Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Immunity ; 41(4): 555-66, 2014 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-25367572

RESUMEN

Bcl-3 is an atypical member of the IκB family that modulates transcription in the nucleus via association with p50 (NF-κB1) or p52 (NF-κB2) homodimers. Despite evidence attesting to the overall physiologic importance of Bcl-3, little is known about its cell-specific functions or mechanisms. Here we demonstrate a T-cell-intrinsic function of Bcl-3 in autoimmunity. Bcl-3-deficient T cells failed to induce disease in T cell transfer-induced colitis and experimental autoimmune encephalomyelitis. The protection against disease correlated with a decrease in Th1 cells that produced the cytokines IFN-γ and GM-CSF and an increase in Th17 cells. Although differentiation into Th1 cells was not impaired in the absence of Bcl-3, differentiated Th1 cells converted to less-pathogenic Th17-like cells, in part via mechanisms involving expression of the RORγt transcription factor. Thus, Bcl-3 constrained Th1 cell plasticity and promoted pathogenicity by blocking conversion to Th17-like cells, revealing a unique type of regulation that shapes adaptive immunity.


Asunto(s)
Autoinmunidad/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Interferón gamma/biosíntesis , Proteínas Proto-Oncogénicas/inmunología , Células TH1/inmunología , Células Th17/inmunología , Factores de Transcripción/inmunología , Animales , Proteínas del Linfoma 3 de Células B , Diferenciación Celular/inmunología , Colitis/inmunología , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad p50 de NF-kappa B/inmunología , Subunidad p52 de NF-kappa B/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Proteínas Proto-Oncogénicas/genética , Células TH1/trasplante , Factores de Transcripción/genética
2.
J Immunol ; 205(3): 830-841, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32591397

RESUMEN

The BCR recognizes foreign Ags to initiate humoral immunity that needs isotype-switched Abs generated via class switch recombination (CSR); however, stimulating the BCR in the absence of costimulation (e.g., CD40) does not induce CSR; thus, it remains elusive whether and how the BCR induces CSR mechanistically. Autoreactive B cells can maintain anergy via unresponsiveness of their BCRs to self-antigens. However, it remains unknown what molecule(s) restrict BCR signaling strength for licensing BCR-induced CSR and whether deficiency of such molecule(s) disrupts autoreactive B cell anergy and causes B cell-mediated diseases by modulating BCR signaling. In this study, we employ mouse models to show that the BCR's capacity to induce CSR is restrained by B cell-intrinsic checkpoints TRAF3 and TRAF2, whose deletion in B cells enables the BCR to induce CSR in the absence of costimulation. TRAF3 deficiency permits BCR-induced CSR by elevating BCR-proximal signaling intensity. Furthermore, NF-κB2 is required for BCR-induced CSR in TRAF3-deficient B cells but not for CD40-induced or LPS-induced CSR, suggesting that TRAF3 restricts NF-κB2 activation to specifically limit the BCR's ability to induce CSR. TRAF3 deficiency also disrupts autoreactive B cell anergy by elevating calcium influx in response to BCR stimulation, leading to lymphoid organ disorders and autoimmune manifestations. We showed that TRAF3 deficiency-associated autoimmune phenotypes can be rectified by limiting BCR repertoires or attenuating BCR signaling strength. Thus, our studies highlight the importance of TRAF3-mediated restraint on BCR signaling strength for controlling CSR, B cell homeostasis, and B cell-mediated disorders.


Asunto(s)
Linfocitos B/inmunología , Anergia Clonal , Cambio de Clase de Inmunoglobulina/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Animales , Linfocitos B/citología , Ratones , Ratones Transgénicos , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/inmunología , Transducción de Señal/genética , Factor 2 Asociado a Receptor de TNF/genética , Factor 2 Asociado a Receptor de TNF/inmunología , Factor 3 Asociado a Receptor de TNF/genética
3.
Immunity ; 37(4): 721-34, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22940098

RESUMEN

Lymph node development during embryogenesis involves lymphotoxin-ß receptor engagement and subsequent differentiation of a poorly defined population of mesenchymal cells into lymphoid tissue organizer cells. Here, we showed that embryonic mesenchymal cells with characteristics of adipocyte precursors present in the microenvironment of lymph nodes gave rise to lymph node organizer cells. Signaling through the lymphotoxin-ß receptor controlled the fate of adipocyte precursor cells by blocking adipogenesis and instead promoting lymphoid tissue stromal cell differentiation. This effect involved activation of the NF-κB2-RelB signaling pathway and inhibition of the expression of the key adipogenic factors Pparγ and Cebpα. In vivo organogenesis assays show that embryonic and adult adipocyte precursor cells can migrate into newborn lymph nodes and differentiate into a variety of lymph node stromal cells. Thus, we propose that adipose tissues act as a source of lymphoid stroma for lymph nodes and other lymphoid structures associated with fat.


Asunto(s)
Adipocitos/inmunología , Ganglios Linfáticos/inmunología , Transducción de Señal , Adipocitos/citología , Animales , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Receptor beta de Linfotoxina/inmunología , Ratones , Subunidad p52 de NF-kappa B/inmunología , Subunidad p52 de NF-kappa B/metabolismo , Fenotipo , Células del Estroma/inmunología , Factor de Transcripción ReIB/inmunología , Factor de Transcripción ReIB/metabolismo
4.
J Immunol ; 203(7): 1867-1881, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31492745

RESUMEN

Marginal zone B cells (MZB) are a mature B cell subset that rapidly respond to blood-borne pathogens. Although the transcriptional changes that occur throughout MZB development are known, the corresponding epigenetic changes and epigenetic modifying proteins that facilitate these changes are poorly understood. The histone demethylase LSD1 is an epigenetic modifier that promotes plasmablast formation, but its role in B cell development has not been explored. In this study, a role for LSD1 in the development of B cell subsets was examined. B cell-conditional deletion of LSD1 in mice resulted in a decrease in MZB whereas follicular B cells and bone marrow B cell populations were minimally affected. LSD1 repressed genes in MZB that were normally upregulated in the myeloid and follicular B cell lineages. Correspondingly, LSD1 regulated chromatin accessibility at the motifs of transcription factors known to regulate splenic B cell development, including NF-κB motifs. The importance of NF-κB signaling was examined through an ex vivo MZB development assay, which showed that both LSD1-deficient and NF-κB-inhibited transitional B cells failed to undergo full MZB development. Gene expression and chromatin accessibility analyses of in vivo- and ex vivo-generated LSD1-deficient MZB indicated that LSD1 regulated the downstream target genes of noncanonical NF-κB signaling. Additionally LSD1 was found to interact with the noncanonical NF-κB transcription factor p52. Together, these data reveal that the epigenetic modulation of the noncanonical NF-κB signaling pathway by LSD1 is an essential process during the development of MZB.


Asunto(s)
Subgrupos de Linfocitos B/metabolismo , Epigénesis Genética/inmunología , Histona Demetilasas/inmunología , Subunidad p52 de NF-kappa B/inmunología , Transducción de Señal/inmunología , Animales , Subgrupos de Linfocitos B/citología , Histona Demetilasas/genética , Ratones , Ratones Noqueados , Subunidad p52 de NF-kappa B/genética , Transducción de Señal/genética
5.
J Allergy Clin Immunol ; 141(3): 1060-1073.e3, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28629746

RESUMEN

BACKGROUND: Autosomal dominant anhidrotic ectodermal dysplasia with immune deficiency (AD EDA-ID) is caused by heterozygous point mutations at or close to serine 32 and serine 36 or N-terminal truncations in IκBα that impair its phosphorylation and degradation and thus activation of the canonical nuclear factor κ light chain enhancer of activated B cells (NF-κB) pathway. The outcome of hematopoietic stem cell transplantation is poor in patients with AD EDA-ID despite achievement of chimerism. Mice heterozygous for the serine 32I mutation in IκBα have impaired noncanonical NF-κB activity and defective lymphorganogenesis. OBJECTIVE: We sought to establish genotype-phenotype correlation in patients with AD EDA-ID. METHODS: A disease severity scoring system was devised. Stability of IκBα mutants was examined in transfected cells. Immunologic, biochemical, and gene expression analyses were performed to evaluate canonical and noncanonical NF-κB signaling in skin-derived fibroblasts. RESULTS: Disease severity was greater in patients with IκBα point mutations than in those with truncation mutations. IκBα point mutants were expressed at significantly higher levels in transfectants compared with truncation mutants. Canonical NF-κB-dependent IL-6 secretion and upregulation of the NF-κB subunit 2/p100 and RELB proto-oncogene, NF-κB subunit (RelB) components of the noncanonical NF-κB pathway were diminished significantly more in patients with point mutations compared with those with truncations. Noncanonical NF-κB-driven generation of the transcriptionally active p100 cleavage product p52 and upregulation of CCL20, intercellular adhesion molecule 1 (ICAM1), and vascular cell adhesion molecule 1 (VCAM1), which are important for lymphorganogenesis, were diminished significantly more in LPS plus α-lymphotoxin ß receptor-stimulated fibroblasts from patients with point mutations compared with those with truncations. CONCLUSIONS: IκBα point mutants accumulate at higher levels compared with truncation mutants and are associated with more severe disease and greater impairment of canonical and noncanonical NF-κB activity in patients with AD EDA-ID.


Asunto(s)
Displasia Ectodérmica/genética , Displasia Ectodérmica/inmunología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Genotipo , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Transducción de Señal , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Quimiocina CCL20/genética , Quimiocina CCL20/inmunología , Displasia Ectodérmica/patología , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Células HEK293 , Humanos , Síndromes de Inmunodeficiencia/patología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Inhibidor NF-kappaB alfa/genética , Inhibidor NF-kappaB alfa/inmunología , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/inmunología , Mutación Puntual , Enfermedades de Inmunodeficiencia Primaria , Proto-Oncogenes Mas , Transducción de Señal/genética , Transducción de Señal/inmunología
6.
J Biol Chem ; 292(27): 11561-11571, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28507099

RESUMEN

CO2 is a physiological gas normally produced in the body during aerobic respiration. Hypercapnia (elevated blood pCO2 >≈50 mm Hg) is a feature of several lung pathologies, e.g. chronic obstructive pulmonary disease. Hypercapnia is associated with increased susceptibility to bacterial infections and suppression of inflammatory signaling. The NF-κB pathway has been implicated in these effects; however, the molecular mechanisms underpinning cellular sensitivity of the NF-κB pathway to CO2 are not fully elucidated. Here, we identify several novel CO2-dependent changes in the NF-κB pathway. NF-κB family members p100 and RelB translocate to the nucleus in response to CO2 A cohort of RelB protein-protein interactions (e.g. with Raf-1 and IκBα) are altered by CO2 exposure, although others are maintained (e.g. with p100). RelB is processed by CO2 in a manner dependent on a key C-terminal domain located in its transactivation domain. Loss of the RelB transactivation domain alters NF-κB-dependent transcriptional activity, and loss of p100 alters sensitivity of RelB to CO2 Thus, we provide molecular insight into the CO2 sensitivity of the NF-κB pathway and implicate altered RelB/p100-dependent signaling in the CO2-dependent regulation of inflammatory signaling.


Asunto(s)
Dióxido de Carbono/inmunología , Hipercapnia/inmunología , Subunidad p52 de NF-kappa B/inmunología , Transducción de Señal/inmunología , Factor de Transcripción ReIB/inmunología , Células A549 , Animales , Humanos , Hipercapnia/genética , Hipercapnia/patología , Ratones , Subunidad p52 de NF-kappa B/genética , Dominios Proteicos , Transducción de Señal/genética , Factor de Transcripción ReIB/genética , Transcripción Genética/genética , Transcripción Genética/inmunología
7.
J Immunol ; 196(6): 2591-601, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26851215

RESUMEN

BAFF is critical for the survival and maturation of mature B cells. BAFF, via BAFFR, activates multiple signaling pathways in B cells, including the alternative NF-κB pathway. The transcription factors RELB and NF-κB2 (p100/p52) are the downstream mediators of the alternative pathway; however, the B cell-intrinsic functions of these NF-κB subunits have not been studied in vivo using conditional alleles, either individually or in combination. We in this study report that B cell-specific deletion of relb led to only a slight decrease in the fraction of mature splenic B cells, whereas deletion of nfkb2 caused a marked reduction. This phenotype was further exacerbated upon combined deletion of relb and nfkb2 and most dramatically affected the maintenance of marginal zone B cells. BAFF stimulation, in contrast to CD40 activation, was unable to rescue relb/nfkb2-deleted B cells in vitro. RNA-sequencing analysis of BAFF-stimulated nfkb2-deleted versus normal B cells suggests that the alternative NF-κB pathway, in addition to its critical role in BAFF-mediated cell survival, may control the expression of genes involved in the positioning of B cells within the lymphoid microenvironment and in the establishment of T cell-B cell interactions. Thus, by ablating the downstream transcription factors of the alternative NF-κB pathway specifically in B cells, we identify in this study a critical role for the combined activity of the RELB and NF-κB2 subunits in B cell homeostasis that cannot be compensated for by the canonical NF-κB pathway under physiological conditions.


Asunto(s)
Linfocitos B/citología , Homeostasis/inmunología , Subunidad p52 de NF-kappa B/inmunología , FN-kappa B/inmunología , Transducción de Señal/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Separación Celular , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Factor de Transcripción ReIB/inmunología , Factor de Transcripción ReIB/metabolismo
8.
J Immunol ; 196(4): 1891-9, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26773153

RESUMEN

Although numerous studies have demonstrated a critical role for canonical NF-κB signaling in inflammation and disease, the function of the noncanonical NF-κB pathway remains ill-defined. In lung tissue from patients with acute respiratory distress syndrome, we identified increased expression of the noncanonical pathway component p100/p52. To investigate the effects of p52 expression in vivo, we generated a novel transgenic mouse model with inducible expression of p52 in Clara cell secretory protein-expressing airway epithelial cells. Although p52 overexpression alone did not cause significant inflammation, p52 overexpression caused increased lung inflammation, injury, and mortality following intratracheal delivery of Escherichia coli LPS. No differences in cytokine/chemokine expression were measured between p52-overexpressing mice and controls, but increased apoptosis of Clara cell secretory protein-positive airway epithelial cells was observed in transgenic mice after LPS stimulation. In vitro studies in lung epithelial cells showed that p52 overexpression reduced cell survival and increased the expression of several proapoptotic genes during cellular stress. Collectively, these studies demonstrate a novel role for p52 in cell survival/apoptosis of airway epithelial cells and implicate noncanonical NF-κB signaling in the pathogenesis of acute respiratory distress syndrome.


Asunto(s)
Apoptosis/inmunología , Subunidad p52 de NF-kappa B/inmunología , Síndrome de Dificultad Respiratoria/patología , Mucosa Respiratoria/patología , Animales , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Lipopolisacáridos/toxicidad , Ratones , Ratones Transgénicos , Subunidad p52 de NF-kappa B/biosíntesis , Neumonía/inmunología , Neumonía/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Síndrome de Dificultad Respiratoria/inmunología , Mucosa Respiratoria/inmunología , Transducción de Señal/inmunología , Regulación hacia Arriba
9.
J Immunol ; 193(1): 422-30, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899500

RESUMEN

The noncanonical NF-κB pathway induces processing of the NF-κB2 precursor protein p100, and thereby mediates activation of p52-containing NF-κB complexes. This pathway is crucial for B cell maturation and humoral immunity, but its role in regulating T cell function is less clear. Using mutant mice that express a nonprocessible p100, NF-κB2(lym1), we show that the noncanonical NF-κB pathway has a T cell-intrinsic role in regulating the pathogenesis of a T cell-mediated autoimmunity, experimental autoimmune encephalomyelitis (EAE). Although the lym1 mutation does not interfere with naive T cell activation, it renders the Th17 cells defective in the production of inflammatory effector molecules, particularly the cytokine GM-CSF. We provide evidence that p52 binds to the promoter of the GM-CSF-encoding gene (Csf2) and cooperates with c-Rel in the transactivation of this target gene. Introduction of exogenous p52 or GM-CSF to the NF-κB2(lym1) mutant T cells partially restores their ability to induce EAE. These results suggest that the noncanonical NF-κB pathway mediates induction of EAE by regulating the effector function of inflammatory T cells.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Subunidad p52 de NF-kappa B/inmunología , Células Th17/inmunología , Activación Transcripcional/inmunología , Animales , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Noqueados , Mutación , Subunidad p52 de NF-kappa B/genética , Proteínas Proto-Oncogénicas c-rel/genética , Proteínas Proto-Oncogénicas c-rel/inmunología , Células Th17/patología , Activación Transcripcional/genética
10.
J Immunol ; 193(10): 4914-4923, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25288570

RESUMEN

The capacity of dendritic cells (DC) to regulate adaptive immunity is controlled by their maturation state and lifespan. Although TNF is a well-known maturation and survival factor for DC, the role of the two TNFR, TNFR1 and TNFR2, in mediating these effects is poorly understood. By using unique TNF variants that selectively signal through TNFR1 and/or TNFR2, we demonstrate differential functions of TNFR in human monocyte-derived and blood CD1c(+) DC. Activation of TNFR1, but not TNFR2, efficiently induced DC maturation, as defined by enhanced expression of cell surface maturation markers (CD83, CD86, and HLA-DR) as well as enhanced T cell stimulatory capacity. In contrast, both TNFR1 and TNFR2 significantly protected DC against cell death, indicating that innate signals can promote DC survival in the absence of DC maturation. We further show differential activation of NF-κB signaling pathways by the TNFR: TNFR1 activated both the p65 and p52 pathways, whereas TNFR2 triggered p52, but not p65, activation. Accordingly, the p65 NF-κB pathway only played a role in the prosurvival effect of TNFR1. However, cell death protection through both TNFR was mediated through the Bcl-2/Bcl-xL pathway. Taken together, our data show that TNFR1, but not TNFR2, signaling induces DC maturation, whereas DC survival can be mediated independently through both TNFR. These data indicate differential but partly overlapping responses through TNFR1 and TNFR2 in both inflammatory and conventional DC, and they demonstrate that DC maturation and DC survival can be regulated through independent signaling pathways.


Asunto(s)
Células Dendríticas/inmunología , Monocitos/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Inmunidad Adaptativa , Antígenos CD/genética , Antígenos CD/inmunología , Antígeno B7-2/genética , Antígeno B7-2/inmunología , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula/inmunología , Proliferación Celular , Supervivencia Celular , Células Dendríticas/citología , Regulación de la Expresión Génica , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/inmunología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Monocitos/citología , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/inmunología , Cultivo Primario de Células , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Transducción de Señal , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Proteína bcl-X/genética , Proteína bcl-X/inmunología , Antígeno CD83
11.
Mol Cell Proteomics ; 13(12): 3250-69, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25106423

RESUMEN

Human respiratory syncytial virus is a major respiratory pathogen for which there are no suitable antivirals or vaccines. A better understanding of the host cell response to this virus may redress this problem. The present report concerns analysis of multiple independent biological replicates of control and 24 h infected lysates of A549 cells by two different proteomic workflows. One workflow involved fractionation of lysates by in-solution protein IEF and individual fractions were digested using trypsin prior to capillary HPLC-LTQ-OrbitrapXL-MS/MS. A second workflow involved digestion of whole cell lysates and analysis by nanoUltraHPLC-LTQ-OrbitrapElite-MS/MS. Both workflows resulted in the quantification of viral proteins exclusively in lysates of infected cells in the relative abundances anticipated from previous studies. Unprecedented numbers (3247 - 5010) of host cell protein groups were also quantified and the infection-specific regulation of a large number (191) of these protein groups was evident based on a stringent false discovery rate cut-off (<1%). Bioinformatic analyses revealed that most of the regulated proteins were potentially regulated by type I, II, and III interferon, TNF-α and noncanonical NF-κB2 mediated antiviral response pathways. Regulation of specific protein groups by infection was validated by quantitative Western blotting and the cytokine-/key regulator-specific nature of their regulation was confirmed by comparable analyses of cytokine treated A549 cells. Overall, it is evident that the workflows described herein have produced the most comprehensive proteomic characterization of host cell responses to human respiratory syncytial virus published to date. These workflows will form the basis for analysis of the impacts of specific genes of human respiratory syncytial virus responses of A549 and other cell lines using a gene-deleted version of the virus. They should also prove valuable for the analysis of the impact of other infectious agents on host cells.


Asunto(s)
Células Epiteliales/inmunología , Interacciones Huésped-Patógeno/inmunología , Proteoma/inmunología , Mucosa Respiratoria/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Extractos Celulares/química , Línea Celular , Cromatografía Líquida de Alta Presión , Células Epiteliales/metabolismo , Células Epiteliales/virología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Interferones/genética , Interferones/inmunología , Interferones/metabolismo , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/inmunología , Subunidad p52 de NF-kappa B/metabolismo , Fragmentos de Péptidos/análisis , Proteolisis , Proteoma/genética , Proteoma/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , Virus Sincitial Respiratorio Humano/metabolismo , Transducción de Señal , Espectrometría de Masas en Tándem , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
12.
Proc Natl Acad Sci U S A ; 110(13): 5127-32, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23493554

RESUMEN

Mutations in the TNF family of proteins have been associated with inherited forms of immune deficiency. Using an array-based sequencing assay, we identified an autosomal-dominant deficiency in TNF-like weak inducer of apoptosis (TWEAK; TNFSF12) in a kindred with recurrent infection and impaired antibody responses to protein and polysaccharide vaccines. This mutation occurs in the sixth exon of TWEAK and results in the amino acid substitution R145C within the conserved TNF-homology domain of the full-length protein. TWEAK mutant protein formed high molecular weight aggregates under nonreducing conditions, suggesting an increased propensity for intermolecular interactions. As a result, mutant TWEAK associated with B-cell-activating factor (BAFF) protein and down-regulated the BAFF-mediated activation of the noncanonical NF-κB pathway through inhibition of p100 processing to p52, resulting in inhibition of BAFF-dependent B-cell survival and proliferation. As BAFF mediates T-cell-independent isotype switching and B-cell survival, our data implicate TWEAK as a disease-susceptibility gene for a humoral immunodeficiency.


Asunto(s)
Linfocitos B/inmunología , Enfermedades Genéticas Congénitas/inmunología , Predisposición Genética a la Enfermedad , Síndromes de Inmunodeficiencia/inmunología , Mutación Missense , Factores de Necrosis Tumoral/inmunología , Adulto , Sustitución de Aminoácidos , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Linfocitos B/patología , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Niño , Preescolar , Citocina TWEAK , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Femenino , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Masculino , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/inmunología , Factores de Necrosis Tumoral/genética
13.
Proc Natl Acad Sci U S A ; 109(35): 14122-7, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22879398

RESUMEN

HIV modulates plasmacytoid dendritic cell (pDC) activation via Toll-like receptor 7, inducing type I IFN and inflammatory cytokines. Simultaneously, pDCs up-regulate the expression of indoleamine 2,3 dioxygenase (IDO), which is essential for the induction of regulatory T cells (Tregs), which function to down-modulate immune activation. Here we demonstrate the crucial importance of the noncanonical NF-κB pathway in the establishment of this immunoregulatory phenotype in pDCs. In response to HIV, the noncanonical NF-κB pathway directly induces IDO and involves the recruitment of TNF receptor-associated factor-3 to the Toll-like receptor/MyD88 complex, NF-κB-inducing kinase-dependent IκB kinase-α activation, and p52/RelB nuclear translocation. We also show that pDC-induced Tregs can inhibit conventional DC (cDC) maturation partially through cytotoxic T-lymphocyte antigen (CTLA)-4 engagement. Furthermore, CTLA-4 induces IDO in cDCs in a NF-κB-inducing kinase-dependent way. These CTLA-4-conditioned cDCs can in turn induce Treg differentiation in an IDO-dependent manner. Thus, the noncanonical NF-κB pathway is integral in controlling immunoregulatory phenotypes of both pDCs and cDCs.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Infecciones por VIH/inmunología , Subunidad p52 de NF-kappa B/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/virología , Expresión Génica/inmunología , Técnicas de Silenciamiento del Gen , Infecciones por VIH/metabolismo , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Quinasa I-kappa B/metabolismo , Inmunofenotipificación , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/virología , Factor 3 Asociado a Receptor de TNF/inmunología , Factor 3 Asociado a Receptor de TNF/metabolismo , Quinasa de Factor Nuclear kappa B
14.
J Biol Chem ; 288(35): 25066-25075, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23873932

RESUMEN

In this study we describe a previously unreported function for NFκB2, an NFκB family transcription factor, in antiviral immunity. NFκB2 is induced in response to poly(I:C), a mimic of viral dsRNA. Poly(I:C), acting via TLR3, induces p52-dependent transactivation of a reporter gene in a manner that requires the kinase activity of IκB kinase ε (IKKε) and the transactivating potential of RelA/p65. We identify a novel NFκB2 binding site in the promoter of the transcription factor Sp1 that is required for Sp1 gene transcription activated by poly(I:C). We show that Sp1 is required for IL-15 induction by both poly(I:C) and respiratory syncytial virus, a response that also requires NFκB2 and IKKε. Our study identifies NFκB2 as a target for IKKε in antiviral immunity and describes, for the first time, a role for NFκB2 in the regulation of gene expression in response to viral infection.


Asunto(s)
Quinasa I-kappa B/inmunología , Interleucina-15/metabolismo , Subunidad p52 de NF-kappa B/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Factor de Transcripción Sp1/inmunología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Inductores de Interferón/farmacología , Interleucina-15/genética , Ratones , Ratones Noqueados , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Poli I-C/farmacología , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/metabolismo , Elementos de Respuesta/genética , Elementos de Respuesta/inmunología , Factor de Transcripción Sp1/biosíntesis , Factor de Transcripción Sp1/genética , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 3/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Factor de Transcripción ReIA/metabolismo
15.
J Immunol ; 189(11): 5185-93, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23087406

RESUMEN

Anergy is a key physiological mechanism for restraining self-reactive B cells. A marked portion of peripheral B cells are anergic B cells that largely depend on BAFF for survival. BAFF activates the canonical and noncanonical NF-κB pathways, both of which are required for B cell survival. In this study we report that deficiency of the adaptor protein B cell lymphoma 10 (Bcl10) impaired the ability of BAFF to support B cell survival in vitro, and it specifically increased apoptosis in anergic B cells in vivo, dramatically reducing anergic B cells in mice. Bcl10-dependent survival of self-reactive anergic B cells was confirmed in the Ig hen egg lysozyme/soluble hen egg lysozyme double-transgenic mouse model of B cell anergy. Furthermore, we found that BAFF stimulation induced Bcl10 association with IκB kinase ß, a key component of the canonical NF-κB pathway. Consistently, Bcl10-deficient B cells were impaired in BAFF-induced IκBα phosphorylation and formation of nuclear p50/c-Rel complexes. Bcl10-deficient B cells also displayed reduced expression of NF-κB2/p100, severely reducing BAFF-induced nuclear accumulation of noncanonical p52/RelB complexes. Consequently, Bcl10-deficient B cells failed to express Bcl-x(L), a BAFF-induced NF-κB target gene. Taken together, these data demonstrate that Bcl10 controls BAFF-induced canonical NF-κB activation directly and noncanonical NF-κB activation indirectly. The BAFF-R/Bcl10/NF-κB signaling axis plays a critical role in peripheral B cell tolerance by regulating the survival of self-reactive anergic B cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Factor Activador de Células B/inmunología , Supervivencia Celular/inmunología , FN-kappa B/inmunología , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis , Factor Activador de Células B/genética , Proteína 10 de la LLC-Linfoma de Células B , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Supervivencia Celular/genética , Anergia Clonal , Regulación de la Expresión Génica/inmunología , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Ratones , Ratones Transgénicos , Muramidasa/inmunología , FN-kappa B/genética , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/inmunología , Fosforilación , Proteínas Proto-Oncogénicas c-rel/genética , Proteínas Proto-Oncogénicas c-rel/inmunología , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/inmunología , Proteína bcl-X/genética , Proteína bcl-X/inmunología
17.
Cell Immunol ; 272(2): 259-68, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22070873

RESUMEN

Tolerogenic dendritic cells represent a promising immunotherapy in autoimmunity. However, the molecular mechanisms that drive tolerogenic DCs functions are not well understood. We used GM-CSF or GM-CSF+IL-4 to generate tolerogenic (GM/DCs) and immunogenic (IL-4/DCs) BMDCs from NOD mice, respectively. GM/DCs were resistant to maturation, produced large amounts of IL-10 but not IL-12p70. GM/DCs displayed a reduced capacity to activate diabetogenic CD8(+) T-cells and were efficient to induce Tregs expansion and conversion. LPS stimulation triggered ERK1/2 activation that was sustained in GM/DCs but not in IL-4/DCs. ERK1/2 and AP-1 were involved in IL-10 production in GM/DCs but not in their resistance to maturation. Supershift analysis showed that NF-κB DNA binding complex contains p52 and p65 in GM/DCs, whereas it contains p52, p65 and RelB in IL-4/DCs. ChIP experiments revealed that p65 was recruited to IL-10 promoter following LPS stimulation of GM/DCs whereas its binding to IL-12p35 promoter was abolished. Our results suggest that immunoregulatory functions of GM/DCs are differentially regulated by ERK1/2, AP-1 and NF-κB pathways.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , FN-kappa B/inmunología , Factor de Transcripción AP-1/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Procesos de Crecimiento Celular/inmunología , Inmunoprecipitación de Cromatina/métodos , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-12/inmunología , Subunidad p35 de la Interleucina-12/genética , Subunidad p35 de la Interleucina-12/inmunología , Interleucina-4/inmunología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos NOD , Subunidad p52 de NF-kappa B/inmunología , Fenotipo , Regiones Promotoras Genéticas , Linfocitos T Reguladores/inmunología , Factor de Transcripción ReIA/inmunología , Factor de Transcripción ReIB/inmunología
18.
J Immunol ; 182(10): 6470-6, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19414801

RESUMEN

The oxygen-sensitive transcription factor hypoxia-inducible factor 1 (HIF-1) is known as the key regulator of hypoxia-induced gene expression. In addition to hypoxia, endotoxins such as bacterial LPS as well as proinflammatory cytokines have been shown to induce HIF-1, suggesting an integrative role for HIF-1 in conditions of hypoxia and inflammation. Cells can become tolerant to endotoxins by repetitive exposure to LPS. Herein, we studied the effect of endotoxin tolerance on HIF-1alpha accumulation and expression of HIF target genes in human monocytic cells and primary mouse peritoneal macrophages. Tolerant cells had reduced levels of HIF-1alpha under hypoxia, which was due to lowered levels of HIF-1alpha mRNA. HIF-1alpha expression is under control of NF-kappaB and increased DNA binding of the p52 subunit of NF-kappaB was found in tolerant cells. Knock down of p52 abolished the effects of endotoxin tolerance on HIF-1alpha expression, which suggest a negative regulatory role of p52 on HIF-1alpha transcription during endotoxin tolerance. Endotoxin tolerant cells showed diminished expression of the HIF target genes phosphoglycerate kinase 1 and adrenomedullin and reduced viability under hypoxic conditions, as well as a significantly reduced invasion. Peritoneal macrophages from endotoxin-tolerant mice made showed significantly reduced HIF-1alpha protein accumulation and subsequent HIF target gene expression. We conclude that endotoxin tolerance impairs HIF-1alpha induction which reduces the ability of monocytic cells to survive and function under hypoxic conditions.


Asunto(s)
Hipoxia de la Célula/inmunología , Endotoxinas/inmunología , Expresión Génica/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Tolerancia Inmunológica/inmunología , Monocitos/inmunología , Animales , Western Blotting , Línea Celular , Supervivencia Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Subunidad p52 de NF-kappa B/inmunología , Subunidad p52 de NF-kappa B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Allergy Clin Immunol ; 135(6): 1641-3, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25605273

Asunto(s)
Hormona Adrenocorticotrópica/deficiencia , Alopecia/inmunología , Inmunodeficiencia Variable Común/inmunología , Enfermedades del Sistema Endocrino/inmunología , Enfermedades Genéticas Congénitas/inmunología , Hipoglucemia/inmunología , Células Asesinas Naturales/inmunología , Mutación , Subunidad p52 de NF-kappa B/inmunología , Infecciones del Sistema Respiratorio/inmunología , Hormona Adrenocorticotrópica/genética , Hormona Adrenocorticotrópica/inmunología , Adulto , Alopecia/complicaciones , Alopecia/genética , Alopecia/patología , Linfocitos B/inmunología , Linfocitos B/patología , Secuencia de Bases , Inmunodeficiencia Variable Común/complicaciones , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/patología , Citotoxicidad Inmunológica , Enfermedades del Sistema Endocrino/complicaciones , Enfermedades del Sistema Endocrino/genética , Enfermedades del Sistema Endocrino/patología , Expresión Génica , Enfermedades Genéticas Congénitas/complicaciones , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Heterocigoto , Humanos , Hipoglucemia/complicaciones , Hipoglucemia/genética , Hipoglucemia/patología , Células Asesinas Naturales/patología , Masculino , Datos de Secuencia Molecular , Subunidad p52 de NF-kappa B/genética , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/patología , Linfocitos T/inmunología , Linfocitos T/patología
20.
J Clin Invest ; 116(11): 2964-71, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17039258

RESUMEN

NF-kappaB2-deficient mice have impaired T and B cell responses. We found, however, that in these mice there was severe infiltration of lymphocytes into multiple organs and increased activity of autoantibodies to peripheral tissue antigens in a manner similar to that of autoimmune regulator-deficient (Aire-deficient) mice. We further demonstrated that NF-kappaB2 was required for thymic Aire gene transcriptional regulation. The Nfkb2(-/-) thymus had distinct cortical and medullar structures, but reduced Aire and target gene expression of peripheral tissue antigens. Engraftment of Nfkb2(-/-) thymic stroma to nude mice recapitulated the autoimmune phenotype of the native Nfkb2(-/-) mice, confirming a key defect in central tolerance. Lymphotoxin beta receptor (LTbetaR) ligation-induced Aire gene expression was also largely abolished in the absence of NF-kappaB2. Thus NF-kappaB2 downstream of LTbetaR plays an important role in the regulation of central tolerance in an Aire-dependent manner.


Asunto(s)
Tolerancia Inmunológica/inmunología , Subunidad p52 de NF-kappa B/inmunología , Subunidad p52 de NF-kappa B/metabolismo , Transducción de Señal , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo , Animales , Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Regulación de la Expresión Génica , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Receptor beta de Linfotoxina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad p52 de NF-kappa B/deficiencia , Subunidad p52 de NF-kappa B/genética , Fenotipo , Receptores del Factor de Necrosis Tumoral/metabolismo , Células Madre/inmunología , Células Madre/efectos de la radiación , Células del Estroma/inmunología , Células del Estroma/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteína AIRE
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda