocitos T Citotóxicos/inmunología , Receptores Toll-Like/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitina/química , Ubiquitinación , Animales , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
12.
Adv Biol Regul ; 75: 100666, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31615747

RESUMEN

Components of bacteria and viruses activate Toll-Like Receptors in host cells, triggering the formation of the Myddosome and a signalling network that culminates in the production and release of the inflammatory mediators required to combat pathogenic infection. The Myddosome initiates signalling by recruiting and activating five E3 ligases that generate hybrid ubiquitin chains and attach them to components of the Myddosome. These ubiquitin chains act as a scaffold for the recruitment and activation of ubiquitin-binding proteins, which include the "master" protein kinases TAK1 and IKKß that drive inflammatory mediator production, as well as other proteins like ABIN1 and A20 that restrict activation of the network to prevent the overproduction of these substances that can lead to autoimmunity and organ damage. Here we review recent developments in our understanding of this network, focusing on the unexpected discovery that the E3 ligase HOIL-1 initiates the formation of hybrid ubiquitin chains by forming an ester bond between the first ubiquitin and the protein components of the Myddosome.


Asunto(s)
Factor 88 de Diferenciación Mieloide/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Ubiquitinación , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Factores de Transcripción/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética
13.
PLoS One ; 8(2): e57063, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23431397

RESUMEN

Morbilliviruses form a closely related group of pathogenic viruses which encode three non-structural proteins V, W and C in their P gene. Previous studies with rinderpest virus (RPV) and measles virus (MeV) have demonstrated that these non-structural proteins play a crucial role in blocking type I (IFNα/ß) and type II (IFNγ) interferon action, and various mechanisms have been proposed for these effects. We have directly compared four important morbilliviruses, rinderpest (RPV), measles virus (MeV), peste des petits ruminants virus (PPRV) and canine distemper virus (CDV). These viruses and their V proteins could all block type I IFN action. However, the viruses and their V proteins had varying abilities to block type II IFN action. The ability to block type II IFN-induced gene transcription correlated with co-precipitation of STAT1 with the respective V protein, but there was no correlation between co-precipitation of either STAT1 or STAT2 and the abilities of the V proteins to block type I IFN-induced gene transcription or the creation of the antiviral state. Further study revealed that the V proteins of RPV, MeV, PPRV and CDV could all interfere with phosphorylation of the interferon-receptor-associated kinase Tyk2, and the V protein of highly virulent RPV could also block the phosphorylation of another such kinase, Jak1. Co-precipitation studies showed that morbillivirus V proteins all form a complex containing Tyk2 and Jak1. This study highlights the ability of morbillivirus V proteins to target multiple components of the IFN signalling pathways to control both type I and type II IFN action.


Asunto(s)
Interferón Tipo I/fisiología , Interferón gamma/fisiología , Morbillivirus/fisiología , Proteínas Virales/fisiología , Animales , Antígenos CD , Línea Celular Tumoral , Núcleo Celular/metabolismo , Chlorocebus aethiops , Perros , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Janus Quinasa 1/metabolismo , Microscopía Fluorescente , Morbillivirus/inmunología , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Receptores de Superficie Celular , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , TYK2 Quinasa/metabolismo , Transcripción Genética , Células Vero
14.
J Exp Med ; 208(6): 1215-28, 2011 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-21606507

RESUMEN

The protein ABIN1 possesses a polyubiquitin-binding domain homologous to that present in nuclear factor κB (NF-κB) essential modulator (NEMO), a component of the inhibitor of NF-κB (IκB) kinase (IKK) complex. To address the physiological significance of polyubiquitin binding, we generated knockin mice expressing the ABIN1[D485N] mutant instead of the wild-type (WT) protein. These mice developed all the hallmarks of autoimmunity, including spontaneous formation of germinal centers, isotype switching, and production of autoreactive antibodies. Autoimmunity was suppressed by crossing to MyD88(-/-) mice, demonstrating that toll-like receptor (TLR)-MyD88 signaling pathways are needed for the phenotype to develop. The B cells and myeloid cells of the ABIN1[D485N] mice showed enhanced activation of the protein kinases TAK, IKK-α/ß, c-Jun N-terminal kinases, and p38α mitogen-activated protein kinase and produced more IL-6 and IL-12 than WT. The mutant B cells also proliferated more rapidly in response to TLR ligands. Our results indicate that the interaction of ABIN1 with polyubiquitin is required to limit the activation of TLR-MyD88 pathways and prevent autoimmunity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Poliubiquitina/química , Animales , Autoinmunidad , Citocinas/metabolismo , Femenino , Humanos , Quinasa I-kappa B/metabolismo , Ligandos , Lisina/química , Masculino , Ratones , Ratones Transgénicos , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Mutación , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Transducción de Señal
15.
Virology ; 385(1): 134-42, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19108859

RESUMEN

The innate immune response, in particular the production of type 1 interferons, is an essential part of the mammalian host response to viral infection. We have previously shown that rinderpest virus, a morbillivirus closely related to the human pathogen measles virus, blocks the actions of type 1 and type 2 interferons. We show here that this virus can also block the induction of type 1 interferon. The viral non-structural C protein appears to be the active agent, since expressing this protein in cells makes them resistant to activation of the interferon-beta promoter while recombinant virus that does not express the C protein activates this promoter much more than virus expressing the C protein. In addition, differences in activation of the interferon-beta promoter by different strains of rinderpest virus are reflected in differing abilities of their respective C proteins to block activation of the promoter by dsRNA. The C protein blocks the activation of this promoter induced by either cytoplasmic dsRNA or by Newcastle disease virus (NDV) infection, as well as activation induced by overexpression of several elements of the signalling pathway, including mda-5, RIG-I and IRF-3. The RPV C protein also blocks transcription from promoters responsive individually to the three transcription factors that make up the interferon-beta promoter enhanceosome, although it does not appear to block the activation of IRF-3.


Asunto(s)
Interferón Tipo I/inmunología , Virus de la Peste Bovina/fisiología , Peste Bovina/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Humanos , Ratones , Peste Bovina/virología , Transfección , Células Vero , Replicación Viral/fisiología
16.
Virology ; 395(2): 223-31, 2009 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-19854460

RESUMEN

The morbilliviruses are a closely related genus which are very similar in their sequences and share a common receptor, but nevertheless show significant restriction in the host species in which they cause disease. One contribution to this restriction might be the nature of the hosts' responses to infection. We have used microarrays to study the changes in the transcriptome of bovine dendritic cells after infection with wild-type (pathogenic) and vaccine (apathogenic) strains of rinderpest virus (RPV), a bovine pathogen, and a wild-type isolate of measles virus (MV), a morbillivirus that causes disease only in humans and some other primates. We found that, as previously observed in human cells, MV induces a rapid interferon response, while that induced by RPV was delayed and much reduced in magnitude. Pathogenic and apathogenic RPV also showed significant differences, with the latter inducing a slightly higher interferon response as well as significant effects on transcription of genes involved in cell cycle regulation.


Asunto(s)
Células Dendríticas/virología , Virus del Sarampión/fisiología , Virus de la Peste Bovina/fisiología , Transcripción Genética/fisiología , Animales , Bovinos , Inmunidad Innata/genética
17.
J Virol ; 80(15): 7555-68, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16840335

RESUMEN

Rinderpest virus (RPV) is a paramyxovirus closely related to the human pathogen Measles virus. It causes severe disease in cattle, buffalo, and some wild animals; although it can infect humans, it does not cause disease. Here, we demonstrate that RPV blocks the action of both type I (alpha) and type II (gamma) interferons (IFNs) by blocking the phosphorylation and nuclear translocation of STAT1 and STAT2 and that this block is not related to species specificity. In addition, both wild-type virulent and vaccine strains of the virus blocked IFN action. Unlike the case with some other paramyxoviruses, neither STAT1 nor STAT2 is degraded upon virus infection. STAT1 is bound by both the viral structural protein P, and thereby recruited to concentrations of viral protein in the cell, and the nonstructural protein V. Although both P and V proteins bind to STAT1 and can block IFN action when expressed in transfected cells, the IFN antagonist activity of the P protein is weaker than that of the V protein. The viral C protein also seems to weakly block IFN-induced activation of STAT1 in transfection experiments. However, studies with knockout viruses showed that the viral V protein appears to be the dominant inhibitor of IFN signaling in the context of virus infection, since prevention of viral V expression restored the IFN sensitivity of infected cells. Although a change in the distribution pattern of STAT2 was observed in virus-infected cells, STAT2 was not bound by any viral protein.


Asunto(s)
Antivirales/farmacología , Interferón-alfa/farmacología , Interferón gamma/farmacología , Virus de la Peste Bovina/fisiología , Proteínas no Estructurales Virales/fisiología , Proteínas Estructurales Virales/fisiología , Animales , Bovinos , Núcleo Celular , Chlorocebus aethiops , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/virología , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/virología , Fosforilación , Transporte de Proteínas , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Piel/citología , Piel/metabolismo , Piel/virología , Células Vero/metabolismo , Células Vero/virología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA