Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Front Immunol ; 12: 662063, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995386

RESUMEN

Phagocytosis is an essential process for the uptake of large (>0.5 µm) particulate matter including microbes and dying cells. Specialized cells in the body perform phagocytosis which is enabled by cell surface receptors that recognize and bind target cells. Professional phagocytes play a prominent role in innate immunity and include macrophages, neutrophils and dendritic cells. These cells display a repertoire of phagocytic receptors that engage the target cells directly, or indirectly via opsonins, to mediate binding and internalization of the target into a phagosome. Phagosome maturation then proceeds to cause destruction and recycling of the phagosome contents. Key subsequent events include antigen presentation and cytokine production to alert and recruit cells involved in the adaptive immune response. Bridging the innate and adaptive immunity, macrophages secrete a broad selection of inflammatory mediators to orchestrate the type and magnitude of an inflammatory response. This review will focus on cytokines produced by NF-κB signaling which is activated by extracellular ligands and serves a master regulator of the inflammatory response to microbes. Macrophages secrete pro-inflammatory cytokines including TNFα, IL1ß, IL6, IL8 and IL12 which together increases vascular permeability and promotes recruitment of other immune cells. The major anti-inflammatory cytokines produced by macrophages include IL10 and TGFß which act to suppress inflammatory gene expression in macrophages and other immune cells. Typically, macrophage cytokines are synthesized, trafficked intracellularly and released in response to activation of pattern recognition receptors (PRRs) or inflammasomes. Direct evidence linking the event of phagocytosis to cytokine production in macrophages is lacking. This review will focus on cytokine output after engagement of macrophage phagocytic receptors by particulate microbial targets. Microbial receptors include the PRRs: Toll-like receptors (TLRs), scavenger receptors (SRs), C-type lectin and the opsonic receptors. Our current understanding of how macrophage receptor stimulation impacts cytokine production is largely based on work utilizing soluble ligands that are destined for endocytosis. We will instead focus this review on research examining receptor ligation during uptake of particulate microbes and how this complex internalization process may influence inflammatory cytokine production in macrophages.


Asunto(s)
Citocinas/inmunología , Macrófagos/inmunología , Fagocitos/inmunología , Fagocitos/microbiología , Transducción de Señal/inmunología , Animales , Antígenos Bacterianos/inmunología , Citocinas/biosíntesis , Humanos , Inmunidad Innata , Ratones , Subunidad p50 de NF-kappa B/inmunología , Fagocitosis/inmunología , Fagosomas/inmunología , Fagosomas/microbiología , Receptores Toll-Like/inmunología
2.
J Immunol ; 206(4): 904-916, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33441438

RESUMEN

Age-related chronic inflammation promotes cellular senescence, chronic disease, cancer, and reduced lifespan. In this study, we wanted to explore the effects of a moderate exercise regimen on inflammatory liver disease and tumorigenesis. We used an established model of spontaneous inflammaging, steatosis, and cancer (nfkb1-/- mouse) to demonstrate whether 3 mo of moderate aerobic exercise was sufficient to suppress liver disease and cancer development. Interventional exercise when applied at a relatively late disease stage was effective at reducing tissue inflammation (liver, lung, and stomach), oxidative damage, and cellular senescence, and it reversed hepatic steatosis and prevented tumor development. Underlying these benefits were transcriptional changes in enzymes driving the conversion of tryptophan to NAD+, this leading to increased hepatic NAD+ and elevated activity of the NAD+-dependent deacetylase sirtuin. Increased SIRT activity was correlated with enhanced deacetylation of key transcriptional regulators of inflammation and metabolism, NF-κB (p65), and PGC-1α. We propose that moderate exercise can effectively reprogram pre-established inflammatory and metabolic pathologies in aging with the benefit of prevention of disease.


Asunto(s)
Envejecimiento/inmunología , Carcinogénesis/inmunología , Hígado Graso/prevención & control , Neoplasias Hepáticas/prevención & control , Condicionamiento Físico Animal , Envejecimiento/genética , Envejecimiento/patología , Animales , Carcinogénesis/patología , Senescencia Celular/inmunología , Hígado Graso/inmunología , Hígado Graso/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/inmunología
3.
Rheumatology (Oxford) ; 60(7): 3420-3431, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33230538

RESUMEN

OBJECTIVE: RA encompasses a complex, heterogeneous and dynamic group of diseases arising from molecular and cellular perturbations of synovial tissues. The aim of this study was to decipher this complexity using an integrative systems approach and provide novel insights for designing stratified treatments. METHODS: An RNA sequencing dataset of synovial tissues from 152 RA patients and 28 normal controls was imported and subjected to filtration of differentially expressed genes, functional enrichment and network analysis, non-negative matrix factorization, and key driver analysis. A naïve Bayes classifier was applied to the independent datasets to investigate the factors associated with treatment outcome. RESULTS: A matrix of 1241 upregulated differentially expressed genes from RA samples was classified into three subtypes (C1-C3) with distinct molecular and cellular signatures. C3 with prominent immune cells and proinflammatory signatures had a stronger association with the presence of ACPA and showed a better therapeutic response than C1 and C2, which were enriched with neutrophil and fibroblast signatures, respectively. C2 was more occupied by synovial fibroblasts of destructive phenotype and carried highly expressed key effector molecules of invasion and osteoclastogenesis. CXCR2, JAK3, FYN and LYN were identified as key driver genes in C1 and C3. HDAC, JUN, NFKB1, TNF and TP53 were key regulators modulating fibroblast aggressiveness in C2. CONCLUSIONS: Deep phenotyping of synovial heterogeneity captured comprehensive and discrete pathophysiological attributes of RA regarding clinical features and treatment response. This result could serve as a template for future studies to design stratified approaches for RA patients.


Asunto(s)
Artritis Reumatoide/genética , Fibroblastos/metabolismo , Neutrófilos/metabolismo , Membrana Sinovial/metabolismo , Anticuerpos Antiproteína Citrulinada/inmunología , Artritis Reumatoide/inmunología , Teorema de Bayes , Bases de Datos Genéticas , Fibroblastos/inmunología , Perfilación de la Expresión Génica , Histona Desacetilasas/genética , Histona Desacetilasas/inmunología , Humanos , Janus Quinasa 3/genética , Janus Quinasa 3/inmunología , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/inmunología , Neutrófilos/inmunología , Osteogénesis/genética , Osteogénesis/inmunología , Fenotipo , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/inmunología , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/inmunología , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/inmunología , Membrana Sinovial/inmunología , Análisis de Sistemas , Transcriptoma , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/inmunología , Familia-src Quinasas/genética , Familia-src Quinasas/inmunología
4.
FEBS J ; 288(2): 640-662, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32386462

RESUMEN

Nuclear factor 'κ-light-chain-enhancer' of activated B cells (NF-κB) signaling is a signaling pathway used by most immune cells to promote immunostimulatory functions. Recent studies have indicated that regulatory T cells (Treg) differentially integrate TCR-derived signals, thereby maintaining their suppressive features. However, the role of NF-κB signaling in the activation of human peripheral blood (PB) Treg has not been fully elucidated so far. We show that the activity of the master transcription factor forkhead box protein 3 (FOXP3) attenuates p65 phosphorylation and nuclear translocation of the NF-κB proteins p50, p65, and c-Rel following activation in human Treg. Using pharmacological and genetic inhibition of canonical NF-κB signaling in FOXP3-transgenic T cells and PB Treg from healthy donors as well as Treg from a patient with a primary NFKB1 haploinsufficiency, we validate that Treg activation and suppressive capacity is independent of NF-κB signaling. Additionally, repression of residual NF-κB signaling in Treg further enhances interleukin-10 (IL-10) production. Blockade of NF-κB signaling can be exploited for the generation of in vitro induced Treg (iTreg) with enhanced suppressive capacity and functional stability. In this respect, dual blockade of mammalian target of rapamycin (mTOR) and NF-κB signaling was accompanied by enhanced expression of the transcription factors FOXP1 and FOXP3 and demethylation of the Treg-specific demethylated region compared to iTreg generated under mTOR blockade alone. Thus, we provide first insights into the role of NF-κB signaling in human Treg. These findings could lead to strategies for the selective manipulation of Treg and the generation of improved iTreg for cellular therapy.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Haploinsuficiencia/inmunología , Subunidad p50 de NF-kappa B/inmunología , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/inmunología , Factor de Transcripción ReIA/inmunología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/inmunología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Activación de Linfocitos , Subunidad p50 de NF-kappa B/deficiencia , Subunidad p50 de NF-kappa B/genética , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Transducción de Señal , Sirolimus/farmacología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Tiazoles/farmacología , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética
5.
Carbohydr Polym ; 251: 117088, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33142629

RESUMEN

Sea cucumbers were nutritional food and traditional Chinese medicine. In this study, fucosylated chondroitin sulfate from sea cucumber Stichopus chloronotus (fCS-Sc), a potential anticoagulant agent and immunological adjuvant, was investigated for its immune activation effects on RAW 264.7 macrophage for the first time. The results indicated that fCS-Sc could significantly promote the proliferation, the pinocytic activity of RAW 264.7 cells, and the production of NO, TNF-α, IL-1ß, and IL-6. The fluorescence labeling assay indicated that fCS-Sc could bind to the macrophage. Moreover, the specific pattern recognition receptor inhibition assays showed that toll-like receptor 4 (TLR4) and TLR2 were involved in the recognition of fCS-Sc. Western blot assays indicated that fCS-Sc could induce degradation of cytoplasm IκB-α, and promotion of NF-κB p65 subunit translocation to nucleus, leading to a functional improvement of macrophage through NF-κB pathway. The results suggested that fCS-Sc might served as a promising candidate of immunomodulator.


Asunto(s)
Sulfatos de Condroitina/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Stichopus/química , Animales , Proliferación Celular/efectos de los fármacos , Sulfatos de Condroitina/aislamiento & purificación , Citocinas/inmunología , Inmunomodulación , Ratones , Subunidad p50 de NF-kappa B/inmunología , Pinocitosis/efectos de los fármacos , Células RAW 264.7
6.
Artículo en Inglés | MEDLINE | ID: mdl-32058033

RESUMEN

Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) lower risk of cardiovascular disease. The primary source of EPA and DHA is fatty fish. Plant-derived alpha linolenic acid (ALA) and stearidonic acid (SDA) could provide sustainable land-based alternatives, but their functionality is underexplored. Omega-3 fatty acids (n-3 FAs) may influence atherogenic processes through changing endothelial cell (EC) function and lowering inflammation. This study compared effects of marine- and plant-derived n-3 FAs on EC inflammatory responses. EA.hy926 cells were exposed to ALA, SDA, EPA or DHA prior to stimulation with tumor necrosis factor (TNF)-α. All FAs were shown to be incorporated into ECs in a dose-dependent manner. SDA (50 µM) decreased both production and cell-surface expression of intercellular adhesion molecule (ICAM)-1; however EPA and DHA resulted in greater reduction of ICAM-1 production and expression. EPA and DHA also significantly lowered production of monocyte chemoattractant protein 1, interleukin (IL)-6 and IL-8. ALA, SDA and DHA (50 µM) all reduced adhesion of THP-1 monocytes to EA.hy926 cells. DHA significantly decreased nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB)p105 gene expression and phosphorylated NFκBp65 protein. Both EPA and DHA (50 µM) significantly decreased cyclooxygenase (COX)-2 protein. Thus, both marine-derived n-3 FAs, particularly DHA, had potent anti-inflammatory effects in this EC model. Of the plant-derived n-3 FAs, SDA showed the greatest inhibition of inflammation. Although neither ALA nor SDA reproduced the anti-inflammatory effects of EPA and DHA in this model, there is some potential for SDA to be a sustainable anti-inflammatory alternative to the marine n-3 FAs.


Asunto(s)
Antiinflamatorios/farmacología , Ácidos Grasos Omega-3/farmacología , Aceites de Pescado/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Fitoquímicos/farmacología , Línea Celular , Quimiocina CCL2/inmunología , Quimiocina CCL2/metabolismo , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Interleucina-8/inmunología , Interleucina-8/metabolismo , Subunidad p50 de NF-kappa B/inmunología , Subunidad p50 de NF-kappa B/metabolismo , Factor de Transcripción ReIA/inmunología , Factor de Transcripción ReIA/metabolismo
7.
J Immunother Cancer ; 8(1)2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31940589

RESUMEN

BACKGROUND: Macrophages and dendritic cells lacking the transcription factor nuclear factor kappa B p50 are skewed toward a proinflammatory phenotype, with increased cytokine expression and enhanced T cell activation; additionally, murine melanoma, fibrosarcoma, colon carcinoma, and glioblastoma grow slower in p50-/- mice. We therefore evaluated the efficacy of p50-negative immature myeloid cells (p50-IMCs) adoptively transferred into tumor-bearing hosts. Immature cells were used to maximize tumor localization, and pretreatment with 5-fluorouracil (5FU) was examined due to its potential to impair marrow production of myeloid cells, to target tumor myeloid cells and to release tumor neoantigens. METHODS: Wild-type (WT)-IMC or p50-IMC were generated by culturing lineage-negative marrow cells from WT or p50-/- mice in media containing thrombopoietin, stem cell factor and Flt3 ligand for 6 days followed by monocyte colony-stimulating factor for 1 day on ultralow attachment plates. Mice inoculated with Hi-Myc prostate cancer (PCa) cells or K-RasG12D pancreatic ductal carcinoma (PDC)-luciferase cells received 5FU followed 5 days later by three doses of 107 immature myeloid cells (IMC) every 3-4 days. RESULTS: PCa cells grew slower in p50-/- mice, and absence of host p50 prolonged the survival of mice inoculated orthotopically with PDC cells. IMC from Cytomegalovirus (CMV)-luciferase mice localized to tumor, nodes, spleen, marrow, and lung. 5FU followed by p50-IMC slowed PCa and PDC tumor growth, ~3-fold on average, in contrast to 5FU followed by WT-IMC, 5FU alone or p50-IMC alone. Slowed tumor growth was evident for 93% of PCa but only 53% of PDC tumors; we therefore focused on PCa for additional IMC analyses. In PCa, p50-IMC matured into F4/80+ macrophages, as well as CD11b+F4/80-CD11c+ conventional dendritic cells (cDCs). In both tumor and draining lymph nodes, p50-IMC generated more macrophages and cDCs than WT-IMC. Activated tumor CD8+ T cells were increased fivefold by p50-IMC compared with WT-IMC, and antibody-mediated CD8+ T cell depletion obviated slower tumor growth induced by 5FU followed by p50-IMC. CONCLUSIONS: 5FU followed by p50-IMC slows the growth of murine prostate and pancreatic carcinoma and depends on CD8+ T cell activation. Deletion of p50 in patient-derived marrow CD34+ cells and subsequent production of IMC for adoptive transfer may contribute to the therapy of these and additional cancers.


Asunto(s)
Carcinoma Ductal Pancreático/terapia , Inmunoterapia Adoptiva/métodos , Células Mieloides/inmunología , Células Mieloides/trasplante , Subunidad p50 de NF-kappa B/deficiencia , Neoplasias Pancreáticas/terapia , Neoplasias de la Próstata/terapia , Animales , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Femenino , Fluorouracilo/farmacología , Masculino , Ratones , Subunidad p50 de NF-kappa B/inmunología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología
8.
Front Immunol ; 10: 2618, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31803180

RESUMEN

Adult-onset primary immunodeficiency is characterized by recurrent infections, hypogammaglobulinemia, and poor antibody response to vaccines. In this study, we have analyzed targeted gene panel sequencing results of 270 patients diagnosed with antibody deficiency and identified five disease-associated variants in NFKB1 in five unrelated families. We detected two single base pair deletions and two single base pair insertions, causing severe protein truncations, and one missense mutation. Immunoblotting, lymphocyte stimulation, immunophenotyping, and ectopic expression assays demonstrated the functional relevance of NFKB1 mutations. Besides antibody deficiency, clinical manifestations included infections, autoimmune features, lymphoproliferation, lymphoma, Addison's disease, type 2 diabetes and asthma. Although partial clinical penetrance was observed in almost all pedigrees, all carriers presented a deficiency in certain serum immunoglobulins and the majority showed a lack of memory B cells (CD19+CD27+). Among all tested genes, NFKB1 alterations were the most common monoallelic cause of antibody deficiency in our cohort.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Subunidad p50 de NF-kappa B/genética , Enfermedades de Inmunodeficiencia Primaria/genética , Adolescente , Adulto , Edad de Inicio , Alelos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Subunidad p50 de NF-kappa B/inmunología , Linaje , Enfermedades de Inmunodeficiencia Primaria/inmunología , Adulto Joven
9.
Front Immunol ; 10: 1369, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31281314

RESUMEN

Myeloid-derived suppressor cells (MDSC) are well-known key negative regulators of the immune response during tumor growth, however scattered is the knowledge of their capacity to influence and adapt to the different tumor microenvironments and of the markers that identify those capacities. Here we show that the secreted protein acidic and rich in cysteine (SPARC) identifies in both human and mouse MDSC with immune suppressive capacity and pro-tumoral activities including the induction of epithelial-to-mesenchymal transition (EMT) and angiogenesis. In mice the genetic deletion of SPARC reduced MDSC immune suppression and reverted EMT. Sparc-/- MDSC were less suppressive overall and the granulocytic fraction was more prone to extrude neutrophil extracellular traps (NET). Surprisingly, arginase-I and NOS2, whose expression can be controlled by STAT3, were not down-regulated in Sparc-/- MDSC, although less suppressive than wild type (WT) counterpart. Flow cytometry analysis showed equal phosphorylation of STAT3 but reduced ROS production that was associated with reduced nuclear translocation of the NF-kB p50 subunit in Sparc-/- than WT MDSC. The limited p50 in nuclei reduce the formation of the immunosuppressive p50:p50 homodimers in favor of the p65:p50 inflammatory heterodimers. Supporting this hypothesis, the production of TNF by Sparc-/- MDSC was significantly higher than by WT MDSC. Although associated with tumor-induced chronic inflammation, TNF, if produced at high doses, becomes a key factor in mediating tumor rejection. Therefore, it is foreseeable that an unbalance in TNF production could skew MDSC toward an inflammatory, anti-tumor phenotype. Notably, TNF is also required for inflammation-driven NETosis. The high level of TNF in Sparc-/- MDSC might explain their increased spontaneous NET formation as that we detected both in vitro and in vivo, in association with signs of endothelial damage. We propose SPARC as a new potential marker of MDSC, in both human and mouse, with the additional feature of controlling MDSC suppressive activity while preventing an excessive inflammatory state through the control of NF-kB signaling pathway.


Asunto(s)
Transición Epitelial-Mesenquimal/inmunología , Células Supresoras de Origen Mieloide/inmunología , Osteonectina/inmunología , Animales , Arginasa/genética , Arginasa/inmunología , Biomarcadores , Transición Epitelial-Mesenquimal/genética , Trampas Extracelulares/genética , Trampas Extracelulares/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células Supresoras de Origen Mieloide/citología , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Osteonectina/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología
10.
Eur J Immunol ; 49(11): 2051-2062, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31269241

RESUMEN

DCs and epithelial cell-derived thymic stromal lymphopoietin (TSLP) have pivotal roles in allergic inflammation. TSLP stimulates myeloid DCs to express OX40-ligand (OX40L) and CCL17, which trigger and maintain Th2 cell responses. We have previously shown that statins, which are HMG-CoA reductase inhibitors, have the ability to suppress type I IFN production by plasmacytoid DCs. Here, we extended our previous work to examine the immunomodulatory effect of statins on allergic responses, particularly the TSLP-dependent Th2 pathway induced by myeloid DCs. We found that treatment of TSLP-stimulated DCs with either pitavastatin or simvastatin suppressed both the DC-mediated inflammatory Th2 cell differentiation and CRTH2+ CD4+ memory Th2 cell expansion and also repressed the expressions of OX40L and CCL17 by DCs. These inhibitory effects of statins were mimicked by treatment with either a geranylgeranyl-transferase inhibitor or Rho-kinase inhibitor and were counteracted by the addition of mevalonate, suggesting that statins induce geranylgeranylated Rho inactivation through a mevalonate-dependent pathway. We also found that statins inhibited the expressions of phosphorylated STA6 and NF-κB-p50 in TSLP-stimulated DCs. This study identified a specific ability of statins to control DC-mediated Th2 responses, suggesting their therapeutic potential for treating allergic diseases.


Asunto(s)
Quimiocina CCL17/inmunología , Células Dendríticas/efectos de los fármacos , Ligando OX40/inmunología , Quinolinas/farmacología , Simvastatina/farmacología , Células Th2/efectos de los fármacos , Anticuerpos Neutralizantes/farmacología , Antígenos CD28/genética , Antígenos CD28/inmunología , Complejo CD3/genética , Complejo CD3/inmunología , Proliferación Celular/efectos de los fármacos , Quimiocina CCL17/antagonistas & inhibidores , Quimiocina CCL17/genética , Técnicas de Cocultivo , Citocinas/farmacología , Células Dendríticas/citología , Células Dendríticas/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Memoria Inmunológica/efectos de los fármacos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucinas/genética , Interleucinas/inmunología , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/inmunología , Ligando OX40/antagonistas & inhibidores , Ligando OX40/genética , Cultivo Primario de Células , Transducción de Señal , Células Th2/citología , Células Th2/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Linfopoyetina del Estroma Tímico
11.
Adv Cancer Res ; 142: 145-186, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30885361

RESUMEN

Somatic mutations in cancer cells may influence tumor growth, survival, or immune interactions in their microenvironment. The tumor necrosis factor receptor family member HVEM (TNFRSF14) is frequently mutated in cancers and has been attributed a tumor suppressive role in some cancer contexts. HVEM functions both as a ligand for the lymphocyte checkpoint proteins BTLA and CD160, and as a receptor that activates NF-κB signaling pathways in response to BTLA and CD160 and the TNF ligands LIGHT and LTα. BTLA functions to inhibit lymphocyte activation, but has also been ascribed a role in stimulating cell survival. CD160 functions to co-stimulate lymphocyte function, but has also been shown to activate inhibitory signaling in CD4+ T cells. Thus, the role of HVEM within diverse cancers and in regulating the immune responses to these tumors is likely context specific. Additionally, development of therapeutics that target proteins within this network of interacting proteins will require a deeper understanding of how these proteins function in a cancer-specific manner. However, the prominent role of the HVEM network in anti-cancer immune responses indicates a promising area for drug development.


Asunto(s)
Neoplasias/genética , Neoplasias/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Linfocitos T/inmunología , Genes Supresores de Tumor , Humanos , Activación de Linfocitos , Mutación , Subunidad p50 de NF-kappa B/inmunología , Subunidad p50 de NF-kappa B/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Microambiente Tumoral
12.
Front Immunol ; 9: 2210, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319651

RESUMEN

Sepsis is a major cause of neonatal morbidity and mortality. The current paradigm suggests that neonatal susceptibility to infection is explained by an innate immune response that is functionally immature. Recent studies in adults have questioned a therapeutic role for IFNß in sepsis; however, the role of IFNß in mediating neonatal sensitivity to sepsis is unknown. We evaluated the transcriptional regulation and expression of IFNß in early neonatal (P0) and adult murine models of endotoxemia (IP LPS, 5 mg/kg). We found that hepatic, pulmonary, and serum IFNß expression was significantly attenuated in endotoxemic neonates when compared to similarly exposed adults. Furthermore, endotoxemia induced hepatic p65/NFκB and IRF3 activation exclusively in adults. In contrast, endotoxemia induced immunotolerant p50/NFκB signaling in neonatal mice without evidence of IRF3 activation. Consistent with impaired IFNß expression and attenuated circulating serum levels, neonatal pulmonary STAT1 signaling and target gene expression was significantly lower than adult levels. Using multiple in vivo approaches, the source of hepatic IFNß expression in endotoxemic adult mice was determined to be the hepatic macrophage, and experiments in RAW 264.7 cells confirmed that LPS-induced IFNß expression was NFκB dependent. Finally, treating neonatal mice with IFNß 2 h after endotoxemia stimulated pulmonary STAT1 signaling and STAT1 dependent gene expression. Furthermore, IFNß treatment of endotoxemic neonatal animals resulted in significantly improved survival following exposure to lethal endotoxemia. In conclusion, endotoxemia induced IFNß expression is attenuated in the early neonatal period, secondary to impaired NFκB-p65/IRF3 signaling. Pre-treatment with IFNß decreases neonatal sensitivity to endotoxemia. These results support further study of the role of impaired IFNß expression and neonatal sensitivity to sepsis.


Asunto(s)
Endotoxemia/inmunología , Tolerancia Inmunológica , Interferón beta/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Transducción de Señal/inmunología , Factores de Edad , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Humanos , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/inmunología , Lipopolisacáridos/toxicidad , Hígado/inmunología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Subunidad p50 de NF-kappa B/inmunología , Células RAW 264.7 , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción ReIA/inmunología , Factor de Transcripción ReIA/metabolismo
13.
J Allergy Clin Immunol ; 142(4): 1041-1051, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30170128

RESUMEN

This manuscript reviews selected topics in primary immunodeficiency diseases (PIDDs) published in 2017. These include (1) the role of follicular T cells in the differentiation of B cells and development of optimal antibody responses; (2) impaired nuclear factor κB subunit 1 signaling in the pathogenesis of common variable immunodeficiency, revealing an association between impaired B-cell maturation and development of inflammatory conditions; (3) autoimmune and inflammatory manifestations in patients with PIDDs in T- and B-cell deficiencies, as well as in neutrophil disorders; (4) newly described gene defects causing PIDDs, including exostosin-like 3 (EXTL3), TNF-α-induced protein 3 (TNFAIP3 [A20]), actin-related protein 2/3 complex-subunit 1B (ARPC1B), v-Rel avian reticuloendotheliosis viral oncogene homolog A (RELA), hypoxia upregulated 1 (HYOU1), BTB domain and CNC homolog 2 (BACH2), CD70, and CD55; (5) use of rapamycin and the phosphoinositide 3-kinase inhibitor leniolisib to reduce autoimmunity and regulate B-cell function in the activated phosphoinositide 3-kinase δ syndrome; (6) improved outcomes in hematopoietic stem cell transplantation for severe combined immunodeficiency (SCID) in the last decade, with an overall 2-year survival of 90% in part caused by early diagnosis through implementation of universal newborn screening; (7) demonstration of the efficacy of lentiviral vector-mediated gene therapy for patients with adenosine deaminase-deficient SCID; (8) the promise of gene editing for PIDDs using CRISPR/Cas9 and zinc finger nuclease technology for SCID and chronic granulomatous disease; and (9) the efficacy of thymus transplantation in Europe, although associated with an unexpected high incidence of autoimmunity. The remarkable progress in the understanding and management of PIDDs reflects the current interest in this area and continues to improve the care of immunodeficient patients.


Asunto(s)
Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/terapia , Animales , Autoinmunidad , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Subunidad p50 de NF-kappa B/inmunología , Esplenectomía , Linfocitos T Colaboradores-Inductores/inmunología , Timectomía
14.
Cytokine ; 111: 84-87, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30125779

RESUMEN

Deposition of amyloid-ß in Alzheimer's disease is accompanied by chronic inflammation, which involves raised levels of pro-inflammatory cytokines TNF-α, IL-6 and IL-1ß. However, the role of Aß1-42 in the inflammatory process, before it gets deposited into aggregates has not been investigated thoroughly. Through this study, we are illustrating the dual role of soluble Aß1-42 (sAß1-42) in activating the NLRP3 inflammasome and simultaneously inhibiting TNF-α secretion. Our data suggested that the treatment of chronically induced THP-1 macrophages and N9 microglial cells with sAß1-42 can suppress the major inflammatory cytokine TNF-α without affecting the level of IL-6. However, the activation of NLRP3 inflammasome was well evidenced by secretion of IL-1ß, increased expression of NLRP3 and caspase-1, implicating sAß1-42 in enhancing and suppressing one or other type of inflammation. Further investigation revealed that sAß1-42 was able to severely abrogate the expression of NF-κB, p50 and restricting the translocation of NF-κB, p65 to nucleus by inhibiting phosphorylation of IκB-α in THP-1 macrophages. These data indicate that the sAß1-42 may play a dual role during inflammatory process, wherein, it may be involved in protecting the cells from inflammatory damage due to TNF-α. This ability of sAß1-42 might be playing some role in protecting the brain cells during the process of aging and Alzheimer's disease, where, chronic inflammatory environment plays a vital role.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Inflamasomas/inmunología , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Fragmentos de Péptidos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Enfermedad de Alzheimer/patología , Humanos , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Macrófagos/patología , Microglía/inmunología , Microglía/patología , Subunidad p50 de NF-kappa B/inmunología , Transducción de Señal/inmunología , Solubilidad , Células THP-1 , Factor de Transcripción ReIA/inmunología
15.
Cell Death Dis ; 8(10): e3115, 2017 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-29022897

RESUMEN

Amyloid-beta (Aß) is a hallmark component of age-related macular degeneration (AMD), which induces secretion of pro-inflammatory cytokines from retinal pigment epithelium (RPE). Previous studies have shown that p50/RelA (p65), a member of NF-κB family, is an essential pro-inflammatory transcription factor responding to Aß1-40 stimulation, but few focused on the other two Rel transcription factor members - RelB and c-Rel - and their role in Aß1-40-mediated inflammation. It was reported that RelA, RelB and c-Rel are also implicated in various NF-κB-mediated inflammatory diseases. Therefore, we infer that Aß1-40-mediated inflammation targets not only the classical inflammation regulator, RelA, but also RelB and c-Rel. In this study, we demonstrate that intravitreally injected Aß1-40 mice develop AMD-like pathologic changes, coupled with Rel protein (RelA, RelB and c-Rel) synthesis and nuclear translocation. To focus on the interaction mechanism of Rel proteins, we found that RelB and c-Rel formed a heterodimer with RelA in mice model. We also found that c-Rel silencing decreased the levels of Aß1-40-dependent RelA expression, indicating that RelB and c-Rel may interact with RelA as coactivator and c-Rel is required to activate the expression of RelA. Moreover, Rel protein silencing decreased the expression of distinct pro-inflammatory cytokines. Together, we demonstrate that besides RelA, RelB and c-Rel can also be activated by Aß1-40, all of which mediate pro-inflammatory cytokine transcription and RPE damage. Our findings imply that RPE-mediated inflammation under the stimulation of Aß1-40 is multi-targeted and RelA, RelB and c-Rel proteins may be the new targets of anti-inflammatory agents.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Degeneración Macular/patología , Fragmentos de Péptidos/inmunología , Proteínas Proto-Oncogénicas c-rel/inmunología , Epitelio Pigmentado de la Retina/metabolismo , Factor de Transcripción ReIA/inmunología , Factor de Transcripción ReIB/inmunología , Péptidos beta-Amiloides/administración & dosificación , Animales , Células Cultivadas , Electrorretinografía , Regulación de la Expresión Génica , Inflamación/inmunología , Degeneración Macular/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/inmunología , Fragmentos de Péptidos/administración & dosificación , Proteínas Proto-Oncogénicas c-rel/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Epitelio Pigmentado de la Retina/citología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIB/genética
16.
Oncogene ; 36(29): 4224-4232, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28368397

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) has been categorized into two molecular subtypes that have prognostic significance, namely germinal center B-cell like (GCB) and activated B-cell like (ABC). Although ABC-DLBCL has been associated with NF-κB activation, the relationships between activation of specific NF-κB signals and DLBCL phenotype remain unclear. Application of novel gene expression classifiers identified two new DLBCL categories characterized by selective p100 (NF-κB2) and p105 (NF-κB1) signaling. Interestingly, our molecular studies showed that p105 signaling is predominantly associated with GCB subtype and histone mutations. Conversely, most tumors with p100 signaling displayed ABC phenotype and harbored ABC-associated mutations in genes such as MYD88 and PIM1. In vitro, MYD88 L265P mutation promoted p100 signaling through TAK1/IKKα and GSK3/Fbxw7a pathways, suggesting a novel role for this protein as an upstream regulator of p100. p100 signaling was engaged during activation of normal B cells, suggesting p100's role in ABC phenotype development. Additionally, silencing p100 in ABC-DLBCL cells resulted in a GCB-like phenotype, with suppression of Blimp, IRF4 and XBP1 and upregulation of BCL6, whereas introduction of p52 or p100 into GC cells resulted in differentiation toward an ABC-like phenotype. Together, these findings identify specific roles for p100 and p105 signaling in defining DLBCL molecular subtypes and posit MYD88/p100 signaling as a regulator for B-cell activation.


Asunto(s)
Linfoma de Células B Grandes Difuso/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Linfocitos B/inmunología , Humanos , Activación de Linfocitos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/inmunología , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/inmunología , Fenotipo , Transducción de Señal
17.
Clin Immunol ; 175: 99-108, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27923702

RESUMEN

NFKB1, a component of the canonical NF-κB pathway, was recently reported to be mutated in a limited number of CVID patients. CVID-associated mutations in NFKB2 (non-canonical pathway) have previously been shown to impair NK cell cytotoxic activity. Although a biological function of NFKB1 in non-human NK cells has been reported, the role of NFKB1 mutations for human NK cell biology and disease has not been investigated yet. We decided therefore to evaluate the role of monoallelic NFKB1 mutations in human NK cell maturation and functions. We show that NFKB1 mutated NK cells present impaired maturation, defective cytotoxicity and reduced IFN-γ production upon in vitro stimulation. Furthermore, human IL-2 activated NFKB1 mutated NK cells fail to up-regulate the expression of the activating marker NKp44 and show reduced proliferative capacity. These data suggest that NFKB1 plays an essential novel role for human NK cell maturation and effector functions.


Asunto(s)
Células Asesinas Naturales/inmunología , Subunidad p50 de NF-kappa B/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Femenino , Humanos , Interferón gamma/inmunología , Interleucina-2/inmunología , Células K562 , Masculino , FN-kappa B/inmunología
18.
Bull Exp Biol Med ; 162(2): 244-247, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27905034

RESUMEN

We studied the effects of Human TruStain FcX buffer (Fcγ receptor blocking solution) in experiments on evaluation of TLR4 level with labeled monoclonal antibodies, intracellular immunofluorescent staining of NF-κB p50, and TNF-α synthesis on human isolated monocytes and whole blood cells. The influence of the blocking buffer on the measured parameters should be taken into account and appropriateness of its use in experiments on isolated cells and whole blood should be considered.


Asunto(s)
Artefactos , Monocitos/metabolismo , Subunidad p50 de NF-kappa B/análisis , Receptores Fc/antagonistas & inhibidores , Receptor Toll-Like 4/análisis , Factor de Necrosis Tumoral alfa/análisis , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Células Sanguíneas/inmunología , Células Sanguíneas/metabolismo , Tampones (Química) , Ensayo de Inmunoadsorción Enzimática/normas , Citometría de Flujo/normas , Expresión Génica , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Indicadores y Reactivos/química , Monocitos/inmunología , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/inmunología , Cultivo Primario de Células , Receptores Fc/genética , Receptores Fc/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
20.
J Exp Med ; 213(4): 621-41, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-27022143

RESUMEN

We examined the role of NFκB1 in the homeostasis and function of peripheral follicular (Fo) B cells. Aging mice lacking NFκB1 (Nfκb1(-/-)) develop lymphoproliferative and multiorgan autoimmune disease attributed in large part to the deregulated activity of Nfκb1(-/-)Fo B cells that produce excessive levels of the proinflammatory cytokine interleukin 6 (IL-6). Despite enhanced germinal center (GC) B cell differentiation, the formation of GC structures was severely disrupted in the Nfκb1(-/-)mice. Bone marrow chimeric mice revealed that the Fo B cell-intrinsic loss of NFκB1 led to the spontaneous generation of GC B cells. This was primarily the result of an increase in IL-6 levels, which promotes the differentiation of Fo helper CD4(+)T cells and acts in an autocrine manner to reduce antigen receptor and toll-like receptor activation thresholds in a population of proliferating IgM(+)Nfκb1(-/-)Fo B cells. We demonstrate that p50-NFκB1 represses Il-6 transcription in Fo B cells, with the loss of NFκB1 also resulting in the uncontrolled RELA-driven transcription of Il-6.Collectively, our findings identify a previously unrecognized role for NFκB1 in preventing multiorgan autoimmunity through its negative regulation of Il-6 gene expression in Fo B cells.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Centro Germinal/inmunología , Interleucina-6/inmunología , Subunidad p50 de NF-kappa B/inmunología , Transcripción Genética/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Linfocitos B/patología , Centro Germinal/patología , Inmunoglobulina M/genética , Inmunoglobulina M/inmunología , Interleucina-6/genética , Ratones , Ratones Noqueados , Subunidad p50 de NF-kappa B/genética , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA