Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Biomolecules ; 13(9)2023 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-37759710

RESUMO

Though originally characterized as an inactive or transcriptionally repressive factor, the NF-κB p50 homodimer has become appreciated as a physiologically relevant driver of specific target gene expression. By virtue of its low affinity for cytoplasmic IκB protein inhibitors, p50 accumulates in the nucleus of resting cells, where it is a binding target for the transcriptional co-activator IκBζ. In this study, we employed X-ray crystallography to analyze the structure of the p50 homodimer on κB DNA from the promoters of human interleukin-6 (IL-6) and neutrophil-gelatinase-associated lipocalin (NGAL) genes, both of which respond to IκBζ. The NF-κB p50 homodimer binds 11-bp on IL-6 κB DNA, while, on NGAL κB DNA, the spacing is 12-bp. This begs the question: what DNA binding mode is preferred by NF-κB p50 homodimer? To address this, we engineered a "Test" κB-like DNA containing the core sequence 5'-GGGGAATTCCCC-3' and determined its X-ray crystal structure in complex with p50. This revealed that, when presented with multiple options, NF-κB p50 homodimer prefers to bind 11-bp, which necessarily imposes asymmetry on the complex despite the symmetry inherent in both the protein and its target DNA, and that the p50 dimerization domain can contact DNA via distinct modes.


Assuntos
Interleucina-6 , Subunidade p50 de NF-kappa B , NF-kappa B , Humanos , Cristalografia por Raios X , DNA , Lipocalina-2 , Raios X , Subunidade p50 de NF-kappa B/química , Subunidade p50 de NF-kappa B/fisiologia
2.
J Mol Neurosci ; 71(1): 101-111, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32557145

RESUMO

This study was designed to investigate the effect of naringin in oxygen-glucose deprivation/reoxygenation (OGD/R) model and its mechanism. The target gene of naringin and the enriched pathways of the gene were searched and identified using bioinformatics analysis. Then OGD/R model was built using PC12 cells, after which the cells were treated with different concentrations of naringin. Subsequently, cell proliferation and apoptosis were evaluated by cell counting kit-8 (CCK-8) and flow cytometry assays, respectively. Meanwhile, the expression of NFKB1 in PC12 cells underwent OGD/R-induced injury was detected by qRT-PCR, while apoptosis-related and pathway-related proteins were checked by Western blot. DCF-DA kit was utilized to measure the level of ROS. Our results revealed that NFKB1, which was upregulated in MACO rats and OGD/R-treated PC12 cells, was a target gene of naringin. Naringin could alleviate OGD/R-induced injury via promoting the proliferation, and repressing the apoptosis of PC12 cells through regulating the expression of NFKB1 and apoptosis-associated proteins and ROS level. Besides, the depletion of NFKB1 was positive to cell proliferation but negative to cell apoptosis. Moreover, the depletion of NFKB1 enhanced the influences of naringin on cell proliferation and apoptosis as well as the expression of apoptosis-related proteins and ROS level. Western blotting indicated that both naringin treatment and depletion of NFKB1 could increase the expression of HIF-1α, p-AKT, and p-mTOR compared with OGD/R group. What's more, treatment by naringin and si-NFKB1 together could significantly increase these effects. Nevertheless, the expression of AKT and mTOR among each group was almost not changed. In conclusion, naringin could prevent the OGD/R-induced injury in PC12 cells in vitro by targeting NFKB1 and regulating HIF-1α/AKT/mTOR-signaling pathway, which might provide novel ideas for the therapy of cerebral ischemia-reperfusion (I/R) injury.


Assuntos
Flavanonas/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Subunidade p50 de NF-kappa B/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Serina-Treonina Quinases TOR/fisiologia , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Flavanonas/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Ontologia Genética , Glucose/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/genética , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/fisiologia , Oxigênio/farmacologia , Células PC12 , Fitoterapia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Proteína X Associada a bcl-2/biossíntese , Proteína X Associada a bcl-2/genética
3.
Int J Mol Sci ; 20(3)2019 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-30717434

RESUMO

The role of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) has been highlighted in mechanisms underlying inflammatory and neuropathic pain processes. The present study was designed to investigate whether NF-κB signaling is associated with pain-related neuropeptide expression in patients with chronic back pain related to degenerative disc disease (DDD). Intervertebral disc (IVD) tissues were collected from forty DDD patients undergoing disc replacement or fusion surgery, and from eighteen postmortem (PM) control subjects. RELA, NFKB1, CGRP, TAC1, TRPV1, and MMP-3 gene expression were analyzed by RT-qPCR, while NF-κB subunit RelA and NF-κB1⁻DNA binding in nuclear extracts and calcitonin gene related peptide (CGRP), substance P (SP), and transient receptor potential, subfamily V, member 1 (TRPV1) protein levels in cytosolic extracts of tissues were assessed by enzyme-linked immunosorbent assay (ELISA). An upregulated NF-κB1⁻DNA binding, and higher CGRP and TRPV1 protein levels were observed in DDD patients compared to PM controls. In DDD patients, NF-κB1⁻DNA binding was positively correlated with nuclear RelA levels. Moreover, NF-κB1⁻DNA binding was positively associated with TRPV1 and MMP-3 gene and SP and TRPV1 protein expression in DDD patients. Our results indicate that the expression of SP and TRPV1 in IVD tissues was associated with NF-κB activation. Moreover, NF-κB may be involved in the generation or maintenance of peripheral pain mechanisms by the regulation of pain-related neuropeptide expression in DDD patients.


Assuntos
Degeneração do Disco Intervertebral/metabolismo , NF-kappa B/metabolismo , Dor/metabolismo , Transdução de Sinais , Substância P/genética , Canais de Cátion TRPV/genética , Adulto , Feminino , Regulação da Expressão Gênica , Humanos , Disco Intervertebral/metabolismo , Disco Intervertebral/fisiopatologia , Degeneração do Disco Intervertebral/complicações , Degeneração do Disco Intervertebral/genética , Degeneração do Disco Intervertebral/fisiopatologia , Masculino , Pessoa de Meia-Idade , NF-kappa B/fisiologia , Subunidade p50 de NF-kappa B/metabolismo , Subunidade p50 de NF-kappa B/fisiologia , Dor/etiologia , Dor/genética , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelA/fisiologia
4.
Cancer Immunol Immunother ; 67(10): 1491-1503, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30030559

RESUMO

High-grade gliomas harbor abundant myeloid cells that suppress anti-tumor immunity and support tumor growth. Targeting transcription factors, such as NF-κB p50, that mediate suppressive myeloid M2 polarization may prove therapeutic. GL261-Luc glioblastoma cells were inoculated into wild-type and p50-/- mice, followed by analysis of tumor growth, survival, tumor myeloid cells, and T cells. The absence of host p50 slows tumor growth and enables regression in 30% of recipients, leading to prolonged survival. Tumors developing in p50-/- mice possess a greater concentration of tumor-infiltrating myeloid cells (TIMs) than those in wild-type mice. TIMs are predominantly F4/80hi macrophages which, along with tumor-associated microglia, express increased pro-inflammatory M1 and reduced immune-suppressive M2 markers. In p50-/- mice, total tumor CD4 T cells are threefold more abundant, whereas CD8 T-cell numbers are unchanged, and both produce increased IFNγ and Granzyme B. Naïve splenic p50-/- CD8 T cells manifest increased activation, whereas naïve p50-/- and WT CD4 T cells show similar Th1, Th2, and Th17 polarization. Antibody targeting CD4, but not CD8, fully obviates the p50-/- survival advantage. Combined CD4 and CD8 T-cell depletion reverses myeloid M2 polarization in wild-type hosts, without affecting myeloid M1 polarization in p50-/- hosts. Finally, gliomas grow similarly in p50(f/f) and p50(f/f);Lysozyme-Cre mice, the latter having reduced p50 specifically in myeloid cells and tumor microglia. Thus, high-grade glioma T cells play a key role in directing M2 polarization of tumor myeloid cells, and reducing NF-κB p50 in both tumor myeloid cells and T cells may contribute to glioma therapy.


Assuntos
Glioblastoma/prevenção & controle , Macrófagos/imunologia , Células Mieloides/imunologia , Subunidade p50 de NF-kappa B/fisiologia , Linfócitos T/imunologia , Animais , Células Cultivadas , Glioblastoma/imunologia , Glioblastoma/mortalidade , Ativação Linfocitária , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Taxa de Sobrevida , Linfócitos T/metabolismo
5.
Cancer Immunol Res ; 6(5): 578-593, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29588321

RESUMO

Although tumor-associated macrophages (TAM) display a M2-skewed tumor-promoting phenotype in most cancers, in colorectal cancer, both TAM polarization and its impact remain controversial. We investigated the role of the M2-polarizing p50 NF-κB subunit in orchestrating TAM phenotype, tumor microenvironment composition, and colorectal cancer progression. We first demonstrated, by parallel studies in colitis-associated cancer (CAC) and in genetically driven ApcMin mouse models, that the p50-dependent inhibition of M1-polarized gut inflammation supported colorectal cancer development. In accordance with these studies, p50-/- mice displayed exacerbated CAC with fewer and smaller tumors, along with enhanced levels of M1/Th1 cytokines/chemokines, including IL12 and CXCL10, whose administration restrained CAC development in vivo The inflammatory profile supporting tumor resistance in colons from p50-/- tumor bearers correlated inversely with TAM load and positively with both recruitment of NK, NKT, CD8+ T cells and number of apoptotic tumor cells. In agreement, myeloid-specific ablation of p50 promoted tumor resistance in mice, whereas in colorectal cancer patients, a high number of p50+ TAMs at the invasive margin was associated with decreased IL12A and TBX21 expression and worse postsurgical outcome. Our findings point to p50 involvement in colorectal cancer development, through its engagement in the protumor activation of macrophages, and identify a candidate for prognostic and target therapeutic intervention. Cancer Immunol Res; 6(5); 578-93. ©2018 AACR.


Assuntos
Neoplasias Colorretais/patologia , Mediadores da Inflamação/fisiologia , Inflamação/complicações , Subunidade p50 de NF-kappa B/fisiologia , Animais , Polaridade Celular/genética , Células Cultivadas , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/metabolismo , Progressão da Doença , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Ativação de Macrófagos/genética , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
6.
Toxicol Appl Pharmacol ; 338: 30-42, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29129777

RESUMO

Trichloroethylene (TCE), a prevalent environmental contaminant, is a potent renal and hepatic toxicant through metabolites such as S-(1, 2-dichlorovinyl)-l-cysteine (DCVC). However, effects of TCE on other target organs such as the placenta have been minimally explored. Because elevated apoptosis and lipid peroxidation in placenta have been observed in pregnancy morbidities involving poor placentation, we evaluated the effects of DCVC exposure on apoptosis and lipid peroxidation in a human extravillous trophoblast cell line, HTR-8/SVneo. We exposed the cells in vitro to 10-100µM DCVC for various time points up to 24h. Following exposure, we measured apoptosis using flow cytometry, caspase activity using luminescence assays, gene expression using qRT-PCR, and lipid peroxidation using a malondialdehyde quantification assay. DCVC significantly increased apoptosis in time- and concentration-dependent manners (p<0.05). DCVC also significantly stimulated caspase 3, 7, 8 and 9 activities after 12h (p<0.05), suggesting that DCVC stimulates the activation of both the intrinsic and extrinsic apoptotic signaling pathways simultaneously. Pre-treatment with the tBID inhibitor Bl-6C9 partially reduced DCVC-stimulated caspase 3 and 7 activity, signifying crosstalk between the two pathways. Additionally, DCVC treatment increased lipid peroxidation in a concentration-dependent manner. Co-treatment with the antioxidant peroxyl radical scavenger (±)-α-tocopherol attenuated caspase 3 and 7 activity, suggesting that lipid peroxidation mediates DCVC-induced apoptosis in extravillous trophoblasts. Our findings suggest that DCVC-induced apoptosis and lipid peroxidation in extravillous trophoblasts could contribute to poor placentation if similar effects occur in vivo in response to TCE exposure, indicating that further studies into this mechanism are warranted.


Assuntos
Apoptose/efeitos dos fármacos , Cisteína/análogos & derivados , Peroxidação de Lipídeos/efeitos dos fármacos , Placenta/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Proteínas de Ciclo Celular/fisiologia , Células Cultivadas , Cisteína/toxicidade , Feminino , Humanos , Subunidade p50 de NF-kappa B/fisiologia , Proteínas Nucleares/fisiologia , Placenta/citologia , Placenta/metabolismo , Gravidez , Primeiro Trimestre da Gravidez , Espécies Reativas de Oxigênio/metabolismo
8.
J Virol ; 91(7)2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28100608

RESUMO

Cyclic GMP-AMP synthase (cGAS) is a newly identified DNA sensor that recognizes foreign DNA, including the genome of herpes simplex virus 1 (HSV-1). Upon binding of viral DNA, cGAS produces cyclic GMP-AMP, which interacts with and activates stimulator of interferon genes (STING) to trigger the transcription of antiviral genes such as type I interferons (IFNs), and the production of inflammatory cytokines. HSV-1 UL24 is widely conserved among members of the herpesviruses family and is essential for efficient viral replication. In this study, we found that ectopically expressed UL24 could inhibit cGAS-STING-mediated promoter activation of IFN-ß and interleukin-6 (IL-6), and UL24 also inhibited interferon-stimulatory DNA-mediated IFN-ß and IL-6 production during HSV-1 infection. Furthermore, UL24 selectively blocked nuclear factor κB (NF-κB) but not IFN-regulatory factor 3 promoter activation. Coimmunoprecipitation analysis demonstrated that UL24 bound to the endogenous NF-κB subunits p65 and p50 in HSV-1-infected cells, and UL24 was also found to bind the Rel homology domains (RHDs) of these subunits. Furthermore, UL24 reduced the tumor necrosis factor alpha (TNF-α)-mediated nuclear translocation of p65 and p50. Finally, mutational analysis revealed that the region spanning amino acids (aa) 74 to 134 of UL24 [UL24(74-134)] is responsible for inhibiting cGAS-STING-mediated NF-κB promoter activity. For the first time, UL24 was shown to play an important role in immune evasion during HSV-1 infection.IMPORTANCE NF-κB is a critical component of the innate immune response and is strongly induced downstream of most pattern recognition receptors (PRRs), leading to the production of IFN-ß as well as a number of inflammatory chemokines and interleukins. To establish persistent infection, viruses have evolved various mechanisms to counteract the host NF-κB pathway. In the present study, for the first time, HSV-1 UL24 was demonstrated to inhibit the activation of NF-κB in the DNA sensing signal pathway via binding to the RHDs of the NF-κB subunits p65 and p50 and abolishing their nuclear translocation.


Assuntos
Herpesvirus Humano 1/fisiologia , Subunidade p50 de NF-kappa B/fisiologia , Transdução de Sinais , Fator de Transcrição RelA/fisiologia , Proteínas Virais/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Chlorocebus aethiops , Regulação Viral da Expressão Gênica , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapas de Interação de Proteínas , Células Vero
9.
J Nephrol ; 30(1): 73-79, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26385798

RESUMO

The amiloride-sensitive epithelial sodium channel (ENaC), located in the apical membrane in the cortical collecting duct of the kidney, mediates the fine-tuned regulation of external Na+ balance. Expression of the alpha-subunit of ENaC (ENaC-α) is regulated by a number of factors in the lung, including transcription factor nuclear factor kappa B (NF-κB). In the present study, we examined the effect of IKKß/p65/p50 on ENaC-α in a murine cortical collecting duct cell line that endogenously expresses ENaC, mpkCCDc14 (CCD) cells. Aldosterone exposure led to up-regulation of ENaC-α and IKKß, and nuclear p65 and p50. Knockdown of IKKß or p65 exhibited >60 % reduction of aldosterone-induced ENaC-α mRNA levels. Chromatin immunoprecipitation and electrophoretic mobility shift assays demonstrated a specific interaction between p65/p50 and ENaC-α gene promoter, which was further confirmed using luciferase reporter-gene vectors transiently transfected into CCD cells. Taken together these data support an important role for p65/p50 in the direct regulation of ENaC-α transcription and have important implications for understanding the role of NF-κB in the regulation of renal function.


Assuntos
Aldosterona/farmacologia , Canais Epiteliais de Sódio/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Rim/metabolismo , Subunidade p50 de NF-kappa B/fisiologia , Fator de Transcrição RelA/fisiologia , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Quinase I-kappa B/fisiologia , Camundongos , Regiões Promotoras Genéticas , RNA Mensageiro/análise
10.
J Immunol ; 197(10): 3927-3935, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27815442

RESUMO

To stimulate a productive T cell response, dendritic cells (DC) must undergo maturation characterized by heightened cell surface expression of MHC and costimulatory molecules as well as cytokine production. Conversely, the inhibition of DC maturation is a central mechanism of immune tolerance. The control of the DC maturation process relies on the integration of several cellular stimulatory or inhibitory signals. The soluble factors and their receptors controlling this central aspect of DC biology are incompletely characterized. We show that murine bone marrow-derived DC (BMDC) maturation induced by LPS, as opposed to polyinosinic:polycytidylic acid or cytosine-phosphate-guanine, is robustly inhibited by vascular endothelial growth factor (VEGF), a previously identified immunosuppressive cytokine. Using BMDC from wild type and conditional knockout mice, we show that neuropilin-1 (NRP-1), a known receptor of VEGF, is necessary to suppress LPS-dependent BMDC maturation. The absence of NRP-1 had no ostensible effects on the biology of BMDC in the absence of VEGF. However, NRP-1-deficient BMDC remained completely insensitive to the VEGF-dependent inhibition of BMDC maturation in culture. In the presence of VEGF, NRP-1 directly interacted with the LPS receptor TLR4 and suppressed downstream signaling through ERK and NF-κß, resulting in a sharp inhibition of MHC class II and costimulatory molecules (CD40, CD86) expression as well as proinflammatory cytokine production. Consequently, we identify NRP-1 as a target to optimize DC maturation within environments that are rich in VEGF, such as tumors.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/fisiologia , Neuropilina-1/imunologia , Neuropilina-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Antígeno B7-2/efeitos dos fármacos , Antígeno B7-2/genética , Células da Medula Óssea/imunologia , Células da Medula Óssea/fisiologia , Antígenos CD40/efeitos dos fármacos , Antígenos CD40/genética , Diferenciação Celular , Células Cultivadas , Citocinas/biossíntese , Citocinas/efeitos dos fármacos , Citocinas/genética , Células Dendríticas/imunologia , Genes MHC da Classe II/efeitos dos fármacos , Genes MHC da Classe II/genética , Tolerância Imunológica/efeitos dos fármacos , Lipopolissacarídeos/imunologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Subunidade p50 de NF-kappa B/fisiologia , Neuropilina-1/deficiência , Poli I-C/farmacologia , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
11.
FEBS J ; 283(10): 1812-22, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26663363

RESUMO

The pleiotropic consequences of nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB) pathway activation result from the combinatorial effects of the five subunits that form the homo- and heterodimeric NF-κB complexes. Although biochemical and gene knockout studies have demonstrated overlapping and distinct functions for these proteins, much is still not known about the mechanisms determining context-dependent functions, the formation of different dimer complexes and transcriptional control in response to diverse stimuli. Here we discuss recent results that reveal that the nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NFKB1) (p105/p50) subunit is an important regulator of NF-κB activity in vivo. These effects are not restricted to being a dimer partner for other NF-κB subunits. Rather p50 homodimers have a critical role as suppressors of the NF-κB response, while the p105 precursor has a variety of NF-κB-independent functions. The importance of Nfkb1 function can be seen in mouse models, where Nfkb1(-/-) mice display increased inflammation and susceptibility to certain forms of DNA damage, leading to cancer, and a rapid ageing phenotype. In humans, low expression of Kip1 ubiquitination-promoting complex 1 (KPC1), a ubiquitin ligase required for p105 to p50 processing, was shown to correlate with a reduction in p50 and glioblastoma incidence. Therefore, while the majority of research in this field has focused on the upstream signalling pathways leading to NF-κB activation or the function of other NF-κB subunits, such as RelA (p65), these data demonstrate a critical role for NFKB1, potentially revealing new strategies for targeting this pathway in inflammatory diseases and cancer.


Assuntos
Envelhecimento/fisiologia , Inflamação/fisiopatologia , Subunidade p50 de NF-kappa B/fisiologia , Neoplasias/fisiopatologia , Animais , Dimerização , Humanos , Sistema Imunitário/fisiologia , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B/química , Subunidade p50 de NF-kappa B/genética
12.
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
13.
Oncogene ; 33(8): 996-1005, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23396362

RESUMO

The biological function of NF-κB1 (p50) in the regulation of protein expression is far from well understood owing to the lack of a transcriptional domain. Here, we report a novel function of p50 in its regulation of p53 protein translation under stress conditions. We found that the deletion of p50 (p50-/-) impaired arsenite-induced p53 protein expression, which could be restored after reconstitutive expression of HA-p50 in p50-/- cells, p50-/-(Ad-HA-p50). Further studies indicated that the amounts of p53 mRNA, p53 promoter-driven transcription activity and p53 protein degradation were comparable between wild-type and p50-/- cells. Moreover, we found that p50 was crucial for Akt/S6 ribosomal protein activation via inhibition of the translation of the PH domain and leucine-rich repeat protein phosphatases 1 (PHLPP1), a phosphatase of Akt. Further studies showed that p50-mediated upregulation of miR-190 was responsible for the inhibition of PHLPP1 translation by targeting the 3'-untranslated region of its mRNA. Collectively, we have identified a novel function of p50 in modulating p53 protein translation via regulation of the miR-190/PHLPP1/Akt-S6 ribosomal protein pathway.


Assuntos
Regulação para Baixo , MicroRNAs/genética , Subunidade p50 de NF-kappa B/fisiologia , Proteínas Nucleares/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Biossíntese de Proteínas/fisiologia , Proteína Supressora de Tumor p53/genética , Animais , Arsenitos/farmacologia , Sequência de Bases , Primers do DNA , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Mol Cell Biol ; 33(24): 4857-71, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24100018

RESUMO

Regulation of microbially induced cytokine secretion is critical in intestinal immune homeostasis. NOD2, the Crohn's disease-associated bacterial peptidoglycan sensor, activates the NF-κB pathway. After chronic NOD2 stimulation in human macrophages, cytokine secretion is significantly attenuated, similar to the situation in the intestinal environment. We find that NF-κB1 (p105/p50) expression is upregulated with chronic NOD2 stimulation and is required for attenuation of cytokine secretion in vitro in human macrophages and in vivo in mice. Upon chronic NOD2 stimulation, regulation of both activating (H3K4Me2 and H4Ac) and inhibitory (H3K27Me3) histone modifications was observed within cytokine gene promoters; these outcomes were NF-κB1 dependent. In addition to enhanced binding to cytokine gene promoters with chronic NOD2 stimulation, NF-κB1 bound to the promoter of the transcriptional repressor, ATF3. ATF3 was then induced and bound to cytokine gene promoters; both features were impaired upon NF-κB1 knockdown. Restoring ATF3 expression under NF-κB1 knockdown conditions restored NOD2-mediated cytokine downregulation. Finally, NF-κB1 and ATF3 cooperate with other inhibitory pathways, including IRAKM and secreted mediators, to downregulate cytokine secretion after chronic NOD2 stimulation. Therefore, we identify NF-κB1 and ATF3 as critical mechanisms through which NOD2 downregulates cytokines and contributes to intestinal immune homeostasis.


Assuntos
Fator 3 Ativador da Transcrição/metabolismo , Citocinas/metabolismo , Subunidade p50 de NF-kappa B/fisiologia , Proteína Adaptadora de Sinalização NOD2/metabolismo , Animais , Células Cultivadas , Citocinas/genética , Epigênese Genética/imunologia , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Humanos , Íleo/imunologia , Íleo/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Adaptadora de Sinalização NOD1/metabolismo , Cultura Primária de Células , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Ativação Transcricional
15.
PLoS One ; 8(6): e67620, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23826329

RESUMO

The immune response is a concerted dynamic multi-cellular process. Upon infection, the dynamics of lymphocyte populations are an aggregate of molecular processes that determine the activation, division, and longevity of individual cells. The timing of these single-cell processes is remarkably widely distributed with some cells undergoing their third division while others undergo their first. High cell-to-cell variability and technical noise pose challenges for interpreting popular dye-dilution experiments objectively. It remains an unresolved challenge to avoid under- or over-interpretation of such data when phenotyping gene-targeted mouse models or patient samples. Here we develop and characterize a computational methodology to parameterize a cell population model in the context of noisy dye-dilution data. To enable objective interpretation of model fits, our method estimates fit sensitivity and redundancy by stochastically sampling the solution landscape, calculating parameter sensitivities, and clustering to determine the maximum-likelihood solution ranges. Our methodology accounts for both technical and biological variability by using a cell fluorescence model as an adaptor during population model fitting, resulting in improved fit accuracy without the need for ad hoc objective functions. We have incorporated our methodology into an integrated phenotyping tool, FlowMax, and used it to analyze B cells from two NFκB knockout mice with distinct phenotypes; we not only confirm previously published findings at a fraction of the expended effort and cost, but reveal a novel phenotype of nfkb1/p105/50 in limiting the proliferative capacity of B cells following B-cell receptor stimulation. In addition to complementing experimental work, FlowMax is suitable for high throughput analysis of dye dilution studies within clinical and pharmacological screens with objective and quantitative conclusions.


Assuntos
Linfócitos B/citologia , Biologia Computacional/métodos , Citometria de Fluxo/métodos , Fluoresceínas/análise , Modelos Biológicos , Software , Succinimidas/análise , Algoritmos , Animais , Proliferação de Células , Células Cultivadas , Corantes Fluorescentes/análise , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B/fisiologia , Proteínas Proto-Oncogênicas c-rel/fisiologia , Coloração e Rotulagem , Fatores de Tempo
16.
Biochim Biophys Acta ; 1832(12): 1980-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23816567

RESUMO

S6K (ribosomal S6 kinase p70, p70S6K) activation requires phosphorylation at two stages. The first phosphorylation is independent of insulin stimulation and mediated by an unknown kinase. The second phosphorylation is mediated by mTOR in insulin dependent manner. In this study, we identified JNK1 (c-Jun N-terminal kinase 1) as a kinase in the first phosphorylation. S6K protein was phosphorylated by JNK1 at S411 and S424 in the carboxyl terminal autoinhibitory domain. The phosphorylation was observed in kinase assay with purified S6K as a substrate, and in cells after JNK1 activation by TNF-α or MEKK1 expression. The phosphorylation was detected in JNK2 null cells, but not in JNK1 null cells after TNF-α treatment. When JNK1 activation was inhibited by MKK7 knockdown, the phosphorylation was blocked in cells. The phosphorylation led to S6K protein degradation in NF-κB deficient cells. The degradation was blocked by inhibition of proteasome activity with MG132. In wide type cells, the phosphorylation did not promote S6K degradation when IKK2 (IKKß, IκB kinase beta) was activated. Instead, the phosphorylation allowed S6K activation by mTOR, which stabilizes S6K protein. In IKK2 null cells or cells treated by IKK2 inhibitor, the phosphorylation led to S6K degradation. These data suggest that S6K is phosphorylated by JNK1 and the phosphorylation makes S6K protein unstable in the absence of IKK2 activation. This study provides a mechanism for regulation of S6K protein stability.


Assuntos
Ativação Enzimática/efeitos dos fármacos , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Subunidade p50 de NF-kappa B/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Ubiquitina/metabolismo , Animais , Western Blotting , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Células HEK293 , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Imunoprecipitação , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Proteólise , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Transdução de Sinais , Ubiquitinação
17.
J Immunol ; 190(8): 3949-58, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23487427

RESUMO

Respiratory diseases are the most frequent chronic illnesses in babies and children. Although a vigorous innate immune system is critical for maintaining lung health, a balanced response is essential to minimize damaging inflammation. We investigated the functional and clinical impact of human genetic variants in the promoter of NFKBIA, which encodes IκBα, the major negative regulator of NF-κB. In this study, we quantified the functional impact of NFKBIA promoter polymorphisms (rs3138053, rs2233406, and rs2233409) on promoter-driven protein expression, allele-specific and total NFKBIA mRNA expression, IκBα protein expression, and TLR responsiveness; mapped innate immune regulatory networks active during respiratory syncytial virus infection, asthma, and bronchopulmonary dysplasia; and genotyped and analyzed independent cohorts of children with respiratory syncytial virus infection, asthma, and bronchopulmonary dysplasia. Genetic variants in the promoter of NFKBIA influenced NFKBIA gene expression, IκBα protein expression, and TLR-mediated inflammatory responses. Using a systems biology approach, we demonstrated that NFKBIA/IκBα is a central hub in transcriptional responses of prevalent childhood lung diseases, including respiratory syncytial virus infection, asthma, and bronchopulmonary dysplasia. Finally, by examining independent pediatric lung disease cohorts, we established that this immunologically relevant genetic variation in the promoter of NFKBIA is associated with differential susceptibility to severe bronchiolitis following infection with respiratory syncytial virus, airway hyperresponsiveness, and severe bronchopulmonary dysplasia. These data highlight the importance of negative innate immune regulators, such as NFKBIA, in pediatric lung disease and begin to unravel common aspects in the genetic predisposition to bronchopulmonary dysplasia, bronchiolitis, and childhood asthma.


Assuntos
Asma/imunologia , Bronquiolite/imunologia , Displasia Broncopulmonar/imunologia , Predisposição Genética para Doença , Variação Genética/imunologia , Subunidade p50 de NF-kappa B/genética , Animais , Asma/genética , Bronquiolite/genética , Bronquiolite/virologia , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/virologia , Células CHO , Criança , Pré-Escolar , Cricetinae , Feminino , Humanos , Lactente , Recém-Nascido , Subunidade p50 de NF-kappa B/fisiologia , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Vírus Sinciciais Respiratórios/imunologia
18.
Urol Int ; 91(2): 206-12, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23295440

RESUMO

OBJECTIVE: Activated nuclear factor-ĸB is associated with the pathogenesis of numerous malignancies, including renal cell carcinoma (RCC). This study aimed to clarify the influence of a common insertion/deletion polymorphism (-94 ins/del ATTG, rs28362491) in the NFKB1 promoter on RCC susceptibility. METHODS: We genotyped the NFKB1 -94 ins/del ATTG promoter polymorphism by the TaqMan method and assessed the association with RCC risk, clinicopathological parameters in a case-control study of 1,027 cases and 1,094 controls. RESULTS: The genotype frequencies were significantly different between RCC cases and controls (p = 0.046). Compared with individuals carrying the ins/del + del/del genotypes, those with the ins/ins genotype had an increased RCC risk [p = 0.036, adjusted odds ratio (OR) = 1.23, 95% confidence interval (CI) = 1.02-1.48], particularly in the subgroup of younger age (p = 0.005, adjusted OR = 1.42, 95% CI = 1.11-1.83) and never smokers (p = 0.013, adjusted OR = 1.34, 95% CI = 1.07-1.69). Furthermore, the polymorphism was significantly associated with the risk of developing localized stage RCC (p = 0.020, OR = 1.26, 95% CI = 1.04-1.53). CONCLUSIONS: The functional NFKB1 promoter polymorphism is associated with an increased risk of RCC.


Assuntos
Carcinoma de Células Renais/genética , Neoplasias Renais/genética , Subunidade p50 de NF-kappa B/genética , Polimorfismo Genético , Regiões Promotoras Genéticas , Adulto , Idoso , Consumo de Bebidas Alcoólicas/efeitos adversos , Índice de Massa Corporal , Carcinoma de Células Renais/diagnóstico , Estudos de Casos e Controles , Genótipo , Humanos , Mutação INDEL , Neoplasias Renais/diagnóstico , Pessoa de Meia-Idade , Subunidade p50 de NF-kappa B/fisiologia , Razão de Chances , Risco , Fumar/efeitos adversos
19.
Sleep Med ; 14(2): 165-71, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23266106

RESUMO

OBJECTIVES: Patients with obstructive sleep apnea have an impaired endothelium-dependent vasodilator response. The mechanisms underlying this impairment remain unclear. We tested the hypothesis that chronic intermittent hypoxia (CIH) impairs endothelium-dependent vasodilatation by NF-κB-mediated down-regulation of endothelial nitric oxide synthase (eNOS) expression. METHODS: Wild type (WT) mice and mice deficient in NF-κB p50 or TNF-α gene were exposed to sham or CIH. Aortic NF-κB activity and aortic expression of TNF-α were determined. Aortic and mesenteric artery levels of eNOS expression were examined and their correlation to endothelium-dependent vasodilator response in vitro and vasodepressor response in vivo were analyzed. RESULTS: WT mice exposed to CIH for five to eight weeks showed significantly reduced eNOS protein expression in aortas and mesenteric arteries, associated with significantly blunted vasodilator and vasodepressor responses to acetylcholine, but not to sodium nitroprusside. CIH activated NF-κB, which preceded TNF-α up-regulation and eNOS down-regulation. NF-κB p50 gene deletion blocked NF-κB activation, inhibited TNF-α expression, prevented eNOS down-regulation and reversed the impaired endothelium-dependent vasodepressor response induced by CIH. TNF-α knockout prevented CIH-induced eNOS down-regulation and restored the endothelium-dependent vasodepressor response. CONCLUSIONS: CIH exposure impairs endothelium-dependent vasodilator mechanism by stimulating NF-κB-mediated TNF-α generation, which in turn, down-regulates eNOS expression, resulting in an impaired endothelium-dependent vasodilatation.


Assuntos
Regulação para Baixo/fisiologia , Hipóxia/fisiopatologia , Subunidade p50 de NF-kappa B/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Apneia Obstrutiva do Sono/fisiopatologia , Acetilcolina/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Humanos , Hipóxia/genética , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B/genética , Óxido Nítrico Sintase Tipo III/genética , Nitroprussiato/farmacologia , Apneia Obstrutiva do Sono/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/fisiologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/genética , Vasodilatação/fisiologia , Vasodilatadores/farmacologia
20.
Carcinogenesis ; 33(10): 1843-53, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22581827

RESUMO

Transcription factors are direct effectors of altered signaling pathways in cancer and frequently determine clinical outcomes in cancer patients. To uncover new transcription factors that would determine clinical outcomes in breast cancer, we systematically analyzed gene expression data from breast cancer patients. Our results revealed that Forkhead box protein M1 (FOXM1) is the top-ranked survival-associated transcription factor in patients with triple-negative breast cancer. Surprisingly, silencing FOXM1 expression led breast cancer cells to become more sensitive to doxorubicin (Dox). We found that FOXM1-dependent resistance to Dox is mediated by regulating DNA repair genes. We further demonstrated that NFκB1 interacts with FOXM1 in the presence of Dox to protect breast cancer cells from DNA damage. Finally, silencing FOXM1 expression in breast cancer cells in a mouse xenograft model significantly sensitized the cells to Dox. Our systematic approaches identified an unexpected role of FOXM1 in Dox resistance by regulating DNA repair genes, and our findings provide mechanistic insights into how FOXM1 mediates resistance to Dox and evidence that FOXM1 may be a promising therapeutic target for sensitizing breast cancer cells to Dox.


Assuntos
Neoplasias da Mama/genética , Reparo do DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Fatores de Transcrição Forkhead/fisiologia , Fatores de Transcrição/fisiologia , Animais , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Subunidade p50 de NF-kappa B/fisiologia , Prognóstico , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA