Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Biol Chem ; 299(12): 105455, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37949232

RESUMEN

The Akt-Rheb-mTORC1 pathway plays a crucial role in regulating cell growth, but the mechanisms underlying the activation of Rheb-mTORC1 by Akt remain unclear. In our previous study, we found that CBAP was highly expressed in human T-ALL cells and primary tumors, and its deficiency led to reduced phosphorylation of TSC2/S6K1 signaling proteins as well as impaired cell proliferation and leukemogenicity. We also demonstrated that CBAP was required for Akt-mediated TSC2 phosphorylation in vitro. In response to insulin, CBAP was also necessary for the phosphorylation of TSC2/S6K1 and the dissociation of TSC2 from the lysosomal membrane. Here we report that CBAP interacts with AKT and TSC2, and knockout of CBAP or serum starvation leads to an increase in TSC1 in the Akt/TSC2 immunoprecipitation complexes. Lysosomal-anchored CBAP was found to override serum starvation and promote S6K1 and 4EBP1 phosphorylation and c-Myc expression in a TSC2-dependent manner. Additionally, recombinant CBAP inhibited the GAP activity of TSC2 complexes in vitro, leading to increased Rheb-GTP loading, likely due to the competition between TSC1 and CBAP for binding to the HBD domain of TSC2. Overexpression of the N26 region of CBAP, which is crucial for binding to TSC2, resulted in a decrease in mTORC1 signaling and an increase in TSC1 association with the TSC2/AKT complex, ultimately leading to increased GAP activity toward Rheb and impaired cell proliferation. Thus, we propose that CBAP can modulate the stability of TSC1-TSC2 as well as promote the translocation of TSC1/TSC2 complexes away from lysosomes to regulate Rheb-mTORC1 signaling.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas de la Membrana , Proteínas Proto-Oncogénicas c-akt , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Humanos , Proliferación Celular , Guanosina Trifosfato/metabolismo , Inmunoprecipitación , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo
2.
Nat Immunol ; 12(2): 144-50, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21170027

RESUMEN

Transcription factor NF-κB and its activating kinase IKKß are associated with inflammation and are believed to be critical for innate immunity. Despite the likelihood of immune suppression, pharmacological blockade of IKKß-NF-κB has been considered as a therapeutic strategy. However, we found neutrophilia in mice with inducible deletion of IKKß (Ikkß(Δ) mice). These mice had hyperproliferative granulocyte-macrophage progenitors and pregranulocytes and a prolonged lifespan of mature neutrophils that correlated with the induction of genes encoding prosurvival molecules. Deletion of interleukin 1 receptor 1 (IL-1R1) in Ikkß(Δ) mice normalized blood cellularity and prevented neutrophil-driven inflammation. However, Ikkß(Δ)Il1r1(-/-) mice, unlike Ikkß(Δ) mice, were highly susceptible to bacterial infection, which indicated that signaling via IKKß-NF-κB or IL-1R1 can maintain antimicrobial defenses in each other's absence, whereas inactivation of both pathways severely compromises innate immunity.


Asunto(s)
Infecciones Bacterianas/inmunología , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Quinasa I-kappa B/metabolismo , Interleucina-1beta/metabolismo , Neutrófilos/metabolismo , Animales , Recuento de Células , Procesos de Crecimiento Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Susceptibilidad a Enfermedades , Células Progenitoras de Granulocitos y Macrófagos/inmunología , Células Progenitoras de Granulocitos y Macrófagos/patología , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Inmunidad Innata/genética , Interleucina-1beta/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Neutrófilos/inmunología , Neutrófilos/patología , Receptores Tipo I de Interleucina-1/genética , Regulación hacia Arriba/genética
3.
Cancer Immunol Immunother ; 71(3): 705-718, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34374812

RESUMEN

BACKGROUND: A major feature of the microenvironment in pancreatic ductal adenocarcinoma (PDAC) is the significant amount of extracellular matrix produced by pancreatic stellate cells (PSCs), which have been reported to enhance the invasiveness of pancreatic cancer cells and negatively impact the prognosis. METHODS: We analyzed the data from two publicly available microarray datasets deposited in the Gene Expression Omnibus and found candidate genes that were differentially expressed in PDAC cells with metastatic potential and PDAC cells cocultured with PSCs. We studied the interaction between PDAC cells and PSCs in vitro and verified our finding with the survival data of patients with PDAC from the website of The Human Protein Atlas. RESULTS: We found that PSCs stimulated PDAC cells to secrete S100A9, which attracted circulatory monocytes into cancer tissue and enhanced the expression of programmed death-ligand 1 (PD-L1) on macrophages. When analyzing the correlation of S100A9 and PD-L1 expression with the clinical outcomes of patients with PDAC, we ascertained that high expression of S100A9 and PD-L1 was associated with poor survival in patients with PDAC. CONCLUSIONS: PSCs stimulated PDAC cells to secrete S100A9, which acts as a chemoattractant to attract circulatory monocytes into cancer microenvironment and induces expression of PD-L1 on macrophages. High expression of S100A9 and PD-L1 was associated with worse overall survival in a cohort of patients with PDAC.


Asunto(s)
Calgranulina B/genética , Carcinoma Ductal Pancreático/etiología , Carcinoma Ductal Pancreático/metabolismo , Comunicación Celular , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/metabolismo , Células del Estroma/metabolismo , Biomarcadores , Calgranulina B/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Comunicación Celular/genética , Comunicación Celular/inmunología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/patología , Pronóstico , Interferencia de ARN , Células del Estroma/patología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
5.
PLoS Pathog ; 13(7): e1006485, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28671985

RESUMEN

Inflammasome is an intracellular protein complex that serves as cytosolic pattern recognition receptor (PRR) to engage with pathogens and to process cytokines of the interleukin-1 (IL-1) family into bioactive molecules. It has been established that interleukin-1ß (IL-1ß) is important to host defense against Histoplasma capsulatum infection. However, the detailed mechanism of how H. capsulatum induces inflammasome activation leading to IL-1ß production has not been studied. Here, we showed in dendritic cells (DCs) that H. capsulatum triggers caspase-1 activation and IL-1ß production through NLRP3 inflammasome. By reciprocal blocking of Dectin-1 or Dectin-2 in single receptor-deficient DCs and cells from Clec4n-/-, Clec7a-/-, and Clec7a-/-Clec4n-/- mice, we discovered that while Dectin-2 operates as a primary receptor, Dectin-1 serves as a secondary one for NLRP3 inflammasome. In addition, both receptors trigger Syk-JNK signal pathway to activate signal 1 (pro-IL-1ß synthesis) and signal 2 (activation of caspase-1). Results of pulmonary infection with H. capsulatum showed that CD103+ DCs are one of the major producers of IL-1ß and Dectin-2 and Dectin-1 double deficiency abolishes their IL-1ß response to the fungus. While K+ efflux and cathepsin B (but not ROS) function as signal 2, viable but not heat-killed H. capsulatum triggers profound lysosomal rupture leading to cathepsin B release. Interestingly, cathepsin B release is regulated by ERK/JNK downstream of Dectin-2 and Dectin-1. Our study demonstrates for the first time the unique roles of Dectin-2 and Dectin-1 in triggering Syk-JNK to activate signal 1 and 2 for H. capsulatum-induced NLRP3 inflammasome activation.


Asunto(s)
Células Dendríticas/inmunología , Histoplasma/fisiología , Histoplasmosis/inmunología , Inflamasomas/inmunología , Lectinas Tipo C/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Animales , Caspasa 1/genética , Caspasa 1/inmunología , Células Dendríticas/microbiología , Histoplasma/genética , Histoplasmosis/genética , Histoplasmosis/microbiología , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Lectinas Tipo C/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética
6.
Am J Pathol ; 188(4): 1031-1042, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29366678

RESUMEN

Highly pathogenic avian influenza A H5N1 virus causes pneumonia and acute respiratory distress syndrome in humans. Virus-induced excessive inflammatory response contributes to severe disease and high mortality rates. Galectin-3, a ß-galactoside-binding protein widely distributed in immune and epithelial cells, regulates various immune functions and modulates microbial infections. Here, we describe galectin-3 up-regulation in mouse lung tissue after challenges with the H5N1 influenza virus. We investigated the effects of endogenous galectin-3 on H5N1 infection and found that survival of galectin-3 knockout (Gal-3KO) mice was comparable with wild-type (WT) mice after infections. Compared with infected WT mice, infected Gal-3KO mice exhibited less inflammation in the lungs and reduced IL-1ß levels in bronchoalveolar lavage fluid. In addition, the bone marrow-derived macrophages (BMMs) from Gal-3KO mice exhibited reduced oligomerization of apoptosis-associated speck-like proteins containing caspase-associated recruitment domains and secreted less IL-1ß compared with BMMs from WT mice. However, similar levels of the inflammasome component of nucleotide oligomerization domain-like receptor protein 3 (NLRP3) were observed in two genotypes of BMMs. Co-immunoprecipitation data indicated galectin-3 and NLRP3 interaction in BMMs infected with H5N1. An association was also observed between galectin-3 and NLRP3/apoptosis-associated speck-like proteins containing caspase-associated recruitment domain complex. Combined, our results suggest that endogenous galectin-3 enhances the effects of H5N1 infection by promoting host inflammatory responses and regulating IL-1ß production by macrophages via interaction with NLRP3.


Asunto(s)
Aves/virología , Galectina 3/metabolismo , Subtipo H5N1 del Virus de la Influenza A/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neumonía/metabolismo , Neumonía/virología , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Perros , Células HEK293 , Humanos , Interleucina-1beta/metabolismo , Pulmón/patología , Pulmón/virología , Macrófagos/metabolismo , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Neumonía/patología , Piroptosis , Análisis de Supervivencia , Regulación hacia Arriba
7.
Mediators Inflamm ; 2018: 3523642, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30647534

RESUMEN

Psoriasis is a chronic inflammatory skin disorder that affects ~2%-3% of the worldwide population. Inappropriate and excessive activation of endosomal Toll-like receptors 7, 8, and 9 (TLRs 7-9) at the psoriatic site has been shown to play a pathogenic role in the onset of psoriasis. Macrophage is a major inflammatory cell type that can be differentiated into phenotypes M1 and M2. M1 macrophages produce proinflammatory cytokines, and M2 macrophages produce anti-inflammatory cytokines. The balance between these two types of macrophages determines the progression of various inflammatory diseases; however, whether macrophage polarization plays a role in psoriatic inflammation activated by endosomal TLRs has not been investigated. In this study, we investigated the function and mechanism of macrophages related to the pathogenic role of TLRs 7-9 in the progression of psoriasis. Analysis of clinical data in database revealed significantly increased expression of macrophage markers and inflammatory cytokines in psoriatic tissues over those in normal tissues. In animal studies, depletion of macrophages in mice ameliorated imiquimod, a TLR 7 agonist-induced psoriatic response. Imiquimod induced expression of genes and cytokines that are signature of M1 macrophage in the psoriatic lesions. In addition, treatment with this TLR 7 agonist shifted macrophages in the psoriatic lesions to a higher M1/M2 ratio. Both of the exogenous and endogenous TLR 7-9 ligands activated M1 macrophage polarization. M1 macrophages expressed higher levels of proinflammatory cytokines and TLRs 7-9 than M2 macrophages. These results suggest that by rendering macrophages into a more inflammatory status and capable of response to their ligands in the psoriatic sites, TLR 7-9 activation drives them to participate in endosomal TLR-activated psoriatic inflammation, resulting in an amplified inflammatory response. Our results also suggest that blocking M1 macrophage polarization could be a strategy which enables inhibition of psoriatic inflammation activated by these TLRs.


Asunto(s)
Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Psoriasis/inmunología , Psoriasis/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Biología Computacional , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Imidazoles/farmacología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Células THP-1 , Receptor Toll-Like 7/metabolismo , Receptores Toll-Like/metabolismo
8.
J Biol Chem ; 291(37): 19299-311, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27458013

RESUMEN

Innate immune responses are important for pathogen elimination and adaptive immune response activation. However, excess inflammation may contribute to immunopathology and disease progression (e.g. inflammation-associated hepatocellular carcinoma). Immune modulation resulting from pattern recognition receptor-induced responses is a potential strategy for controlling immunopathology and related diseases. This study demonstrates that the mycotoxin patulin suppresses Toll-like receptor- and RIG-I/MAVS-dependent cytokine production through GSH depletion, mitochondrial dysfunction, the activation of p62-associated mitophagy, and p62-TRAF6 interaction. Blockade of autophagy restored the immunosuppressive activity of patulin, and pharmacological activation of p62-dependent mitophagy directly reduced RIG-I-like receptor-dependent inflammatory cytokine production. These results demonstrated that p62-dependent mitophagy has an immunosuppressive role to innate immune response and might serve as a potential immunomodulatory target for inflammation-associated diseases.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Mitofagia/efectos de los fármacos , Micotoxinas/farmacología , Patulina/farmacología , Proteína Sequestosoma-1/inmunología , Animales , Células HEK293 , Humanos , Ratones , Mitofagia/inmunología , Células RAW 264.7
9.
Proc Natl Acad Sci U S A ; 110(40): 16079-84, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24043792

RESUMEN

The NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome, a multiprotein complex, triggers caspase-1 activation and maturation of the proinflammatory cytokines IL-1ß and IL-18 upon sensing a wide range of pathogen- and damage-associated molecules. Dysregulation of NLRP3 inflammasome activity contributes to the pathogenesis of many diseases, but its regulation remains poorly defined. Here we show that depletion of plasminogen activator inhibitor type 2 (PAI-2), a serine protease inhibitor, resulted in NLRP3- and ASC (apoptosis-associated Speck-like protein containing a C-terminal caspase recruitment domain)-dependent caspase-1 activation and IL-1ß secretion in macrophages upon Toll-like receptor 2 (TLR2) and TLR4 engagement. TLR2 or TLR4 agonist induced PAI-2 expression, which subsequently stabilized the autophagic protein Beclin 1 to promote autophagy, resulting in decreases in mitochondrial reactive oxygen species, NLRP3 protein level, and pro-IL-1ß processing. Likewise, overexpressing Beclin 1 in PAI-2-deficient cells rescued the suppression of NLRP3 activation in response to LPS. Together, our data identify a tier of TLR signaling in controlling NLRP3 inflammasome activation and reveal a cell-autonomous mechanism which inversely regulates TLR- or Escherichia coli-induced mitochondrial dysfunction, oxidative stress, and IL-1ß-driven inflammation.


Asunto(s)
Autofagia/inmunología , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Inhibidor 2 de Activador Plasminogénico/metabolismo , Proteolisis , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Caspasa 1/metabolismo , Citocinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Silenciador del Gen , Immunoblotting , Interleucina-1beta/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
PLoS Pathog ; 8(7): e1002812, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22829768

RESUMEN

Group B Streptococcus (GBS) is a leading cause of invasive bacterial infections in human newborns and immune-compromised adults. The pore-forming toxin (PFT) ß hemolysin/cytolysin (ßh/c) is a major virulence factor for GBS, which is generally attributed to its cytolytic functions. Here we show ßh/c has immunomodulatory properties on macrophages at sub-lytic concentrations. ßh/c-mediated activation of p38 MAPK drives expression of the anti-inflammatory and immunosuppressive cytokine IL-10, and inhibits both IL-12 and NOS2 expression in GBS-infected macrophages, which are critical factors in host defense. Isogenic mutant bacteria lacking ßh/c fail to activate p38-mediated IL-10 production in macrophages and promote increased IL-12 and NOS2 expression. Furthermore, targeted deletion of p38 in macrophages increases resistance to invasive GBS infection in mice, associated with impaired IL-10 induction and increased IL-12 production in vivo. These data suggest p38 MAPK activation by ßh/c contributes to evasion of host defense through induction of IL-10 expression and inhibition of macrophage activation, a new mechanism of action for a PFT and a novel anti-inflammatory role for p38 in the pathogenesis of invasive bacterial infection. Our studies suggest p38 MAPK may represent a new therapeutic target to blunt virulence and improve clinical outcome of invasive GBS infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Interleucina-10/biosíntesis , Macrófagos/inmunología , Macrófagos/microbiología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/patogenicidad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Inmunidad Innata , Interleucina-12/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Ratones , Óxido Nítrico Sintasa/biosíntesis , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/inmunología , Streptococcus agalactiae/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
11.
Nat Commun ; 15(1): 3791, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710704

RESUMEN

Fas-associated protein with death domain (FADD), procaspase-8, and cellular FLICE-inhibitory proteins (cFLIP) assemble through death-effector domains (DEDs), directing death receptor signaling towards cell survival or apoptosis. Understanding their three-dimensional regulatory mechanism has been limited by the absence of atomic coordinates for their ternary DED complex. By employing X-ray crystallography and cryogenic electron microscopy (cryo-EM), we present the atomic coordinates of human FADD-procaspase-8-cFLIP complexes, revealing structural insights into these critical interactions. These structures illustrate how FADD and cFLIP orchestrate the assembly of caspase-8-containing complexes and offer mechanistic explanations for their role in promoting or inhibiting apoptotic and necroptotic signaling. A helical procaspase-8-cFLIP hetero-double layer in the complex appears to promote limited caspase-8 activation for cell survival. Our structure-guided mutagenesis supports the role of the triple-FADD complex in caspase-8 activation and in regulating receptor-interacting protein kinase 1 (RIPK1). These results propose a unified mechanism for DED assembly and procaspase-8 activation in the regulation of apoptotic and necroptotic signaling across various cellular pathways involved in development, innate immunity, and disease.


Asunto(s)
Apoptosis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Caspasa 8 , Proteína de Dominio de Muerte Asociada a Fas , Humanos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/química , Caspasa 8/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/genética , Células HEK293 , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal
12.
Int J Biol Sci ; 19(9): 2897-2913, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37324951

RESUMEN

Vaccines are a powerful medical intervention for preventing epidemic diseases. Efficient inactivated or protein vaccines typically rely on an effective adjuvant to elicit an immune response and boost vaccine activity. In this study, we investigated the adjuvant activities of combinations of Toll-like receptor 9 (TLR9) and stimulator of interferon genes (STING) agonists in a SARS-CoV-2 receptor binding domain protein vaccine. Adjuvants formulated with a TLR9 agonist, CpG-2722, with various cyclic dinucleotides (CDNs) that are STING agonists increased germinal center B cell response and elicited humoral immune responses in immunized mice. An adjuvant containing CpG-2722 and 2'3'-c-di-AM(PS)2 effectively boosted the immune response to both intramuscularly and intranasally administrated vaccines. Vaccines adjuvanted with CpG-2722 or 2'3'-c-di-AM(PS)2 alone were capable of inducing an immune response, but a cooperative adjuvant effect was observed when both were combined. CpG-2722 induced antigen-dependent T helper (Th)1 and Th17 responses, while 2'3'-c-di-AM(PS)2 induced a Th2 response. The combination of CpG-2722 and 2'3'-c-di-AM(PS)2 generated a distinct antigen-dependent Th response profile characterized by higher Th1 and Th17, but lower Th2 responses. In dendritic cells, CpG-2722 and 2'3'-c-di-AM(PS)2 showed a cooperative effect on inducing expression of molecules critical for T cell activation. CpG-2722 and 2'3'-c-di-AM(PS)2 have distinct cytokine inducing profiles in different cell populations. The combination of these two agonists enhanced the expression of cytokines for Th1 and Th17 responses and suppressed the expression of cytokines for Th2 response in these cells. Thus, the antigen-dependent Th responses observed in the animals immunized with different vaccines were shaped by the antigen-independent cytokine-inducing profiles of their adjuvant. The expanded targeting cell populations, the increased germinal center B cell response, and reshaped T helper responses are the molecular bases for the cooperative adjuvant effect of the combination of TLR9 and STING agonists.


Asunto(s)
COVID-19 , Vacunas , Animales , Ratones , Vacunas contra la COVID-19 , Receptor Toll-Like 9/agonistas , SARS-CoV-2 , Oligodesoxirribonucleótidos/farmacología , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Citocinas , Inmunidad , Centro Germinal
13.
J Exp Med ; 220(8)2023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37158982

RESUMEN

Type I interferons are important antiviral cytokines, but prolonged interferon production is detrimental to the host. The TLR3-driven immune response is crucial for mammalian antiviral immunity, and its intracellular localization determines induction of type I interferons; however, the mechanism terminating TLR3 signaling remains obscure. Here, we show that the E3 ubiquitin ligase ZNRF1 controls TLR3 sorting into multivesicular bodies/lysosomes to terminate signaling and type I interferon production. Mechanistically, c-Src kinase activated by TLR3 engagement phosphorylates ZNRF1 at tyrosine 103, which mediates K63-linked ubiquitination of TLR3 at lysine 813 and promotes TLR3 lysosomal trafficking and degradation. ZNRF1-deficient mice and cells are resistant to infection by encephalomyocarditis virus and SARS-CoV-2 because of enhanced type I interferon production. However, Znrf1-/- mice have exacerbated lung barrier damage triggered by antiviral immunity, leading to enhanced susceptibility to respiratory bacterial superinfections. Our study highlights the c-Src-ZNRF1 axis as a negative feedback mechanism controlling TLR3 trafficking and the termination of TLR3 signaling.


Asunto(s)
COVID-19 , Interferón Tipo I , Animales , Ratones , Antivirales , SARS-CoV-2 , Receptor Toll-Like 3 , Genes src
14.
Opt Lett ; 37(19): 4056-8, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23027277

RESUMEN

A design of microfluidic devices is presented to integrate single-mode, liquid-core waveguides with microfluidic channels that generate and deliver disklike emulsion microdroplet cavities doped with an organic dye. The microcavity modes can be directly coupled to the liquid waveguide. Cavity-enhanced spontaneous emission was observed at the waveguide with low pump pulse energy.

15.
FASEB J ; 25(8): 2700-10, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21543763

RESUMEN

Autophagy is one of the downstream effector mechanisms for elimination of intracellular microbes following activation of the Toll-like receptors (TLRs). Although the detailed molecular mechanism for this cellular process is still unclear, Beclin 1, a key molecule for autophagy, has been suggested to play a role. Heat shock protein 90 (Hsp90) is a molecular chaperone that regulates the stability of signaling proteins. Herein, we show that Hsp90 forms a complex with Beclin 1 through an evolutionarily conserved domain to maintain the stability of Beclin 1. In monocytic cells, geldanamycin (GA), an Hsp90 inhibitor, effectively promoted proteasomal degradation of Beclin 1 in a concentration-dependent (EC(50) 100 nM) and time-dependent (t(50) 2 h) manner. In contrast, KNK437/Hsp inhibitor I had no effect. Hsp90 specifically interacted with Beclin 1 but not with other adapter proteins in the TLR signalsome. Treatment of cells with GA inhibited TLR3- and TLR4-mediated autophagy. In addition, S. typhimurium infection-induced autophagy was blocked by GA treatment. This further suggested a role of the Hsp90/Beclin 1 in controlling autophagy in response to microbial infections. Taken together, our data revealed that by maintaining the homeostasis of Beclin 1, Hsp90 plays a novel role in TLR-mediated autophagy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Toll-Like/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/efectos de los fármacos , Autofagia/inmunología , Beclina-1 , Benzoquinonas/farmacología , Línea Celular , Evolución Molecular , Células HEK293 , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Inmunidad Innata , Interferón beta/metabolismo , Lactamas Macrocíclicas/farmacología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Modelos Biológicos , Complejos Multiproteicos , FN-kappa B/metabolismo , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Ubiquitinación
16.
Cancer Cell ; 6(3): 297-305, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15380520

RESUMEN

We used an experimental murine cancer metastasis model in which a colon adenocarcinoma cell line generates lung metastases, whose growth is stimulated in response to injection of bacterial lipopolysaccharide (LPS), to investigate the role of NF-kappaB in inflammation-induced tumor growth. We found that LPS-induced metastatic growth response in this model depends on both TNFalpha production by host hematopoietic cells and NF-kappaB activation in tumor cells. Inhibition of NF-kappaB in both colon and mammary carcinoma cells converts the LPS-induced growth response to LPS-induced tumor regression. The latter response is TNFalpha-independent, but depends on another member of the TNF superfamily, TRAIL, whose receptor is induced in NF-kappaB-deficient cancer cells.


Asunto(s)
Inflamación/patología , Neoplasias Pulmonares/enzimología , Glicoproteínas de Membrana/fisiología , FN-kappa B/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Adenocarcinoma/secundario , Animales , Proteínas Reguladoras de la Apoptosis , Neoplasias del Colon/patología , Activación Enzimática , Inflamación/inducido químicamente , Lipopolisacáridos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Ligando Inductor de Apoptosis Relacionado con TNF , Células Tumorales Cultivadas
17.
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
18.
Cell Rep ; 38(8): 110354, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35196483

RESUMEN

Excessive generation and accumulation of highly reactive oxidizing molecules causes oxidative stress and oxidative damage to cellular components. Accumulating evidence indicates that autophagy diminishes oxidative damage in cells and maintains redox homeostasis by degrading and recycling intracellular damaged components. Here, we show that TRAF6 E3 ubiquitin ligase and A20 deubiquitinase coordinate to regulate ATG9A ubiquitination and autophagy activation in cells responding to oxidative stress. The ROS-dependent TRAF6-mediated non-proteolytic, K48/63-linked ubiquitination of ATG9A enhances its association with Beclin 1 and the assembly of VPS34-UVRAG complex, thereby stimulating autophagy. Notably, expression of the ATG9A ubiquitination mutants impairs ROS-induced VPS34 activation and autophagy. We further find that lipopolysaccharide (LPS)-induced ROS production also stimulates TRAF6-mediated ATG9A ubiquitination. Ablation of ATG9A causes aberrant TLR4 endosomal trafficking and decreases IRF-3 phosphorylation in LPS-stimulated macrophages. Our findings provide important insights into how K48/K63-linked ubiquitination of ATG9A contributes to the regulation of oxidative stress-induced autophagy.


Asunto(s)
Factor 6 Asociado a Receptor de TNF , Ubiquitina-Proteína Ligasas , Autofagia/fisiología , Estrés Oxidativo , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
19.
J Exp Med ; 201(10): 1677-87, 2005 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-15897281

RESUMEN

Transcription factor, nuclear factor kappaB (NF-kappaB), is required for osteoclast formation in vivo and mice lacking both of the NF-kappaB p50 and p52 proteins are osteopetrotic. Here we address the relative roles of the two catalytic subunits of the IkappaB kinase (IKK) complex that mediate NF-kappaB activation, IKKalpha and IKKbeta, in osteoclast formation and inflammation-induced bone loss. Our findings point out the importance of the IKKbeta subunit as a transducer of signals from receptor activator of NF-kappaB (RANK) to NF-kappaB. Although IKKalpha is required for RANK ligand-induced osteoclast formation in vitro, it is not needed in vivo. However, IKKbeta is required for osteoclastogenesis in vitro and in vivo. IKKbeta also protects osteoclasts and their progenitors from tumor necrosis factor alpha-induced apoptosis, and its loss in hematopoietic cells prevents inflammation-induced bone loss.


Asunto(s)
Resorción Ósea/metabolismo , FN-kappa B/metabolismo , Osteoclastos/metabolismo , Precursores de Proteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Apoptosis , Resorción Ósea/genética , Resorción Ósea/patología , Proteínas Portadoras/metabolismo , Diferenciación Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Quinasa I-kappa B , Inflamación/genética , Inflamación/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Células Progenitoras Mieloides/metabolismo , FN-kappa B/genética , Subunidad p50 de NF-kappa B , Subunidad p52 de NF-kappa B , Precursores de Proteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
20.
Proc Natl Acad Sci U S A ; 105(22): 7803-8, 2008 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-18511561

RESUMEN

NOD2, a NOD-like receptor (NLR), is an intracellular sensor of bacterial muramyl dipeptide (MDP) that was suggested to promote secretion of the proinflammatory cytokine IL-1beta. Yet, the molecular mechanism by which NOD2 can stimulate IL-1beta secretion, and its biological significance were heretofore unknown. We found that NOD2 through its N-terminal caspase recruitment domain directly binds and activates caspase-1 to trigger IL-1beta processing and secretion in MDP-stimulated macrophages, whereas the C-terminal leucine-rich repeats of NOD2 prevent caspase-1 activation in nonstimulated cells. MDP challenge induces the association of NOD2 with another NLR protein, NALP1, and gel filtration analysis revealed the formation of a complex consisting of NOD2, NALP1, and caspase-1. Importantly, Bacillus anthracis infection induces IL-1beta secretion in a manner that depended on caspase-1 and NOD2. In vitro, Anthrax lethal toxin strongly potentiated IL-1beta secretion, and that response was NOD2 and caspase-1-dependent. Thus, NOD2 plays a key role in the B. anthracis-induced inflammatory response by being a critical mediator of IL-1beta secretion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carbunco/inmunología , Proteínas Reguladoras de la Apoptosis/metabolismo , Bacillus anthracis/inmunología , Caspasa 1/metabolismo , Interleucina-1beta/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Acetilmuramil-Alanil-Isoglutamina/inmunología , Animales , Caspasa 1/genética , Línea Celular , Cromatografía en Gel , Humanos , Inflamación/inmunología , Inflamación/microbiología , Macrófagos/inmunología , Ratones , Ratones Endogámicos , Mutación , Proteína Adaptadora de Señalización NOD2/genética , Mapeo de Interacción de Proteínas , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA