Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
EMBO Rep ; 22(12): e52254, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34633746

RESUMEN

Promyelocytic leukemia protein (PML) is a tumor suppressor possessing multiple modes of action, including induction of apoptosis. We unexpectedly find that PML promotes necroptosis in addition to apoptosis, with Pml-/- macrophages being more resistant to TNF-mediated necroptosis than wild-type counterparts and PML-deficient mice displaying resistance to TNF-induced systemic inflammatory response syndrome. Reduced necroptosis in PML-deficient cells is associated with attenuated receptor-interacting protein kinase 1 (RIPK1) activation, as revealed by reduced RIPK1[S166] phosphorylation, and attenuated RIPK1-RIPK3-MLKL necrosome complex formation. We show that PML deficiency leads to enhanced TNF-induced MAPK-activated kinase 2 (MK2) activation and elevated RIPK1[S321] phosphorylation, which suppresses necrosome formation. MK2 inhibitor treatment or MK2 knockout abrogates resistance to cell death induction in PML-null cells and mice. PML binds MK2 and p38 MAPK, thereby inhibiting p38-MK2 interaction and MK2 activation. Moreover, PML participates in autocrine production of TNF induced by cellular inhibitors of apoptosis 1 (cIAP1)/cIAP2 degradation, since PML-knockout attenuates autocrine TNF. Thus, by targeting MK2 activation and autocrine TNF, PML promotes necroptosis and apoptosis, representing a novel tumor-suppressive activity for PML.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Proteínas Serina-Treonina Quinasas , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal , Animales , Apoptosis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Necroptosis , Fosforilación , Proteína de la Leucemia Promielocítica/genética , Proteína de la Leucemia Promielocítica/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Rheumatology (Oxford) ; 58(4): 719-728, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30629240

RESUMEN

OBJECTIVE: Deletion of Deltex1 (DTX1) in mice caused hyperactivation of T cells and lupus-like autoimmune syndromes, however, the association of DTX1 with human autoimmune diseases is totally unknown. This study investigated the role of DTX1 in human T cell functions and its correlation with disease activity in patients with SLE. METHODS: The influence of DTX1 on T cell function was evaluated using human primary cells. DTX1 expression in peripheral blood mononuclear cells (PBMCs) from healthy controls and SLE patients was measured by quantitative real-time PCR and the SLEDAI was used to assess disease activity. RESULTS: After stimulation with anti-CD3 and anti-CD28, silencing of DTX1 expression enhanced IFN-γ secretion by human T cells. The expression of DTX1 in PBMCs was significantly lower in 100 SLE patients than in 50 age- and sex-matched healthy controls (DTX1/glyceraldehyde 3-phosphate dehydrogenase, 0.452 vs 1.269, P < 0.001). The area under the receiver operator characteristics curve of the model was 0.737 (95% CI 0.658, 0.815). Intriguingly, a low DTX1 level in T cells led to high IFN-γ production in SLE patients and had a correlation with severe disease activity. In addition, low DTX1 expression in SLE patients was associated with active LN, lung involvement or hypocomplementaemia. CONCLUSION: Knockdown DTX1 expression in human T cells reduced IFN-γ secretion. DTX1 expression in the PBMCs was significantly lower in SLE patients and had an inverse correlation with disease activity, indicating that the DTX1 level may be a good disease marker of SLE.


Asunto(s)
Lupus Eritematoso Sistémico/sangre , Linfocitos T/metabolismo , Ubiquitina-Proteína Ligasas/sangre , Biomarcadores/sangre , Humanos , Interferón gamma/metabolismo , Leucocitos Mononucleares/metabolismo
3.
Immunity ; 31(1): 72-83, 2009 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-19592273

RESUMEN

The molecular process underlying T cell anergy is incompletely understood. Deltex1 (DTX1) is a Notch target with unknown physiological function. Here we show that Dtx1 was a transcription target of nuclear factor of activated T cells (NFAT) and participated in T cell anergy. DTX1 protein was upregulated during T cell anergy, and transgenic expression of Dtx1 attenuated T cell activation. DTX1 inhibited T cell activation by both E3-dependent and E3-independent mechanisms. In addition, DTX1 suppressed T cell activation in the absence of its Notch-binding domain. Importantly, DTX1 regulated the expression of two anergy-associated molecules, growth arrest and DNA-damage-inducible 45 beta (Gadd45 beta) and Cbl-b. DTX1 interacted with early growth response 2 (Egr-2) for optimum expression of Cbl-b. Furthermore, deficiency of DTX1 augmented T cell activation, conferred resistance to anergy induction, enhanced autoantibody generation, and increased inflammation. DTX1 therefore represents a component downstream of calcium-NFAT signaling that regulates T cell anergy.


Asunto(s)
Anergia Clonal/genética , Proteínas de Unión al ADN/genética , Factores de Transcripción NFATC/metabolismo , Linfocitos T/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Autoinmunidad/inmunología , Proteínas de Unión al ADN/inmunología , Proteína 2 de la Respuesta de Crecimiento Precoz/inmunología , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Proteínas Inmediatas-Precoces/inmunología , Proteínas Inmediatas-Precoces/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-cbl/inmunología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Linfocitos T/metabolismo , Ubiquitina-Proteína Ligasas , Regulación hacia Arriba/inmunología
4.
J Immunol ; 195(6): 2612-23, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26238491

RESUMEN

Cellular FLIP (c-FLIP) specifically inhibits caspase-8 and suppresses death receptor-induced apoptosis. c-FLIP has also been reported to transmit activation signals. In this study, we report a novel function of c-FLIP involving inhibition of myeloid cell activation through antagonizing the selective innate signaling pathway. We found that conditional knockout of c-FLIP in dendritic cells (DCs) led to neutrophilia and splenomegaly. Peripheral DC populations, including CD11b(+) conventional DCs (cDCs), CD8(+) cDCs, and plasmacytoid DCs, were not affected by c-FLIP deficiency. We also found that c-FLIP knockout cDCs, plasmacytoid DCs, and bone marrow-derived DCs (BMDCs) displayed enhanced production of TNF-α, IL-2, or G-CSF in response to stimulation of TLR4, TLR2, and dectin-1. Consistent with the ability of c-FLIP to inhibit the activation of p38 MAPK, the enhanced activation of c-FLIP-deficient BMDCs could be partly linked to an elevated activation of p38 MAPK after engagement of innate receptors. Increased activation was also found in c-FLIP(+/-) macrophages. Additionally, the increased activation in c-FLIP-deficient DCs was independent of caspase-8. Our results reveal a novel inhibitory role of c-FLIP in myeloid cell activation and demonstrate the unexpected anti-inflammatory activity of c-FLIP. Additionally, our observations suggest that cancer therapy targeting c-FLIP downregulation may facilitate DC activation and increase T cell immunity.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/inmunología , Caspasa 8/inmunología , Células Dendríticas/inmunología , Células Mieloides/inmunología , Animales , Antiinflamatorios , Presentación de Antígeno/inmunología , Apoptosis/inmunología , Antígeno B7-1/biosíntesis , Antígeno B7-2/biosíntesis , Células de la Médula Ósea/inmunología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Caspasa 8/genética , Activación Enzimática/genética , Activación Enzimática/inmunología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Antígenos de Histocompatibilidad Clase II/biosíntesis , Inflamación/inmunología , Interleucina-2/biosíntesis , Lectinas Tipo C/inmunología , Recuento de Leucocitos , Ratones , Ratones Noqueados , Células Mieloides/citología , Neutrófilos/citología , Neutrófilos/inmunología , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/inmunología , Esplenomegalia/inmunología , Linfocitos T/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Blood ; 124(18): 2847-57, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25190756

RESUMEN

Emerging evidence indicates that innate immunodeficiency syndromes are linked to mutations in innate receptors and to specific infections. X-linked lymphoproliferative syndrome type-2 (XLP-2) is associated with deficiency in X-linked inhibitor of apoptosis protein (XIAP), with poorly understood molecular mechanisms. Here we showed that XIAP deficiency selectively impaired B-cell chronic lymphocytic leukemia/lymphoma 10 (BCL10)-mediated innate responses to dectin-1 ligands but did not affect responses to various Toll-like receptor agonists. Consequently, Xiap(-/-) mice became highly vulnerable on Candida albicans infection. The compromised early innate responses led to the persistent presence of C albicans and inflammatory cytokines in Xiap(-/-) mice. Furthermore, priming of Xiap(-/-) mice with the dectin-1 ligand curdlan alone resulted in XLP-2-like syndromes. Restoration of dectin-1-induced Rac1 activation and phagocytosis by resolvin D1, but not up-regulation of nuclear factor-κB, rescued Xiap(-/-) mice from C albicans lethal infection. Therefore, development of XLP-2 in XIAP-deficient patients could be partly due to sustained inflammation as a consequence of defective BCL10-dependent innate immunity toward specific pathogens. Importantly, our results suggest the potential therapeutic value of resolvin D1 in the treatment of XLP-2 and innate immunodeficiency syndromes.


Asunto(s)
Candidiasis/inmunología , Candidiasis/patología , Inmunidad Innata , Proteínas Inhibidoras de la Apoptosis/deficiencia , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteína 10 de la LLC-Linfoma de Células B , Candida albicans/efectos de los fármacos , Candida albicans/fisiología , Candidiasis/microbiología , Receptores ErbB/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Imidazoles/farmacología , Inmunidad Innata/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Lectinas Tipo C/agonistas , Lectinas Tipo C/metabolismo , Lipopéptidos/farmacología , Lipopolisacáridos/farmacología , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/patología , Lisina/metabolismo , Lisofosfolípidos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , FN-kappa B/metabolismo , Fagocitosis/efectos de los fármacos , Poli I-C/farmacología , Unión Proteica/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitinación/efectos de los fármacos , beta-Glucanos
6.
J Immunol ; 193(4): 1672-80, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25000980

RESUMEN

The generation of T cell anergy is associated with upregulation of ubiquitin E3 ligases including Casitas B-lineage lymphoma (Cbl-b), Itch, gene related to anergy in lymphocyte, and deltex1 (DTX1). These E3 ligases attenuate T cell activation by targeting to signaling molecules. For example, Cbl-b and Itch promote the degradation of protein kinase Cθ (PKCθ) and phospholipase C-γ1 (PLC-γ1) in anergic Th1 cells. How these anergy-associated E3 ligases coordinate during T cell anergy remains largely unknown. In the current study, we found that PKCθ and PLC-γ1 are also downregulated by DTX1. DTX1 interacted with PKCθ and PLC-γ1 and stimulated the degradation of PKCθ and PLC-γ1. T cell anergy-induced proteolysis of PKCθ was prevented in Dtx1(-/-) T cells, supporting the essential role of DTX1 in PKCθ downregulation. Similar to Cbl-b and Itch, DTX1 promoted monoubiquitination of PKCθ. Proteasome inhibitor did not inhibit DTX1-directed PKCθ degradation, but instead DTX1 directed the relocalization of PKCθ into the lysosomal pathway. In addition, DTX1 interacted with Cbl-b and increased the protein levels of Cbl-b. We further demonstrated the possibility that, through the downregulation of PKCθ, DTX1 prevented PKCθ-induced Cbl-b degradation and increased Cbl-b protein stability. Our results suggest the coordination between E3 ligases during T cell anergy; DTX1 acts with Cbl-b to assure a more extensive silencing of PKCθ, whereas DTX1-mediated PKCθ degradation further stabilizes Cbl-b.


Asunto(s)
Proteínas de Unión al ADN/genética , Isoenzimas/metabolismo , Proteína Oncogénica v-cbl/biosíntesis , Proteína Quinasa C/metabolismo , Proteolisis , Células TH1/inmunología , Animales , Calcimicina/farmacología , Línea Celular , Anergia Clonal , Proteínas de Unión al ADN/biosíntesis , Regulación hacia Abajo , Células HEK293 , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Células Jurkat , Activación de Linfocitos/inmunología , Lisosomas/inmunología , Ratones , Ratones Noqueados , Proteína Oncogénica v-cbl/genética , Fosfolipasa C gamma/biosíntesis , Fosfolipasa C gamma/metabolismo , Inhibidores de Proteasoma/farmacología , Proteína Quinasa C/biosíntesis , Proteína Quinasa C/genética , Proteína Quinasa C-theta , Interferencia de ARN , ARN Interferente Pequeño , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteína Tirosina Quinasa ZAP-70/biosíntesis
7.
Blood ; 121(16): 3185-94, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23430110

RESUMEN

The functional activities of the tumor suppressor promyelocytic leukemia protein (PML) are mostly associated with its nuclear location. In the present study, we discovered an unexpected role of PML in NLRP3 inflammasome activation. In PML-deficient macrophages, the production of IL-1ß was strongly impaired. The expression of pro-IL-1ß, NLRP3, ASC, and procaspase-1 was not affected in Pml(-/-) macrophages. PML deficiency selectively reduced the processing of procaspase-1. We further showed that PML is required for the assembly of the NLRP3 inflammasome in reconstitution experiment. All PML isoforms were capable of stimulating NLRP3 inflammasome activation. In Pml(-/-) macrophages, the generation of reactive oxygen species and release of mitochondrial DNA were decreased. The involvement of PML in inflammasome activation constitutes an important activity of PML and reveals a new mechanism underlying the inflammasome activation. In addition, downregulation of PML by arsenic trioxide suppressed monosodium urate (MSU)-induced IL-1ß production, suggesting that targeting to PML could be used to treat NLRP3 inflammasome-associated diseases.


Asunto(s)
Proteínas Portadoras/inmunología , Inflamasomas/inmunología , Proteínas Nucleares/inmunología , Factores de Transcripción/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Trióxido de Arsénico , Arsenicales/farmacología , Proteínas Portadoras/genética , Caspasa 1/inmunología , Línea Celular , Células Cultivadas , ADN Mitocondrial/inmunología , Regulación hacia Abajo/efectos de los fármacos , Eliminación de Gen , Inhibidores de Crecimiento/farmacología , Humanos , Interleucina-1beta/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Nucleares/genética , Óxidos/farmacología , Proteína de la Leucemia Promielocítica , Especies Reactivas de Oxígeno/inmunología , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
8.
Apoptosis ; 19(2): 357-63, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24185831

RESUMEN

Death-associated protein kinase (DAPK) is a tumor suppressor and negatively regulates several activation signals. Consistent with its potential anti-inflammatory activity, DAPK promotes the formation of IFN-γ-activated inhibitor of translation (GAIT) complex that suppresses the translation of selected inflammatory genes. DAPK has been found to inhibit tumor necrosis factor-α (TNF-α)- or lipopolysaccharides (LPS)-induced NF-κB activation and pro-inflammatory cytokine expression. Inflammation is always associated with T cell activation, while DAPK attenuates T cell activation by a selective suppression in T cell receptor-triggered NF-κB activation. Recent studies, however, also reveal a contribution of DAPK to pro-inflammatory processes. DAPK is shown to mediate pro-inflammatory signaling downstream of TNF-α, LPS, IL-17, or IL-32. In addition, DAPK is required for the full formation of NLRP3 inflammasome, essential for the generation of IL-1ß and IL-18. These results suggest the complicated role of DAPK in the regulation of inflammation that is likely dependent on cell types and environmental cues.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Inflamación/metabolismo , Animales , Citocinas/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular/inmunología , Regulación hacia Abajo , Humanos , Inflamasomas/metabolismo , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
J Immunol ; 188(6): 2914-21, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22308310

RESUMEN

Syndecan is the major transmembrane proteoglycan in cells. Of the four syndecans, syndecan-1 is the dominant form expressed in multiple myeloma and is an indicator of poor prognosis. In the current study, we observed that early TRAIL-induced apoptotic processes were accompanied by cleavage of syndecan-1 intracellular region, and explored the possibility whether removal of syndecan-1 promotes apoptotic processes. We found that syndecan-1 knockdown by specific small interfering RNA in multiple myeloma enhanced TRAIL-induced apoptosis, even though the expression of TRAIL receptors and several apoptosis-associated molecules was unaffected. The enhanced TRAIL-mediated apoptosis in syndecan-1-deficient cells was not due to a decrease in surface heparan sulfate or a reduction in TRAIL receptor endocytosis. The increase in TRAIL-induced cell death was accompanied by an elevated caspase-8 activation and an enhanced formation of death-inducing signaling complexes, which could be attributed to an increased expression of TRAIL receptor O-glycosylation enzyme in syndecan-1-deficient cells. We also found that in H9 lymphoma and Jurkat cells, knockdown of the predominant syndecan member also led to an increase in Fas ligand-induced apoptosis. Our results demonstrate that syndecan plays a negative role in death receptor-mediated cell death, suggesting potential application of syndecan downregulation in the treatment of myeloma in combination with TRAIL.


Asunto(s)
Apoptosis/fisiología , Mieloma Múltiple/metabolismo , Sindecano-1/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/inmunología , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inmunoprecipitación , Mieloma Múltiple/inmunología , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Sindecano-1/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología
11.
Eur J Immunol ; 42(8): 2165-75, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22865050

RESUMEN

Paxillin is an adaptor protein associated with focal adhesion complex, and is activated by tyrosine phosphorylation through focal adhesion kinase (FAK) and Src kinase. Recent studies reveal that serine phosphorylation of paxillin by JNK and p38 MAPK is essential for cell migration or neurite extension, but their cellular targets remain unclear. In this study, we examined the requirement of paxillin phosphorylation by p38 MAPK or JNK in T-cell motility and activation using paxillin mutants at the respective phosphorylation sites, Ser85, and Ser178. (S85A)-paxillin, (S178A)-paxillin, or (S85A/S178A)-paxillin inhibited the motility of NIH/3T3 fibroblasts, but did not interfere with T-cell migration and integrin-mediated T-cell adhesion. In contrast, activation of T cells was effectively suppressed by (S85A/S178A)-paxillin. Transgenic (S85A/S178A)-paxillin expression inhibited T-cell proliferation and reduced the production of IL-2, IFN-γ, and IL-4. In searching for signals modulated by (S85A/S178A)-paxillin, we found that NFAT activation was specifically blocked by (S85A/S178A)-paxillin. This could be partly attributed to diminished stromal interaction molecule 1 (STIM1) expression and attenuated TCR-induced Ca(2+) influx. Our results demonstrate that dual phosphorylation of paxillin by JNK and p38 MAPK is essential for T-cell activation and suggest that NFAT is a functional target of the JNK/p38 phosphorylated paxillin.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factores de Transcripción NFATC/metabolismo , Paxillin/metabolismo , Linfocitos T/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células 3T3 , Animales , Canales de Calcio , Adhesión Celular/inmunología , Línea Celular , Quimiotaxis de Leucocito , Integrinas/metabolismo , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Interleucina-4/biosíntesis , Activación de Linfocitos , Glicoproteínas de Membrana/biosíntesis , Ratones , Factores de Transcripción NFATC/biosíntesis , Paxillin/genética , Fosforilación , Molécula de Interacción Estromal 1 , Linfocitos T/fisiología
12.
Blood ; 117(3): 960-70, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21041719

RESUMEN

Interleukin-1ß (IL-1ß) is critical for inflammation and control of infection. The production of IL-1ß depends on expression of pro-IL-1ß and inflammasome component induced by inflammatory stimuli, followed by assembly of inflammasome to generate caspase-1 for cleavage of pro-IL-1ß. Here we show that tumor suppressor death-associated protein kinase (DAPK) deficiency impaired IL-1ß production in macrophages. Generation of tumor necrosis factor-α in macrophages, in contrast, was not affected by DAPK knockout. Two tiers of defects in IL-1ß generation were found in DAPK-deficient macrophages: decreased pro-IL-1ß induction by some stimuli and reduced caspase-1 activation by all inflammatory stimuli examined. With a normal NLRP3 induction in DAPK-deficient macrophages, the diminished caspase-1 generation is attributed to impaired inflammasome assembly. There is a direct binding of DAPK to NLRP3, suggesting an involvement of DAPK in inflammasome formation. We further illustrated that the formation of NLRP3 inflammasome in situ induced by inflammatory signals was impaired by DAPK deficiency. Taken together, our results identify DAPK as a molecule required for full production of IL-1ß and functional assembly of the NLRP3 inflammasome. In addition, DAPK knockout reduced uric acid crystal-triggered peritonitis, suggesting that DAPK may serve as a target in the treatment of IL-1ß-associated autoinflammatory diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Adaptadoras de Señalización CARD , Proteínas Quinasas Dependientes de Calcio-Calmodulina/deficiencia , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Línea Celular , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular , Células HEK293 , Humanos , Immunoblotting , Inflamación/metabolismo , Interleucina-1beta/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteína con Dominio Pirina 3 de la Familia NLR , Unión Proteica , Interferencia de ARN , Transfección , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
13.
Sci Adv ; 8(45): eabn9912, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36367942

RESUMEN

Caspase-8 activity controls the switch from cell death to pyroptosis when apoptosis and necroptosis are blocked, yet how caspase-8 inactivation induces inflammasome assembly remains unclear. We show that caspase-8 inhibition via IETD treatment in Toll-like receptor (TLR)-primed Fadd-/-Ripk3-/- myeloid cells promoted interleukin-1ß (IL-1ß) and IL-18 production through inflammasome activation. Caspase-8, caspase-1/11, and functional GSDMD, but not NLRP3 or RIPK1 activity, proved essential for IETD-triggered inflammasome activation. Autophagy became prominent in IETD-treated Fadd-/-Ripk3-/- macrophages, and inhibiting it attenuated IETD-induced cell death and IL-1ß/IL-18 production. In contrast, inhibiting GSDMD or autophagy did not prevent IETD-induced septic shock in Fadd-/-Ripk3-/- mice, implying distinct death processes in other cell types. Cathepsin-B contributes to IETD-mediated inflammasome activation, as its inhibition or down-regulation limited IETD-elicited IL-1ß production. Therefore, the autophagy and cathepsin-B axis represents one of the pathways leading to atypical inflammasome activation when apoptosis and necroptosis are suppressed and capase-8 is inhibited in myeloid cells.


Asunto(s)
Inflamasomas , Interleucina-18 , Ratones , Animales , Inflamasomas/metabolismo , Caspasa 8/genética , Interleucina-18/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Autofagia
14.
FEBS J ; 288(17): 5021-5041, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33971084

RESUMEN

Inborn errors of immunity (IEIs) are a group of genetically defined disorders leading to defective immunity. Some IEIs have been linked to mutations of immune receptors or signaling molecules, resulting in defective signaling of respective cascades essential for combating specific pathogens. However, it remains incompletely understood why in selected IEIs, such as X-linked lymphoproliferative syndrome type 2 (XLP-2), hypo-immune response to specific pathogens results in persistent inflammation. Moreover, mechanisms underlying the generation of anticytokine autoantibodies are mostly unknown. Recently, IEIs have been associated with coronavirus disease 2019 (COVID-19), with a small proportion of patients that contract severe COVID-19 displaying loss-of-function mutations in genes associated with type I interferons (IFNs). Moreover, approximately 10% of patients with severe COVID-19 possess anti-type I IFN-neutralizing autoantibodies. Apart from IEIs that impair immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), SARS-CoV-2 encodes several proteins that suppress early type I IFN production. One primary consequence of the lack of type I IFNs during early SARS-CoV-2 infection is the increased inflammation associated with COVID-19. In XLP-2, resolution of inflammation rescued experimental subjects from infection-induced mortality. Recent studies also indicate that targeting inflammation could alleviate COVID-19. In this review, we discuss infection-induced inflammation in IEIs, using XLP-2 and COVID-19 as examples. We suggest that resolving inflammation may represent an effective therapeutic approach to these diseases.


Asunto(s)
COVID-19/genética , Interferón Tipo I/genética , Errores Innatos del Metabolismo/genética , SARS-CoV-2/patogenicidad , COVID-19/inmunología , COVID-19/virología , Humanos , Inmunidad/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/virología , Errores Innatos del Metabolismo/inmunología , SARS-CoV-2/inmunología , Virosis/genética , Virosis/inmunología , Virosis/virología
15.
Front Immunol ; 12: 686060, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34211474

RESUMEN

Toll-like receptor (TLR) signaling is critical for defense against pathogenic infection, as well as for modulating tissue development. Activation of different TLRs triggers common inflammatory responses such as cytokine induction. Here, we reveal differential impacts of TLR3 and TLR7 signaling on transcriptomic profiles in bone marrow-derived macrophages (BMDMs). Apart from self-regulation, TLR3, but not TLR7, induced expression of other TLRs, suggesting that TLR3 activation globally enhances innate immunity. Moreover, we observed diverse influences of TLR3 and TLR7 signaling on genes involved in methylation, caspase and autophagy pathways. We compared endogenous TLR3 and TLR7 by using CRISPR/Cas9 technology to knock in a dual Myc-HA tag at the 3' ends of mouse Tlr3 and Tlr7. Using anti-HA antibodies to detect endogenous tagged TLR3 and TLR7, we found that both TLRs display differential tissue expression and posttranslational modifications. C-terminal tagging did not impair TLR3 activity. However, it disrupted the interaction between TLR7 and myeloid differentiation primary response 88 (MYD88), the Tir domain-containing adaptor of TLR7, which blocked its downstream signaling necessary to trigger cytokine and chemokine expression. Our study demonstrates different properties for TLR3 and TLR7, and also provides useful mouse models for further investigation of these two RNA-sensing TLRs.


Asunto(s)
Epítopos/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/fisiología , Neuronas/metabolismo , Receptor Toll-Like 3/fisiología , Receptor Toll-Like 7/fisiología , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Epítopos/inmunología , Femenino , Perfilación de la Expresión Génica , Inmunidad Innata , Macrófagos/inmunología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Factor 88 de Diferenciación Mieloide/fisiología , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
16.
Nat Commun ; 11(1): 5005, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33024109

RESUMEN

Hypoxia-inducible factor 1α (HIF-1α) and HIF-2α are master transcription factors that regulate cellular responses to hypoxia, but the exact function in regulatory T (Treg) cells is controversial. Here, we show that Treg cell development is normal in mice with Foxp3-specific knockout (KO) of HIF-1α or HIF-2α. However, HIF-2α-KO (but not HIF-1α-KO) Treg cells are functionally defective in suppressing effector T cell-induced colitis and inhibiting airway hypersensitivity. HIF-2α-KO Treg cells have enhanced reprogramming into IL-17-secreting cells. We show crosstalk between HIF-2α and HIF-1α, and that HIF-2α represses HIF-1α expression. HIF-1α is upregulated in HIF-2α-KO Treg cells and further deletion of HIF-1α restores the inhibitory function of HIF-2α-KO Treg cells. Mice with Foxp3-conditional KO of HIF-2α are resistant to growth of MC38 colon adenocarcinoma and metastases of B16F10 melanoma. Together, these results indicate that targeting HIF-2α to destabilize Treg cells might be an approach for regulating the functional activity of Treg cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linfocitos T Reguladores/fisiología , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hiperreactividad Bronquial/genética , Linfocitos T CD4-Positivos/citología , Diferenciación Celular , Reprogramación Celular , Colitis/etiología , Colitis/patología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Femenino , Factores de Transcripción Forkhead/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-17/metabolismo , Masculino , Melanoma/genética , Melanoma/patología , Ratones Noqueados , Linfocitos T Reguladores/metabolismo
17.
Cell Death Dis ; 11(5): 305, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366830

RESUMEN

Death-associated protein kinase 1 (DAPK1, DAPk, DAPK) is known for its involvement in apoptosis and autophagy-associated cell death. Here, we identified an unexpected function of DAPK1 in suppressing necroptosis. DAPK1-deficiency renders macrophages and dendritic cells susceptible to necroptotic death. We also observed an inhibitory role for DAPK1 in necroptosis in HT-29 cells, since knockdown or knockout of DAPK1 in such cells increased their sensitivity to necroptosis. Increased necroptosis was associated with enhanced formation of the RIPK1-RIPK3-MLKL complex in these DAPK1-deficient cells. We further found that DAPK1-deficiency led to decreased MAPK activated kinase 2 (MK2) activation and reduced RIPK1 S321 phosphorylation, with this latter representing a critical step controlling necrosome formation. Most TNF signaling pathways, including ERK, JNK, and AKT, were not regulated by DAPK. In contrast, DAPK bound p38 MAPK and selectively promoted p38 MAPK activation, resulting in enhanced MK2 phosphorylation. Our results reveal a novel role for DAPK1 in inhibiting necroptosis and illustrate an unexpected selectivity for DAPK1 in promoting p38 MAPK-MK2 activation. Importantly, our study suggests that modulation of necroptosis and p38/MK2-mediated inflammation may be achieved by targeting DAPK1.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Necroptosis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Caspasa 8/metabolismo , Supervivencia Celular , Proteínas Quinasas Asociadas a Muerte Celular/deficiencia , Regulación hacia Abajo , Activación Enzimática , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Técnicas de Silenciamiento del Gen , Células HT29 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/enzimología , Células Mieloides/patología , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Choque Séptico/metabolismo , Choque Séptico/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa
18.
Mol Cell Biol ; 25(4): 1367-78, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15684388

RESUMEN

Deltex is known as a Notch signal mediator, but its physiological action mechanism is poorly understood. Here we identified a new regulatory role of Deltex in T-cell activation. Deltex expression was constitutive in resting T cells and was reduced upon T-cell receptor (TCR)-stimulated activation. The biological role of Deltex is supported by the enhanced T-cell activation when Deltex1 was down-regulated by small interfering RNA. Overexpression of Deltex1 suppressed T-cell activation but not the proximal TCR activation events. The impaired activation of mitogen-activated protein kinase by Deltex could be partly attributed to a selective down-regulation of MEKK1 protein in T cells. We further found that Deltex promoted degradation of the C-terminal catalytic fragment of MEKK1 [MEKK1(C)]. Deltex1 interacted directly with MEKK1(C) and stimulated the ubiquitination of MEKK1(C) as shown by in vivo and in vitro ubiquitination analysis. Therefore, MEKK1(C), the dominant form of MEKK1 in T cells, is a target of Deltex E3 ubiquitin ligase. Our results reveal a novel mechanism as to how Deltex selectively suppresses T-cell activation through degradation of a key signaling molecule, MEKK1.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/fisiología , Activación de Linfocitos/fisiología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Linfocitos T/metabolismo , Animales , Interleucina-2/metabolismo , Ratones , Células 3T3 NIH , Fosforilación , Desnaturalización Proteica/fisiología , ARN Interferente Pequeño/metabolismo , Linfocitos T/fisiología , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas
19.
J Leukoc Biol ; 104(5): 911-918, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29901858

RESUMEN

Hypoxia-inducible factor 1α (HIF-1α) regulates cellular responses to hypoxia. However, conflicting roles for HIF-1α in the functions of regulatory T cells (Tregs) have been reported. In this review, we summarize observations on the requirement for HIF-1α for FOXP3 expression and Tregs development, as well as for HIF-1α-mediated downregulation of FOXP3 and Tregs destabilization. We also examine the association of HIF-1α with Tregs under pathogenic conditions. Based on these findings, we suggest that HIF-1α mainly plays a detrimental role in the function and stability of Tregs and that HIF-1α is disposable for the development and suppressive function of Tregs.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead/biosíntesis , Regulación de la Expresión Génica/inmunología , Humanos
20.
Nat Commun ; 9(1): 463, 2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29386580

RESUMEN

X-linked lymphoproliferative syndrome type-2 (XLP-2) is a primary immunodeficiency disease attributed to XIAP mutation and is triggered by infection. Here, we show that mouse Xiap-/- regulatory T (Treg) cells and human XIAP-deficient Treg cells are defective in suppressive function. The Xiap-/- Treg cell defect is linked partly to decreased SOCS1 expression. XIAP binds SOCS1 and promotes SOCS1 stabilization. Foxp3 stability is reduced in Xiap-/- Treg cells. In addition, Xiap-/- Treg cells are prone to IFN-γ secretion. Transfer of wild-type Treg cells partly rescues infection-induced inflammation in Xiap-/- mice. Notably, inflammation-induced reprogramming of Xiap-/- Treg cells can be prevented by blockade of the IL-6 receptor (IL-6R), and a combination of anti-IL-6R and Xiap-/- Treg cells confers survival to inflammatory infection in Xiap-/- mice. Our results suggest that XLP-2 can be corrected by combination treatment with autologous iTreg (induced Treg) cells and anti-IL-6R antibody, bypassing the necessity to transduce Treg cells with XIAP.


Asunto(s)
Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Trastornos Linfoproliferativos/inmunología , Receptores de Interleucina-6/antagonistas & inhibidores , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead/metabolismo , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Noqueados , Estabilidad Proteica , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Linfocitos T Reguladores/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA