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













Base de datos
Intervalo de año de publicación
1.
Cell Mol Immunol ; 18(1): 230-242, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32203192

RESUMEN

The exact relationships between group 2 innate lymphoid cells (ILC2s) and Th2 cells in type 2 pathology, as well as the mechanisms that restrain the responses of these cells, remain poorly defined. Here we examined the roles of ILC2s and Th2 cells in type 2 lung pathology in vivo using germline and conditional Relb-deficient mice. We found that mice with germline deletion of Relb (Relb-/-) spontaneously developed prominent type 2 pathology in the lung, which contrasted sharply with mice with T-cell-specific Relb deletion (Relbf/fCd4-Cre), which were healthy with no observed autoimmune pathology. We also found that in contrast to wild-type B6 mice, Relb-deficient mice showed markedly expanded ILC2s but not ILC1s or ILC3s. Moreover, adoptive transfer of naive CD4+ T cells into Rag1-/-Relb-/- hosts induced prominent type 2 lung pathology, which was inhibited by depletion of ILC2s. Mechanistically, we showed that Relb deletion led to enhanced expression of Bcl11b, a key transcription factor for ILC2s. We concluded that RelB plays a critical role in restraining ILC2s, primarily by suppressing Bcl11b activity, and consequently inhibits type 2 lung pathology in vivo.


Asunto(s)
Proteínas de Homeodominio/fisiología , Inmunidad Innata , Pulmón/patología , Linfocitos/patología , Proteínas Represoras/metabolismo , Células Th2/inmunología , Factor de Transcripción ReIB/fisiología , Proteínas Supresoras de Tumor/metabolismo , Traslado Adoptivo , Animales , Citocinas/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/genética
2.
Cell Immunol ; 349: 104043, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32044112

RESUMEN

Type I Interferon (IFN) signaling plays a critical role in dendritic cell (DC) development and functions. Inhibition of hyper type I IFN signaling promotes cDC2 subtype development. Relb is essential to development of cDC2 subtype and here we analyzed its effect on type I IFN signaling in DCs. We show that Relb suppresses the homeostatic type I IFN signaling in cDC2 cultures. TLR stimulation of FL-DCs led to RelB induction coinciding with fall in IFN signatures; conforming with the observation Relb expression reduced TLR stimulated IFN induction along with decrease in ISGs. Towards understanding mechanism, we show that effects of RelB are mediated by increased levels of IκBα. We demonstrate that RelB dampened antiviral responses by lowering ISG levels and the defect in cDC2 development in RelB null mice can be rescued in Ifnar1-/- background. Overall, we propose a novel role of RelB as a negative regulator of the type I IFN signaling pathway; fine tuning development of cDC2 subtype.


Asunto(s)
Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Inhibidor NF-kappaB alfa/fisiología , Factor de Transcripción ReIB/fisiología , Secuencia de Aminoácidos , Animales , Diferenciación Celular , Células Cultivadas , Cruzamientos Genéticos , Células Dendríticas/clasificación , Células Dendríticas/citología , Regulación de la Expresión Génica/inmunología , Ratones , Células 3T3 NIH , Virus de la Enfermedad de Newcastle/inmunología , Péptidos/farmacología , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/fisiología , Transducción de Señal/inmunología , Bazo/citología , Factor de Transcripción ReIB/deficiencia , Factor de Transcripción ReIB/genética , Carga Viral
3.
Am J Pathol ; 189(12): 2516-2530, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31539516

RESUMEN

NF-κB signals through canonical transcription factor p65 (RelA)/p50 and noncanonical avian reticuloendotheliosis viral oncogene related B (RelB)/p52 pathways. The RelA/p50 is involved in basal and inflammatory lymphangiogenesis. However, the role of RelB/p52 in lymphatic vessel biology is unknown. Herein, we investigated changes in lymphatic vessels (LVs) in mice deficient in noncanonical NF-κB signaling and the function of RelB in lymphatic endothelial cells (LECs). LVs were examined in Relb-/-, p52-/-, or control mice, and the gene expression profiles in LECs with RelB knockdown. Relb-/-, but not p52-/-, mice exhibited multiple LV abnormalities. They include the following: i) increased capillary vessel diameter, ii) reduced smooth muscle cell (SMC) coverage of mature vessels, iii) leakage, and iv) loss of active and passive lymphatic flow. Relb-/- mature LVs had thinner vessel walls, more apoptotic LECs and SMCs, and fewer LEC junctions. RelB knockdown LECs had decreased growth, survival, and adhesion, and dysregulated signaling pathways involving these cellular events. These results suggest that Relb-/- mice have abnormal LVs, mainly in mature vessels with reduced SMC coverage, leakage, and loss of contractions. RelB knockdown in LECs leads to reduced growth, survival, and adhesion. RelB plays a vital role in LEC-mediated LV maturation and function.


Asunto(s)
Proliferación Celular , Células Endoteliales/patología , Vasos Linfáticos/patología , Factor de Transcripción ReIB/fisiología , Animales , Apoptosis , Movimiento Celular , Células Cultivadas , Células Endoteliales/metabolismo , Vasos Linfáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B , Transducción de Señal
4.
Proc Natl Acad Sci U S A ; 113(32): 9063-8, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27457956

RESUMEN

The NF-κB signaling cascade relays external signals essential for B-cell growth and survival. This cascade is frequently hijacked by cancers that arise from the malignant transformation of germinal center (GC) B cells, underscoring the importance of deciphering the function of NF-κB in these cells. The NF-κB signaling cascade is comprised of two branches, the canonical and alternative NF-κB pathways, mediated by distinct transcription factors. The expression and function of the transcription factors of the alternative pathway, RELB and NF-κB2, in late B-cell development is incompletely understood. Using conditional deletion of relb and nfkb2 in GC B cells, we here report that ablation of both RELB and NF-κB2, but not of the single transcription factors, resulted in the collapse of established GCs. RELB/NF-κB2 deficiency in GC B cells was associated with impaired cell-cycle entry and reduced expression of the cell-surface receptor inducible T-cell costimulator ligand that promotes optimal interactions between B and T cells. Analysis of human tonsillar tissue revealed that plasma cells and their precursors in the GC expressed high levels of NF-κB2 relative to surrounding lymphocytes. Accordingly, deletion of nfkb2 in murine GC B cells resulted in a dramatic reduction of antigen-specific antibody-secreting cells, whereas deletion of relb had no effect. These results demonstrate that the transcription factors of the alternative NF-κB pathway control distinct stages of late B-cell development, which may have implications for B-cell malignancies that aberrantly activate this pathway.


Asunto(s)
Linfocitos B/fisiología , Centro Germinal/fisiología , FN-kappa B/fisiología , Factores de Transcripción/fisiología , Animales , Antígenos CD40/fisiología , Células Cultivadas , Humanos , Ratones , Transducción de Señal/fisiología , Factor de Transcripción ReIB/fisiología
5.
Cell Death Dis ; 6: e1942, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26492375

RESUMEN

The plasticity of tumour-associated macrophages (TAMs) has implicated an influential role in hepatocellular carcinoma (HCC). Repolarisation of TAM towards M1 phenotype characterises an immune-competent microenvironment that favours tumour regression. To investigate the role and mechanism of TAM repolarisation in suppression of HCC by a natural compound baicalin, Orthotopic HCC implantation model was used to investigate the effect of baicalin on HCC; liposome-clodronate was introduced to suppress macrophage populations in mice; bone marrow-derived monocytes (BMDMs) were induced to unpolarised, M1-like, M2-like macrophages and TAM using different conditioned medium. We observed that oral administration of baicalin (50 mg/kg) completely blocked orthotopic growth of implanted HCC. Suppression of HCC by baicalin was diminished when mice macrophage was removed by clodronate treatment. Baicalin induced repolarisation of TAM to M1-like phenotype without specific toxicity to either phenotype of macrophages. Baicalin initiated TAM reprogramming to M1-like macrophage, and promoted pro-inflammatory cytokines production. Co-culturing of HCC cells with baicalin-treated TAMs resulted in reduced proliferation and motility in HCC. Baicalin had minimal effect on derivation of macrophage polarisation factors by HCC cells, while directly induced repolarisation of TAM and M2-like macrophage. This effect was associated with elevated autophagy, and transcriptional activation of RelB/p52 pathway. Suppression of autophagy or RelB abolished skewing of baicalin-treated TAM. Autophagic degradation of TRAF2 in baicalin-treated TAM might be responsible for RelB/p52 activation. Our findings unveil the essential role of TAM repolarisation in suppressive effect of baicalin on HCC, which requires autophagy-associated activation of RelB/p52.


Asunto(s)
Carcinoma Hepatocelular/patología , Flavonoides/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas Experimentales/patología , Subunidad p52 de NF-kappa B/fisiología , Factor de Transcripción ReIB/fisiología , Animales , Autofagia/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Polaridad Celular/efectos de los fármacos , Flavonoides/uso terapéutico , Interleucina-12/genética , Interleucina-12/metabolismo , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Subunidad p52 de NF-kappa B/metabolismo , Transducción de Señal , Factor de Transcripción ReIB/metabolismo , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
J Bone Miner Res ; 29(4): 866-77, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24115294

RESUMEN

RelA-mediated NF-κB canonical signaling promotes mesenchymal progenitor cell (MPC) proliferation, but inhibits differentiation of mature osteoblasts (OBs) and thus negatively regulates bone formation. Previous studies suggest that NF-κB RelB may also negatively regulate bone formation through noncanonical signaling, but they involved a complex knockout mouse model, and the molecular mechanisms involved were not investigated. Here, we report that RelB(-/-) mice develop age-related increased trabecular bone mass associated with increased bone formation. RelB(-/-) bone marrow stromal cells expanded faster in vitro and have enhanced OB differentiation associated with increased expression of the osteoblastogenic transcription factor, Runt-related transcription factor 2 (Runx2). In addition, RelB directly targeted the Runx2 promoter to inhibit its activation. Importantly, RelB(-/-) bone-derived MPCs formed bone more rapidly than wild-type cells after they were injected into a murine tibial bone defect model. Our findings indicate that RelB negatively regulates bone mass as mice age and limits bone formation in healing bone defects, suggesting that inhibition of RelB could reduce age-related bone loss and enhance bone repair.


Asunto(s)
Desarrollo Óseo/fisiología , Diferenciación Celular , Osteoblastos/citología , Factor de Transcripción ReIB/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa
7.
Leukemia ; 28(4): 871-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24056880

RESUMEN

In this study, we determined the respective roles of RelA and RelB NF-κB subunits in Epstein-Barr virus (EBV)-transformed B cells. Using different EBV-immortalized B-cell models, we showed that only RelA activation increased both survival and cell growth. RelB activity was induced secondarily to RelA activation and repressed RelA DNA binding by trapping the p50 subunit. Reciprocally, RelA activation repressed RelB activity by increasing expression of its inhibitor p100. To search for such reciprocal inhibition at the transcriptional level, we studied gene expression profiles of our RelA and RelB regulatable cellular models. Ten RelA-induced genes and one RelB-regulated gene, ARNTL2, were repressed by RelB and RelA, respectively. Apart from this gene, RelB signature was included in that of RelA Functional groups of RelA-regulated genes were for control of energy metabolism, genetic instability, protection against apoptosis, cell cycle and immune response. Additional functions coregulated by RelA and/or RelB were autophagy and plasma cell differentiation. Altogether, these results demonstrate a cross-inhibition between RelA and RelB and suggest that, in fine, RelB was subordinated to RelA. In the view of future drug development, RelA appeared to be pivotal in both classical and alternative activation pathways, at least in EBV-transformed B cells.


Asunto(s)
Transformación Celular Viral , Herpesvirus Humano 4/patogenicidad , Linfoma de Células B/etiología , Factor de Transcripción ReIA/fisiología , Factor de Transcripción ReIB/fisiología , Línea Celular , Humanos , FN-kappa B/fisiología , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIB/antagonistas & inhibidores , Factor de Transcripción ReIB/genética , Transcriptoma
8.
J Leukoc Biol ; 94(5): 941-51, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23922380

RESUMEN

RelB is one of the more unusual members of the NF-κB family. This family, arguably the best known group of transcription regulators, regulates an astonishing array of cell types and biological processes. This includes regulation of cell growth, differentiation and death by apoptosis, and the development and function of the innate and adaptive-immune system. RelB is best known for its roles in lymphoid development, DC biology, and noncanonical signaling. Within the last few years, however, surprising functions of RelB have emerged. The N-terminal leucine zipper motif of RelB, a motif unique among the NF-κB family, may associate with more diverse DNA sequences than other NF-κB members. RelB is capable of direct binding to the AhR that supports the xenobiotic-detoxifying pathway. RelB can regulate the circadian rhythm by directly binding to the BMAL partner of CLOCK. Finally, RelB also couples with bioenergy NAD(+) sensor SIRT1 to integrate acute inflammation with changes in metabolism and mitochondrial bioenergetics. In this review, we will explore these unique aspects of RelB, specifically with regard to its role in immunity.


Asunto(s)
Leucocitos/fisiología , Factor de Transcripción ReIB/fisiología , Ritmo Circadiano , Humanos , FN-kappa B/fisiología , Estabilidad Proteica , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Sirtuina 1/fisiología , Factor de Transcripción ReIB/química , Factor de Transcripción ReIB/genética
9.
Bing Du Xue Bao ; 29(1): 44-50, 2013 Jan.
Artículo en Chino | MEDLINE | ID: mdl-23547379

RESUMEN

Vpr, an auxiliary protein of HIV-1(Human immunodeficiency virus type 1), exerts important functions to promote viral replication and AIDS progression. In this study, we performed a yeast two-hybrid screening assay using human cDNA library to further investigate the molecular mechanism of various functions of Vpr RelB, a key protein in NF-kappaB signaling pathway, was identified as a Vpr interaction protein by co-immunoprecipitation. Further investigations indicated that RelB not only promoted the Vpr-mediated activation of NF-kappaB reporter gene, but also enhanced the transactivation of HIV LTR. Moreover, the results showed that RelB promoted Vpr-induced cell cycle G2/M arrest. Collectively, these results indicated that RelB might interact with Vpr and regulate its transcriptional activation and cell cycle arrest.


Asunto(s)
Puntos de Control del Ciclo Celular , División Celular , Fase G2 , Factor de Transcripción ReIB/fisiología , Activación Transcripcional , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/fisiología , Duplicado del Terminal Largo de VIH , Células HeLa , Humanos , FN-kappa B/genética
10.
Mol Endocrinol ; 26(8): 1356-69, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22734038

RESUMEN

Placental CRH may be part of a clock that governs the length of human gestation. The mechanism underlying differential regulation of CRH in the human placenta is poorly understood. We report here that constitutively activated RelB/nuclear factor-κB2 (NF-κB)-2 (p100/p52) acts as an endogenous stimulatory signal to regulate CRH by binding to an NF-κB enhancer of CRH gene promoter in the human placenta. Nuclear staining of NF-κB2 and RelB in villous syncytiotrophoblasts and cytotrophoblasts was coupled with cytoplasmic CRH in syncytial knots of cytotrophoblasts. Chromatin immunoprecipitation identified that CRH gene associated with both RelB and NF-κB2 (p52). Dexamethasone increased synthesis and nuclear translocation of RelB and NF-κB2 (p52) and their association with the CRH gene. In contrast, progesterone, a down-regulator of placental CRH, repressed NF-κB2 (p100) processing, nuclear translocation of RelB and NF-κB2 (p52), and their association with the CRH gene. Luciferase reporter assay determined that the NF-κB enhancer of CRH was sufficient to regulate transcriptional activity of a heterologous promoter in primary cytotrophoblasts. RNA interference-mediated repression of RelB or NF-κB2 resulted in significant inhibition of CRH at both transcriptional and translational levels and prevented the dexamethasone-mediated up-regulation of CRH transcription and translation. These results suggest that the noncanonical NF-κB pathway regulates CRH production in the human placenta and is responsible for the positive regulation of CRH by glucocorticoids.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Subunidad p52 de NF-kappa B/fisiología , Placenta/metabolismo , Factor de Transcripción ReIB/fisiología , Análisis de Varianza , Secuencia de Bases , Células Cultivadas , Hormona Liberadora de Corticotropina/genética , Dexametasona/farmacología , Elementos de Facilitación Genéticos , Femenino , Regulación de la Expresión Génica , Humanos , Leupeptinas/farmacología , Subunidad p52 de NF-kappa B/metabolismo , Placenta/citología , Embarazo , Progesterona/farmacología , Progesterona/fisiología , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Transducción de Señal , Factor de Transcripción ReIB/metabolismo , Transcripción Genética , Trofoblastos/metabolismo
11.
Cell Rep ; 1(4): 317-24, 2012 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-22545247

RESUMEN

Monocytes serve as a central defense system against infection and injury but can also promote pathological inflammatory responses. Considering the evidence that monocytes exist in at least two subsets committed to divergent functions, we investigated whether distinct factors regulate the balance between monocyte subset responses in vivo. We identified a microRNA (miRNA), miR-146a, which is differentially regulated both in mouse (Ly-6C(hi)/Ly-6C(lo)) and human (CD14(hi)/CD14(lo)CD16(+)) monocyte subsets. The single miRNA controlled the amplitude of the Ly-6C(hi) monocyte response during inflammatory challenge whereas it did not affect Ly-6C(lo) cells. miR-146a-mediated regulation was cell-intrinsic and depended on Relb, a member of the noncanonical NF-κB/Rel family, which we identified as a direct miR-146a target. These observations not only provide mechanistic insights into the molecular events that regulate responses mediated by committed monocyte precursor populations but also identify targets for manipulating Ly-6C(hi) monocyte responses while sparing Ly-6Clo monocyte activity.


Asunto(s)
MicroARNs/fisiología , Monocitos/fisiología , Factor de Transcripción ReIB/fisiología , Animales , Movimiento Celular , Proliferación Celular , Regulación de la Expresión Génica , Humanos , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Factor de Transcripción ReIB/metabolismo
12.
J Immunol ; 188(5): 2380-6, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22287708

RESUMEN

HMGB1 is a chromatin architectural protein that is released by dead or damaged cells at sites of tissue injury. Extracellular HMGB1 functions as a proinflammatory cytokine and chemoattractant for immune effector and progenitor cells. Previously, we have shown that the inhibitor of NF-κB kinase (IKK)ß- and IKKα-dependent NF-κB signaling pathways are simultaneously required for cell migration to HMGB1. The IKKß-dependent canonical pathway is needed to maintain expression of receptor for advanced glycation end products, the ubiquitously expressed receptor for HMGB1, but the target of the IKKα non-canonical pathway was not known. In this study, we show that the IKKα-dependent p52/RelB noncanonical pathway is critical to sustain CXCL12/SDF1 production in order for cells to migrate toward HMGB1. Using both mouse bone marrow-derived macrophages and mouse embryo fibroblasts (MEFs), it was observed that neutralization of CXCL12 by a CXCL12 mAb completely eliminated chemotaxis to HMGB1. In addition, the HMGB1 migration defect of IKKα KO and p52 KO cells could be rescued by adding recombinant CXCL12 to cells. Moreover, p52 KO MEFs stably transduced with a GFP retroviral vector that enforces physiologic expression of CXCL12 also showed near normal migration toward HMGB1. Finally, both AMD3100, a specific antagonist of CXCL12's G protein-coupled receptor CXCR4, and an anti-CXCR4 Ab blocked HMGB1 chemotactic responses. These results indicate that HMGB1-CXCL12 interplay drives cell migration toward HMGB1 by engaging receptors of both chemoattractants. This novel requirement for a second receptor-ligand pair enhances our understanding of the molecular mechanisms regulating HMGB1-dependent cell recruitment to sites of tissue injury.


Asunto(s)
Comunicación Autocrina/inmunología , Movimiento Celular/inmunología , Quimiocina CXCL12/biosíntesis , Proteína HMGB1/fisiología , Quinasa I-kappa B/fisiología , Subunidad p52 de NF-kappa B/fisiología , Transducción de Señal/inmunología , Factor de Transcripción ReIB/fisiología , Animales , Transformación Celular Neoplásica , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/fisiología , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/deficiencia , Ratones , Ratones Noqueados , Ratones Transgénicos , Subunidad p52 de NF-kappa B/biosíntesis , Subunidad p52 de NF-kappa B/deficiencia , Factor de Transcripción ReIB/biosíntesis , Células Tumorales Cultivadas
14.
PLoS One ; 5(7): e11875, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20686703

RESUMEN

Human Immunodeficiency Virus-1 (HIV-1)-associated neurocognitive disorder (HAND) is likely neuroinflammatory in origin, believed to be triggered by inflammatory and oxidative stress responses to cytokines and HIV protein gene products such as the HIV transactivator of transcription (Tat). Here we demonstrate increased messenger RNA for nuclear factor-kappa B (NF-kappaB) family member, transcription factor RelB, in the brain of doxycycline-induced Tat transgenic mice, and increased RelB synthesis in Tat-exposed microglial cells. Since genetic ablation of RelB in mice leads to multi-organ inflammation, we hypothesized that Tat-induced, newly synthesized RelB inhibits cytokine production by microglial cells, possibly through the formation of transcriptionally inactive RelB/RelA complexes. Indeed, tumor necrosis factor-alpha (TNFalpha) production in monocytes isolated from RelB deficient mice was significantly higher than in monocytes isolated from RelB expressing controls. Moreover, RelB overexpression in microglial cells inhibited Tat-induced TNFalpha synthesis in a manner that involved transcriptional repression of the TNFalpha promoter, and increased phosphorylation of RelA at serine 276, a prerequisite for increased RelB/RelA protein interactions. The Rel-homology-domain within RelB was necessary for this interaction. Overexpression of RelA itself, in turn, significantly increased TNFalpha promoter activity, an effect that was completely blocked by RelB overexpression. We conclude that RelB regulates TNFalpha cytokine synthesis by competitive interference binding with RelA, which leads to downregulation of TNFalpha production. Moreover, because Tat activates both RelB and TNFalpha in microglia, and because Tat induces inflammatory TNFalpha synthesis via NF-kappaB, we posit that RelB serves as a cryoprotective, anti-inflammatory, counter-regulatory mechanism for pathogenic NF-kappaB activation. These findings identify a novel regulatory pathway for controlling HIV-induced microglial activation and cytokine production that may have important therapeutic implications for the management of HAND.


Asunto(s)
Factor de Transcripción ReIB/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/fisiología , Animales , Encéfalo/metabolismo , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/metabolismo , Factor de Necrosis Tumoral alfa/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
15.
Ann N Y Acad Sci ; 1201: 129-36, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20649549

RESUMEN

Radiation therapy is in the front line for treatment of localized prostate cancer. However, a significant percentage of patients have radiation-resistant disease. The NF-kappaB pathway is an important factor for radiation resistance, and the classical (canonical) pathway is thought to confer protection of prostate cancer cells from ionizing radiation. Recently, the alternative (non-canonical) pathway, which is involved in prostate cancer aggressiveness, has also been shown to be important for radiation resistance in prostate cancer. The alternative NF-kappaB pathway component RelB protects prostate cancer cells from the detrimental effects of ionizing radiation, in part, by stimulating expression of the mitochondria-localized antioxidant enzyme manganese superoxide dismutase (MnSOD). Blocking RelB activation suppresses MnSOD expression and sensitizes prostate cancer cells to radiation. These results suggest that RelB-mediated modulation of the antioxidant capacity of prostate cancer cells is an important mechanism of radiation resistance. Therefore, targeting RelB activation may prove to be a valuable weapon in the oncologist's arsenal to defeat aggressive and radiation-resistant prostate cancer.


Asunto(s)
Apoptosis , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia , Superóxido Dismutasa/metabolismo , Factor de Transcripción ReIB/fisiología , Antioxidantes/metabolismo , Humanos , Masculino , Mitocondrias/metabolismo , Modelos Biológicos , FN-kappa B/metabolismo , Estrés Oxidativo , Neoplasias de la Próstata/patología , Radiación Ionizante , Radioterapia/métodos , Especies Reactivas de Oxígeno
16.
Biochem Pharmacol ; 77(4): 734-45, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18955032

RESUMEN

The discovery of the new crosstalk between the aryl hydrocarbon receptor (AhR) and the NF-kappaB subunit RelB may extend our understanding of the biological functions of the AhR and at the same time raises a number of questions, which will be addressed in this review. The characteristics of this interaction differ from that of AhR with RelA in that the latter appears to be mostly negative unlike the collaborative interactions of AhR/RelB. The AhR/RelB dimer is capable of binding to DNA response elements including the dioxin response element (DRE) as well as NF-kappaB binding sites supporting the activation of target genes of the AhR as well as NF-kappaB pathway. Further studies show that AhR/RelB complexes can be found not only in lymphoid cells but also in a human hepatoma cell line (HepG2) or breast cancer cell line (MDA-MB-231). RelB has been implicated in carcinogenesis of breast cancer for instance and RelB is known to be a critical factor for the function and differentiation of dendritic cells; interestingly the participation of AhR in both processes has been suggested recently, which offers the great potential to expand the scope of the physiological roles of the AhR. There is evidence indicating that RelB may serve as a pro-survival factor, including its ability to promote "inflammation resolution" besides the association of RelB with inflammatory disorders. Based on such information, a hypothesis has been proposed in this review that AhR together with RelB functions as a coordinator of inflammatory responses.


Asunto(s)
Receptor Cross-Talk/fisiología , Receptores de Hidrocarburo de Aril/fisiología , Factor de Transcripción ReIB/fisiología , Animales , Línea Celular Tumoral , Humanos , Ligandos , Neoplasias/etiología , Neoplasias/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptor Cross-Talk/efectos de los fármacos , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/metabolismo
17.
Biochem J ; 416(1): 117-27, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18620544

RESUMEN

RelB is the key component of the alternative NF-kappaB (nuclear factor kappaB) signalling pathway. However, RelB exerts also a negative effect via the recruitment of a DNMT1 (DNA methyltransferase 1)-Daxx (death domain-associated protein) complex to NF-kappaB target genes. Importantly, the molecular mechanisms which determine the functions of RelB are still largely unknown. In the present study, we aimed to analyse whether ubiquitination of RelB might be involved in the regulation of RelB. Indeed, RelB is constitutively polyubiquitinated in the B-cell lines Namalwa and 70Z/3. Although a PMA+ionomycin-induced augmentation of RelB polyubiquitination was linked to its proteasomal degradation in B-cells, the constitutive RelB polyubiquitination seems to affect non-proteasomal functions. Consistently, a significant RelB polyubiquitination in HEK (human embryonic kidney)-293 cells correlated with an augmentation of the transcriptional activity of RelB. Yet, neither nuclear localization nor DNA binding was enhanced by RelB polyubiquitination. Interestingly, basal RelB polyubiquitination depends neither on Lys(48) nor on Lys(63) conjugates, but might involve unconventional ubiquitin conjugates. Mapping of the ubiquitination target sites in RelB revealed the existence of various lysine residues, which serve as ubiquitination acceptors. However, only the substitution of Lys(273/274) and Lys(305/308) significantly decreased the basal RelB activity and the ubiquitin-induced augmentation of the RelB activity. Collectively, these results imply a dual role of RelB polyubiquitination for the stability and activity of this transcription factor.


Asunto(s)
Lisina/fisiología , Factor de Transcripción ReIB/fisiología , Ubiquitina/química , Ubiquitinación/fisiología , Línea Celular , Regulación de la Expresión Génica , Humanos , Riñón/embriología , Factor de Transcripción ReIB/química , Transcripción Genética , Ubiquitina/metabolismo
18.
Blood ; 112(3): 551-9, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18505785

RESUMEN

Nuclear factor-kappaB (NF-kappaB) transcription factors regulate B-cell development and survival. However, whether they also have a role during early steps of B-cell differentiation is largely unclear. Here, we show that constitutive activation of the alternative NF-kappaB pathway in p100(-/-) knockin mice resulted in a block of early B-cell development at the transition from the pre-pro-B to the pro-B-cell stage due to enhanced RelB activity. Expression of the essential B-cell transcription factors EBF and in particular Pax5 was reduced in p100(-/-) B-cell precursors in a RelB-dependent manner, resulting in reduced mRNA levels of B lineage-specific genes. Moreover, enhanced RelB function in p100(-/-) B-cell precursors was accompanied by increased expression of B lineage-inappropriate genes, such as C/EBP alpha, correlating with a markedly increased myeloid differentiation potential of p100(-/-) progenitor B cells. Ectopic expression of Pax5 in hematopoietic progenitors restored early B-cell development in p100(-/-) bone marrow, suggesting that impaired early B lymphopoiesis in mice lacking the p100 inhibitor may be due to down-regulation of Pax5 expression. Thus, tightly controlled p100 processing and RelB activation is essential for normal B lymphopoiesis and lymphoid/myeloid lineage decision in bone marrow.


Asunto(s)
Linfocitos B/citología , Linfopoyesis , Subunidad p52 de NF-kappa B/fisiología , Factor de Transcripción ReIB/fisiología , Animales , Médula Ósea , Linaje de la Célula , Ratones , Ratones Noqueados , Subunidad p52 de NF-kappa B/deficiencia , Factor de Transcripción PAX5/genética , Transactivadores/genética
19.
Med Sci (Paris) ; 24(12): 1083-8, 2008 Dec.
Artículo en Francés | MEDLINE | ID: mdl-19116119

RESUMEN

The family of NF-kappaB transcription factors plays a key role in diverse biological processes, such as inflammatory and immune responses, cell survival and tumor development. A new NF-kappaB activation pathway has been recently uncovered, the so-called alternative NF-kappaB activation pathway. It has been shown that this pathway mainly controls the activity of RelB, a member of the NF-kappaB family, and its activation requires IKKalpha, one of the two catalytic subunits of the IKK complex. Recent studies suggest that constitutive activation of the alternative pathway may be involved in tumor development, but most surprisingly it is also reported that IKKalpha exerts a protective function against uncontrolled cell proliferation and cancer development. This review discusses recent progresses in the understanding on how the alternative pathway may participate in cancer development, focusing more specifically on RelB and IKKalpha.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , FN-kappa B/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias/genética , Animales , Apoptosis , División Celular , Dimerización , Humanos , Quinasa I-kappa B/fisiología , Linfoma/genética , Linfoma/metabolismo , Ratones , Ratones Transgénicos , Modelos Genéticos , FN-kappa B/química , Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Receptores de Citocinas/fisiología , Factor de Transcripción ReIB/fisiología , Ubiquitinación , Vertebrados/genética , Vertebrados/metabolismo
20.
Biochem Soc Trans ; 35(Pt 2): 270-2, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17371257

RESUMEN

Inflammation is a beneficial response to insult or injury which plays an important role in orchestrating the adaptive immune response. The resolution of acute inflammation is an active process that involves the release of anti-inflammatory mediators and the termination of pro-inflammatory signalling pathways coincident with leucocyte apoptosis and phagocytic clearance and the migration of antigen-presenting cells from the site of inflammation to the local lymphatic tissue. The latter process is required for the development of adaptive immunity and immunological memory. The NF-kappaB (nuclear factor kappaB) pathway is an important regulator of inflammation and immunity; NF-kappaB activation is controlled by IKK [IkappaB (inhibitor of NF-kappaB) kinase] complex, which regulates NF-kappaB activation in response to pro-inflammatory stimuli. The IKK complex has two catalytic subunits, IKKalpha and IKKbeta; recent research shows that these highly homologous kinases have distinct roles in inflammation and adaptive immunity. Here, we discuss the emerging roles for IKKalpha in the tight regulation of inflammation and the development of adaptive immune responses.


Asunto(s)
Quinasa I-kappa B/fisiología , Inmunidad Innata , Inflamación/fisiopatología , Apoptosis , Humanos , Quinasa I-kappa B/genética , Inflamación/inmunología , Modelos Inmunológicos , FN-kappa B/fisiología , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA