Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(2): e0261854, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35108274

RESUMO

The nuclear factor-kappa B (NF-κB) is a transcription factor with important roles in inflammation, immune response, and oncogenesis. Dysregulation of NF-κB signaling is associated with inflammation and certain cancers. We developed a gene expression biomarker predictive of NF-κB modulation and used the biomarker to screen a large compendia of gene expression data. The biomarker consists of 108 genes responsive to tumor necrosis factor α in the absence but not the presence of IκB, an inhibitor of NF-κB. Using a set of 450 profiles from cells treated with immunomodulatory factors with known NF-κB activity, the balanced accuracy for prediction of NF-κB activation was > 90%. The biomarker was used to screen a microarray compendium consisting of 12,061 microarray comparisons from human cells exposed to 2,672 individual chemicals to identify chemicals that could cause toxic effects through NF-κB. There were 215 and 49 chemicals that were identified as putative or known NF-κB activators or suppressors, respectively. NF-κB activators were also identified using two high-throughput screening assays; 165 out of the ~3,800 chemicals (ToxCast assay) and 55 out of ~7,500 unique compounds (Tox21 assay) were identified as potential activators. A set of 32 chemicals not previously associated with NF-κB activation and which partially overlapped between the different screens were selected for validation in wild-type and NFKB1-null HeLa cells. Using RT-qPCR and targeted RNA-Seq, 31 of the 32 chemicals were confirmed to be NF-κB activators. These results comprehensively identify a set of chemicals that could cause toxic effects through NF-κB.


Assuntos
Biomarcadores/metabolismo , Regulação da Expressão Gênica/genética , NF-kappa B/metabolismo , Linhagem Celular , Bases de Dados de Compostos Químicos , Regulação da Expressão Gênica/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , NF-kappa B/agonistas , NF-kappa B/antagonistas & inibidores , Subunidade p50 de NF-kappa B/deficiência , Subunidade p50 de NF-kappa B/genética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
2.
Front Immunol ; 12: 669906, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34721373

RESUMO

Since its discovery over 30 years ago the NF-ĸB family of transcription factors has gained the status of master regulator of the immune response. Much of what we understand of the role of NF-ĸB in immune development, homeostasis and inflammation comes from studies of mice null for specific NF-ĸB subunit encoding genes. The role of inflammation in diseases that affect a majority of individuals with health problems globally further establishes NF-ĸB as an important pathogenic factor. More recently, genomic sequencing has revealed loss of function mutations in the NFKB1 gene as the most common monogenic cause of common variable immunodeficiencies in Europeans. NFKB1 encodes the p105 subunit of NF-ĸB which is processed to generate the NF-ĸB p50 subunit. NFKB1 is the most highly expressed transcription factor in macrophages, key cellular drivers of inflammation and immunity. Although a key role for NFKB1 in the control of the immune system is apparent from Nfkb1-/- mouse studies, we know relatively little of the role of NFKB1 in regulating human macrophage responses. In this study we use the THP1 monocyte cell line and CRISPR/Cas9 gene editing to generate a model of NFKB1-/- human macrophages. Transcriptomic analysis reveals that activated NFKB1-/- macrophages are more pro-inflammatory than wild type controls and express elevated levels of TNF, IL6, and IL1B, but also have reduced expression of co-stimulatory factors important for the activation of T cells and adaptive immune responses such as CD70, CD83 and CD209. NFKB1-/- THP1 macrophages recapitulate key observations in individuals with NFKB1 haploinsufficiency including decreased IL10 expression. These data supporting their utility as an in vitro model for understanding the role of NFKB1 in human monocytes and macrophages and indicate that of loss of function NFKB1 mutations in these cells is an important component in the associated pathology.


Assuntos
Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Inflamação/genética , Macrófagos/metabolismo , Subunidade p50 de NF-kappa B/genética , Transcriptoma , Imunidade Adaptativa , Sistemas CRISPR-Cas , Citocinas/genética , Citocinas/metabolismo , Humanos , Imunidade Celular , Inflamação/imunologia , Inflamação/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Subunidade p50 de NF-kappa B/deficiência , Fenótipo , RNA-Seq , Células THP-1 , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
3.
FEBS J ; 288(2): 640-662, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32386462

RESUMO

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.


Assuntos
Fatores de Transcrição Forkhead/imunologia , Haploinsuficiência/imunologia , Subunidade p50 de NF-kappa B/imunologia , Linfócitos T Reguladores/imunologia , Serina-Treonina Quinases TOR/imunologia , Fator de Transcrição RelA/imunologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/imunologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/imunologia , Núcleo Celular/metabolismo , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Humanos , Interleucina-10/genética , Interleucina-10/imunologia , Ativação Linfocitária , Subunidade p50 de NF-kappa B/deficiência , Subunidade p50 de NF-kappa B/genética , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Transdução de Sinais , Sirolimo/farmacologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Tiazóis/farmacologia , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/genética
5.
J Immunother Cancer ; 8(1)2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31940589

RESUMO

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.


Assuntos
Carcinoma Ductal Pancreático/terapia , Imunoterapia Adotiva/métodos , Células Mieloides/imunologia , Células Mieloides/transplante , Subunidade p50 de NF-kappa B/deficiência , Neoplasias Pancreáticas/terapia , Neoplasias da Próstata/terapia , Animais , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/patologia , Feminino , Fluoruracila/farmacologia , Masculino , Camundongos , Subunidade p50 de NF-kappa B/imunologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia
6.
Sci Rep ; 9(1): 15912, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31685905

RESUMO

Mice emit ultrasonic vocalizations (USVs) in different social conditions: pups maternal separation, juveniles play, adults mating and social investigation. The USVs measurement has become an important instrument for behavioural phenotyping in neurodevelopmental disorders (NDDs). Recently, we have demonstrated that the deletion of the NFκB1 gene, which encodes the p50 NF-κB subunit, causes NDDs phenotype in mice. In this study, we investigated the ultrasonic communication and the effects of an early social enrichment in mice lacking the NF-κB p50 subunit (p50 KO). In particular, USVs of wild-type (WT), p50 KO and KO exposed to early social enrichment (KO enriched) were recorded using an ultrasound sensitive microphone and analysed by Avisoft software. USVs analysis showed that p50 KO pups emit more and longer vocalizations compared to WT pups. On the contrary, in adulthood, p50 KO mice emit less USVs than WT mice. We also found significant qualitative differences in p50 KO mice USVs compared to WT mice; the changes specifically involved two USVs categories. Early social enrichment had no effect on USVs number, duration and type in p50 KO mice. Together, these data revealed social communication alterations in a mouse model of NDDs; these deficits were not recovered by early social enrichment, strengthening the fact that genetic background prevails on environmental enrichment.


Assuntos
Transtornos do Neurodesenvolvimento/patologia , Vocalização Animal , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B/deficiência , Subunidade p50 de NF-kappa B/genética , Transtornos do Neurodesenvolvimento/metabolismo , Fenótipo
7.
J Neuroinflammation ; 16(1): 60, 2019 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-30871598

RESUMO

BACKGROUND: While NF-κB p50 function is impaired in central nervous system disease, aging in non-CNS tissues, and response to reactive oxygen species, the role of NF-κB p50 in aging-associated microglial pro-inflammatory priming is poorly understood. METHODS: Male NF-κB p50+/+ and NF-κB p50-/- mice at three different ages (1.5-3.0 month old, 8.0-11.0 month old, and 16.0-18.0 month old) were treated with LPS (5 mg/kg, IP) to trigger peripheral inflammation, where circulating cytokines, neuroinflammation, microglia morphology, and NF-κB p50/p65 function in brain tissue were determined 3 h later. RESULTS: Peripheral LPS injection in 9-month-old C57BL/6 mice resulted in lower NF-κB p50 DNA binding of nuclear extracts from the whole brain, when compared to 3-week-old C57BL/6 mice, revealing differences in LPS-induced NF-κB p50 activity in the brain across the mouse lifespan. To examine the consequences of loss NF-κB p50 function with aging, NF-κB p50+/+ and NF-κB p50-/- mice of three different age groups (1.5-3.0 month old, 8.0-11.0 month old, and 16.0-18.0 month old) were injected with LPS (5 mg/kg, IP). NF-κB p50-/- mice showed markedly elevated circulating, midbrain, and microglial TNFα when compared to NF-κB p50+/+ mice at all ages. Notably, the 16.0-18.0-month-old (middle aged) NF-κB p50-/- mice exhibited synergistically augmented LPS-induced serum and midbrain TNFα when compared to the younger (1.5-3.0 month old, young adult) NF-κB p50-/- mice. The 16.0-18.0-month-old LPS-treated NF-κB p50-/- mice also had the highest midbrain IL-1ß expression, largest number of microglia with changes in morphology, and greatest elevation of pro-inflammatory factors in isolated adult microglia. Interestingly, aging NF-κB p50-/- mice exhibited decreased brain NF-κB p65 expression and activity. CONCLUSIONS: These findings support that loss of NF-κB p50 function and aging in middle-aged mice may interact to excessively augment peripheral/microglial pro-inflammatory responses and point to a novel neuroinflammation signaling mechanism independent the NF-κB p50/p65 transcription factor in this process.


Assuntos
Envelhecimento/patologia , Encéfalo/metabolismo , Inflamação/patologia , Microglia/patologia , Subunidade p50 de NF-kappa B/deficiência , Fatores Etários , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Inflamação/induzido quimicamente , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Subunidade p50 de NF-kappa B/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Physiol Rep ; 6(18): e13836, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30251338

RESUMO

Mice with a deletion of the p50 subunit of the proinflammatory nuclear factor kappa B pathway (NF-κB p50) have reduced weight compared to wild-type control mice. However, the physiological underpinning of this phenotype remains unknown. This study addressed this issue. Compared to littermate controls, lean male p50 null mice (p50-/- ) had an increased metabolic rate (~20%) that was associated with increased skeletal muscle (SkM, ~35%), but not liver, oxidative metabolism. These metabolic alterations were accompanied by decreases in adiposity, and tissue and plasma triglyceride levels (all ~30%). Notably, there was a marked decrease in skeletal muscle, but not liver, DGAT2 gene expression (~70%), but a surprising reduction in muscle PPARα and CPT1 (both ~20%) gene expression. Exposure to a high-fat diet accentuated the diminished adiposity of p50-/- mice despite elevated caloric intake, whereas plasma triglycerides and free fatty acids (both ~30%), and liver (~40%) and SkM (~50%) triglyceride accumulation were again reduced compared to WT. Although SkM cytokine expression (IL-6 and TNFα, each ~100%) were increased in p50-/- mice, neither cytokine acutely increased SkM oxidative metabolism. We conclude that the reduced susceptibility to diet-induced obesity and dyslipidemia in p50-/- mice results from an increase in metabolic rate, which is associated with elevated skeletal muscle oxidative metabolism and decreased DGAT2 expression.


Assuntos
Metabolismo Basal/fisiologia , Mediadores da Inflamação/metabolismo , Músculo Esquelético/metabolismo , Subunidade p50 de NF-kappa B/deficiência , Obesidade/metabolismo , Estresse Oxidativo/fisiologia , Animais , Dieta Hiperlipídica/efeitos adversos , Dieta Hiperlipídica/tendências , Metabolismo Energético/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/prevenção & controle
9.
Neuropharmacology ; 133: 366-374, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29454840

RESUMO

Several studies on humans and mice support oxytocin's role in improving social behaviour, but its use in pharmacotherapy presents some important limiting factors. To date, it is emerging a pharmacological potential for melanocortin 4 receptor (MC4R) agonism in social deficits treatment. Recently, we demonstrated that the deletion of the NFKB1 gene, which encodes the p50 NF-κB subunit, causes impairment in social behaviours, with reductions in social interactions in mice. In this work, we tested the acute effects of THIQ, a selective melanocortin 4 receptor (MC4R) agonist. THIQ treatment increased social interactions both in wild type and p50-/- mice. In particular, after treatment with THIQ, p50-/- mice showed a prosocial behaviour analogous to that of basal WT mice. Moreover, intranasal treatment with an oxytocin antagonist blocked social interactions induced by THIQ, demonstrating that its prosocial effects are mediated by the oxytocin pathway. The data obtained reinforce using MC4R agonists to ameliorate social impairment in NDDs.


Assuntos
Ocitocina/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Transdução de Sinais/fisiologia , Transtornos do Comportamento Social/metabolismo , Animais , Relação Dose-Resposta a Droga , Comportamento Exploratório/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Relações Interpessoais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Subunidade p50 de NF-kappa B/deficiência , Subunidade p50 de NF-kappa B/genética , Ocitocina/genética , RNA Mensageiro/metabolismo , Radioimunoensaio , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/genética , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transtornos do Comportamento Social/tratamento farmacológico , Transtornos do Comportamento Social/genética , Tetra-Hidroisoquinolinas/uso terapêutico , Triazóis/uso terapêutico
10.
Front Immunol ; 9: 3148, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30761159

RESUMO

Background: NF-κB1 is a master regulator of both acquired and innate responses. NFKB1 loss-of-function mutations elicit a wide clinical phenotype with asymptomatic individuals at one end of the spectrum and patients with common variable immunodeficiency, combined immunodeficiency or autoinflammation at the other. Impairment of acquired and innate immunity and disseminated Mycobacterium genavense infection expands the clinical and immunological phenotype of NF-κB1 deficiency. Objective: Functional and molecular characterization of a patient with a novel phenotype of NF-κB1 deficiency. Methods: Circulating T, B, dendritic cell subsets and innate or unconventional T-cells were quantified. The cytokine production in stimulated whole blood samples was assessed and molecular characterization by next generation sequencing and gene expression assays were also performed. Results: We report a patient presenting with features of combined immunodeficiency (CID) and disseminated Mycobacterium genavense infection. Sequencing of genomic DNA identified a novel synonymous mutation (c.705G > A) in NFKB1 gene which resulted in exon 8 skipping and haploinsufficiency of the NF-κB1 subunit p50. The susceptibility to atypical mycobacterial infection has not been previously reported and may be the result of a dendritic cell deficiency. A selective deficiency of circulating follicular helper T (cTFH) cells responsible for mediating the differentiation of naive B cells into memory and plasma cells was also present in the patient. It could affect the maturation of innate or unconventional T cells where NF-κB1 could also be involved. Conclusion: These findings showed that the role of NF-κB1 in humans could be critical for the development of acquired and innate immunity and further highlights the role of human T cells in anti-mycobacterial immunity.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Síndromes de Imunodeficiência/complicações , Síndromes de Imunodeficiência/genética , Infecções por Mycobacterium/diagnóstico , Infecções por Mycobacterium/etiologia , Mycobacterium/imunologia , Subunidade p50 de NF-kappa B/deficiência , Biópsia , Medula Óssea/metabolismo , Criança , Citocinas/metabolismo , Humanos , Imunofenotipagem , Masculino , Mutação , Linhagem , Fenótipo , Pele/patologia
11.
Cell Death Dis ; 8(7): e2939, 2017 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-28726772

RESUMO

Tamoxifen (TAM) has recently been shown to cause acute gastric atrophy and metaplasia in mice. We have previously demonstrated that the outcome of Helicobacter felis infection, which induces similar gastric lesions in mice, is altered by deletion of specific NF-κB subunits. Nfkb1-/- mice developed more severe gastric atrophy than wild-type (WT) mice 6 weeks after H. felis infection. In contrast, Nfkb2-/- mice were protected from this pathology. We therefore hypothesized that gastric lesions induced by TAM may be similarly regulated by signaling via NF-κB subunits. Groups of five female C57BL/6 (WT), Nfkb1-/-, Nfkb2-/- and c-Rel-/- mice were administered 150 mg/kg TAM by IP injection. Seventy-two hours later, gastric corpus tissues were taken for quantitative histological assessment. In addition, groups of six female WT and Nfkb1-/- mice were exposed to 12 Gy γ-irradiation. Gastric epithelial apoptosis was quantified 6 and 48 h after irradiation. TAM induced gastric epithelial lesions in all strains of mice, but this was more severe in Nfkb1-/- mice than in WT mice. Nfkb1-/- mice exhibited more severe parietal cell loss than WT mice, had increased gastric epithelial expression of Ki67 and had an exaggerated gastric epithelial DNA damage response as quantified by γH2AX. To investigate whether the difference in gastric epithelial DNA damage response of Nfkb1-/- mice was unique to TAM-induced DNA damage or a generic consequence of DNA damage, we also assessed gastric epithelial apoptosis following γ-irradiation. Six hours after γ-irradiation, gastric epithelial apoptosis was increased in the gastric corpus and antrum of Nfkb1-/- mice. NF-κB1-mediated signaling regulates the development of gastric mucosal pathology following TAM administration. This is associated with an exaggerated gastric epithelial DNA damage response. This aberrant response appears to reflect a more generic sensitization of the gastric mucosa of Nfkb1-/- mice to DNA damage.


Assuntos
Dano ao DNA , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Subunidade p50 de NF-kappa B/deficiência , Gastropatias/metabolismo , Tamoxifeno/efeitos adversos , Animais , Feminino , Mucosa Gástrica/patologia , Infecções por Helicobacter/induzido quimicamente , Infecções por Helicobacter/genética , Infecções por Helicobacter/patologia , Helicobacter felis/metabolismo , Camundongos , Camundongos Knockout , Subunidade p52 de NF-kappa B/genética , Subunidade p52 de NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-rel/genética , Proteínas Proto-Oncogênicas c-rel/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Gastropatias/induzido quimicamente , Gastropatias/genética , Gastropatias/patologia , Tamoxifeno/farmacologia
13.
Chemistry ; 23(20): 4929-4935, 2017 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-28211965

RESUMO

NF-κB is a critical transcription factor that plays an important role in mediating inflammation, the immune response, and cell proliferation. The activation of NF-κB leads to an enhancement of proinflammatory mediator expression, which is implicated in the pathogenesis of a variety of diseases. Therefore, methods that allow the intracellular tracking of NF-κB are particularly attractive because they can provide information regarding the pathways or stimulation responses that are involved in the activation of NF-κB. In this work, we report a novel platform to track intracellular NF-κB by employing the conjugated iridium(III) complex 1, which was synthesized through the unique combination of a luminescent iridium(III) moiety with the natural product oridonin. Experiments conducted with p50 knockdown cells revealed that complex 1 could detect the p50 subunit of NF-κB in cellulo. Furthermore, complex 1 tracked NF-κB translocation induced by tumor necrosis factor-α (TNF-α) as a model stimulus, without affecting the translocation process itself. To the best of our knowledge, complex 1 is the first metal-based compound that has been reported to be capable of monitoring intracellular NF-κB in living cells.


Assuntos
Complexos de Coordenação/síntese química , Diterpenos do Tipo Caurano/química , Irídio/química , NF-kappa B/metabolismo , Complexos de Coordenação/química , Complexos de Coordenação/metabolismo , Células HeLa , Humanos , Microscopia de Fluorescência , Subunidade p50 de NF-kappa B/deficiência , Subunidade p50 de NF-kappa B/genética , Ativação Transcricional/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
14.
Oncogene ; 35(18): 2299-310, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-26300007

RESUMO

Nuclear factor-κB (NF-κB) is generally believed to be pro-tumorigenic. Here we report a tumor-suppressive function for NF-κB1, the prototypical member of NF-κB. While NF-κB1 downregulation is associated with high lung cancer risk in humans and poor patient survival, NF-κB1-deficient mice are more vulnerable to lung tumorigenesis induced by the smoke carcinogen, urethane. Notably, the tumor-suppressive function of NF-κB1 is independent of its classical role as an NF-κB factor, but instead through stabilization of the Tpl2 kinase. NF-κB1-deficient tumors exhibit 'normal' NF-κB activity, but a decreased protein level of Tpl2. Reconstitution of Tpl2 or the NF-κB1 p105, but not p50 (the processed product of p105), inhibits the tumorigenicity of NF-κB1-deficient lung tumor cells. Remarkably, Tpl2-knockout mice resemble NF-κB1 knockouts in urethane-induced lung tumorigenesis. Mechanistic studies indicate that p105/Tpl2 signaling is required for suppressing urethane-induced lung damage and inflammation, and activating mutations of the K-Ras oncogene. These studies reveal an unexpected, NF-κB-independent but Tpl2-depenednt role of NF-κB1 in lung tumor suppression. These studies also reveal a previously unexplored role of p105/Tpl2 signaling in lung homeostasis.


Assuntos
Carcinogênese , Neoplasias Pulmonares/patologia , MAP Quinase Quinase Quinases/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Carcinogênese/induzido quimicamente , Carcinogênese/genética , Linhagem Celular Tumoral , Estabilidade Enzimática/efeitos dos fármacos , Estabilidade Enzimática/genética , Técnicas de Inativação de Genes , Genes ras/genética , Homeostase/efeitos dos fármacos , Homeostase/genética , Humanos , Pulmão/efeitos dos fármacos , Pulmão/patologia , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , Camundongos , Mutação , Subunidade p50 de NF-kappa B/deficiência , Subunidade p50 de NF-kappa B/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Uretana/farmacologia
15.
Immunol Cell Biol ; 94(1): 66-78, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26084385

RESUMO

Defects in apoptosis can cause autoimmune disease. Loss-of-function mutations in the 'death receptor' FAS impair the deletion of autoreactive lymphocytes in the periphery, leading to progressive lymphadenopathy and systemic lupus erythematosus-like autoimmune disease in mice (Fas(lpr/lpr) (mice homozygous for the lymphoproliferation inducing spontaneous mutation)) and humans. The REL/nuclear factor-κB (NF-κB) transcription factors regulate a broad range of immune effector functions and are also implicated in various autoimmune diseases. We generated compound mutant mice to investigate the individual functions of the NF-κB family members NF-κB1, NF-κB2 and c-REL in the various autoimmune pathologies of Fas(lpr/lpr) mutant mice. We show that loss of each of these transcription factors resulted in amelioration of many classical features of autoimmune disease, including hypergammaglobulinaemia, anti-nuclear autoantibodies and autoantibodies against tissue-specific antigens. Remarkably, only c-REL deficiency substantially reduced immune complex-mediated glomerulonephritis and extended the lifespan of Fas(lpr/lpr) mice. Interestingly, compared with the Fas(lpr/lpr) animals, Fas(lpr/lpr)nfkb2(-/-) mice presented with a dramatic acceleration and augmentation of lymphadenopathy that was accompanied by severe lung pathology due to extensive lymphocytic infiltration. The Fas(lpr/lpr)nfkb1(-/-) mice exhibited the combined pathologies caused by defects in FAS-mediated apoptosis and premature ageing due to loss of NF-κB1. These findings demonstrate that different NF-κB family members exert distinct roles in the development of the diverse autoimmune and lymphoproliferative pathologies that arise in Fas(lpr/lpr) mice, and suggest that pharmacological targeting of c-REL should be considered as a strategy for therapeutic intervention in autoimmune diseases.


Assuntos
Lúpus Eritematoso Sistêmico/complicações , Doenças Linfáticas/complicações , Subunidade p50 de NF-kappa B/deficiência , Subunidade p52 de NF-kappa B/deficiência , Proteínas Proto-Oncogênicas c-rel/metabolismo , Receptor fas/metabolismo , Animais , Autoanticorpos/sangue , Quimiocinas/sangue , Quimiocinas/metabolismo , Dermatite/sangue , Dermatite/complicações , Dermatite/imunologia , Fatores de Transcrição Forkhead/metabolismo , Genótipo , Hipergamaglobulinemia/sangue , Hipergamaglobulinemia/complicações , Tolerância Imunológica/imunologia , Leucócitos/patologia , Longevidade , Lúpus Eritematoso Sistêmico/sangue , Doenças Linfáticas/sangue , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Subunidade p50 de NF-kappa B/metabolismo , Subunidade p52 de NF-kappa B/metabolismo , Especificidade de Órgãos , Esplenomegalia/sangue , Fatores de Transcrição/metabolismo , Proteína AIRE
17.
FEBS J ; 282(1): 129-41, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25312244

RESUMO

Although the transcription factor nuclear factor-κB (NF-κB) is known to regulate cell death and survival, its precise role in cell death within the central nervous system remains unknown. We previously reported that mice with a homozygous deficiency for NF-κBp50 spontaneously develop optic neuropathy. The aim of the present study was to demonstrate the expression and activation of the proapoptotic factor(s) that mediate optic neuropathy in p50-deficient mice. Recombination-activating gene (Rag) 1 is known to activate the recombination of immunoglobulin V(D)J. In this study, experiments with genetically engineered mice revealed the involvement of Rag1 expression in apoptosis of Brn3a-positive retinal ganglion cells, and also demonstrated the specific effect of p50 deficiency on the activation of Rag1 gene transcription. Furthermore, genetic analysis of murine neuronal stem-like cells clarified the biological significance of Rag1 in N-methyl-D-aspartate-induced neuronal apoptosis. We also detected the apoptosis-regulating factors Bax and cleaved caspase 3, 8 and 9 in HEK293 cells transfected-molecule of Rag1, and a human histological examination revealed the expression of Rag1 in retinal ganglion cells. The results of the present study indicate that Rag1 plays a role in optic neuropathy as a proapoptotic candidate in p50-deficient mice. This finding may lead to new therapeutic targets in optic neuropathy.


Assuntos
Proteínas de Homeodomínio/fisiologia , Doenças do Nervo Óptico/patologia , Doenças do Nervo Óptico/fisiopatologia , Animais , Apoptose/genética , Apoptose/fisiologia , Técnicas de Silenciamento de Genes , Genes RAG-1 , Células HEK293 , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Neurológicos , N-Metilaspartato/toxicidade , Subunidade p50 de NF-kappa B/deficiência , Subunidade p50 de NF-kappa B/genética , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Doenças do Nervo Óptico/etiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Transdução de Sinais , Fator de Transcrição Brn-3A/metabolismo
18.
Biochem Biophys Res Commun ; 447(4): 563-8, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24751519

RESUMO

p27(Kip1) is a potent inhibitor of the cyclin-dependent kinases that drive G1 to S phase transition. Since deregulation of p27(Kip1) is found in many malignancies and is associated with the poor prognosis, elucidation of the molecular bases for regulation of p27(Kip1) expression is of great significance, not only in providing insight into the understanding of biological p27(Kip1), but also in the development of new cancer therapeutic tactics. We here explored the inhibitory regulation of IKKß on p27(Kip1) expression following arsenite exposure. We found that although the basal level of p27(Kip1) expression in the IKKß(-/-) cells is much lower than that in the IKKß(+/+) cells, the deletion of IKKß in the MEFs led to a marked increase in p27(Kip1) protein induction due to arsenite exposure in comparison to that in the IKKß(+/+) cells. The IKKß regulatory effect on p27(Kip1) expression was also verified in the IKKß(-/-) and IKKß(-/-) cells with IKKß reconstitutional expression, IKKß(-/-) (IKKß). Further studies indicated that IKKß-mediated p27(Kip1) downregulation occurred at protein degradation level via p65-dependent and p50-independent manner. Moreover, the results obtained from the comparison of arsenite-induced GSK3ß activation among transfectants of WT, IKKß(-/-) and IKKß(-/-) (IKKß), and the utilization of GSKß shRNA, demonstrated that IKKß regulation of p27 protein degradation was mediated by GSK3ß following arsenite exposure.


Assuntos
Arsenitos/farmacologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Quinase I-kappa B/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Antineoplásicos/farmacologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/deficiência , Inibidor de Quinase Dependente de Ciclina p27/genética , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Técnicas de Inativação de Genes , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células HeLa , Humanos , Quinase I-kappa B/deficiência , Quinase I-kappa B/genética , Camundongos , Subunidade p50 de NF-kappa B/deficiência , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/metabolismo , Fosforilação , Proteólise/efeitos dos fármacos , RNA Interferente Pequeno/genética , Fator de Transcrição RelA/deficiência , Fator de Transcrição RelA/genética
19.
Neurobiol Dis ; 64: 16-29, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24345324

RESUMO

Inflammation is a major mechanism of acute brain injury and chronic neurodegeneration. This neuroinflammation is known to be substantially regulated by the transcription factor NF-κB, which is predominantly found in the form of heterodimer of p65 (RelA) and p50 subunit, with p50/p50 homodimers being also common. The p65 subunit has a transactivation domain, whereas p50 is chiefly involved in DNA binding. Binding of the p65/p50 heterodimers is thought to induce expression of numerous proinflammatory genes in microglia. Here we show that cultured microglia deficient for the gene (Nfkb1) encoding p50 subunit show reduced induction of proinflammatory mediators, increased expression of anti-inflammatory genes, and increased expression of CD45, an immunoregulatory molecule, in response to lipopolysaccharide (LPS) exposure, but increased capacity to take up ß-amyloid (Aß) which is associated with enhanced release of tumor necrosis factor alpha (TNFα). However, Nfkb1 deficiency strongly increases leukocyte infiltration and the expression of proinflammatory genes in response to intrahippocampal administration of LPS. Also, when crossing Nfkb1 deficient mice with APdE9 transgenic mice the expression of proinflammatory genes was strongly enhanced, whereas Aß burden was slightly but significantly reduced. These alterations in expression of inflammatory mediators in Nfkb1 deficient mice were associated with reduced expression of CD45. Our data demonstrates a crucial and complex role p50 subunit of NF-κB in brain inflammation, especially in regulating the phenotype of microglia after acute and chronic inflammatory insults relevant to clinical conditions, contributing to both pro-inflammatory and anti-inflammatory responses of microglia, infiltration of leukocytes, and clearance of Aß in Alzheimer's disease.


Assuntos
Hipocampo/imunologia , Microglia/imunologia , Subunidade p50 de NF-kappa B/deficiência , Subunidade p50 de NF-kappa B/fisiologia , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica/imunologia , Humanos , Antígenos Comuns de Leucócito/metabolismo , Leucócitos/fisiologia , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Subunidade p50 de NF-kappa B/genética , Fagocitose/fisiologia , Presenilina-1/genética , Presenilina-1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
20.
Aging (Albany NY) ; 6(11): 931-43, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25553648

RESUMO

NF-κB is a major regulator of age-dependent gene expression and the p50/NF-κB1 subunit is an integral modulator of NF-κB signaling. Here, we examined Nfkb1-/- mice to investigate the relationship between this subunit and aging. Although Nfkb1-/- mice appear similar to littermates at six months of age, by 12 months they have a higher incidence of several observable age-related phenotypes. In addition, aged Nfkb1-/- animals have increased kyphosis, decreased cortical bone, increased brain GFAP staining and a decrease in overall lifespan compared to Nfkb1+/+. In vitro, serially passaged primary Nfkb1-/- MEFs have more senescent cells than comparable Nfkb1+/+ MEFs. Also, Nfkb1-/- MEFs have greater amounts of phospho-H2AX foci and lower levels of spontaneous apoptosis than Nfkb1+/+, findings that are mirrored in the brains of Nfkb1-/- animals compared to Nfkb1+/+. Finally, in wildtype animals a substantial decrease in p50 DNA binding is seen in aged tissue compared to young. Together, these data show that loss of Nfkb1 leads to early animal aging that is associated with reduced apoptosis and increased cellular senescence. Moreover, loss of p50 DNA binding is a prominent feature of aged mice relative to young. These findings support the strong link between the NF-κB pathway and mammalian aging.


Assuntos
Senilidade Prematura , Envelhecimento/metabolismo , Subunidade p50 de NF-kappa B/deficiência , Fatores Etários , Envelhecimento/genética , Envelhecimento/patologia , Animais , Apoptose , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Senescência Celular , DNA/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Genótipo , Gliose , Histonas/metabolismo , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Cifose/genética , Cifose/metabolismo , Cifose/patologia , Longevidade , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidade p50 de NF-kappa B/genética , Fenótipo , Fosforilação , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...