Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34810256

RESUMO

Global inactivation of IκB kinase (IKK)-α results in defective lymph node (LN) formation and B cell maturation, and loss of IKK-α-dependent noncanonical NF-κB signaling in stromal organizer and hematopoietic cells is thought to underlie these distinct defects. We previously demonstrated that this pathway is also activated in vascular endothelial cells (ECs). To determine the physiologic function of EC-intrinsic IKK-α, we crossed IkkαF/F mice with Tie2-cre or Cdh5-cre mice to ablate IKK-α in ECs. Notably, the compound defects of global IKK-α inactivation were recapitulated in IkkαTie2 and IkkαCdh5 mice, as both lacked all LNs and mature follicular and marginal zone B cell numbers were markedly reduced. However, as Tie2-cre and Cdh5-cre are expressed in all ECs, including blood forming hemogenic ECs, IKK-α was also absent in hematopoietic cells (HC). To determine if loss of HC-intrinsic IKK-α affected LN development, we generated IkkαVav mice lacking IKK-α in only the hematopoietic compartment. While mature B cell numbers were significantly reduced in IkkαVav mice, LN formation was intact. As lymphatic vessels also arise during development from blood ECs, we generated IkkαLyve1 mice lacking IKK-α in lymphatic ECs (LECs) to determine if IKK-α in lymphatic vessels impacts LN development. Strikingly, while mature B cell numbers were normal, LNs were completely absent in IkkαLyve1 mice. Thus, our findings reveal that IKK-α in distinct EC-derived compartments is uniquely required to promote B cell homeostasis and LN development, and we establish that LEC-intrinsic IKK-α is absolutely essential for LN formation.


Assuntos
Linfócitos B/metabolismo , Quinase I-kappa B/fisiologia , Linfonodos/metabolismo , Animais , Linfócitos B/fisiologia , Linhagem Celular , Células Endoteliais/metabolismo , Feminino , Homeostase/fisiologia , Quinase I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Linfonodos/fisiologia , Tecido Linfoide/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Organogênese/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
3.
J Biol Chem ; 291(16): 8440-52, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26826124

RESUMO

T cell activation following antigen binding to the T cell receptor (TCR) involves the mobilization of intracellular Ca(2+) to activate the key transcription factors nuclear factor of activated T lymphocytes (NFAT) and NF-κB. The mechanism of NFAT activation by Ca(2+) has been determined. However, the role of Ca(2+) in controlling NF-κB signaling is poorly understood, and the source of Ca(2+) required for NF-κB activation is unknown. We demonstrate that TCR- but not TNF-induced NF-κB signaling upstream of IκB kinase activation absolutely requires the influx of extracellular Ca(2+) via STIM1-dependent Ca(2+) release-activated Ca(2+)/Orai channels. We further show that Ca(2+) influx controls phosphorylation of the NF-κB protein p65 on Ser-536 and that this posttranslational modification controls its nuclear localization and transcriptional activation. Notably, our data reveal that this role for Ca(2+) is entirely separate from its upstream control of IκBα degradation, thereby identifying a novel Ca(2+)-dependent distal step in TCR-induced NF-κB activation. Finally, we demonstrate that this control of distal signaling occurs via Ca(2+)-dependent PKCα-mediated phosphorylation of p65. Thus, we establish the source of Ca(2+) required for TCR-induced NF-κB activation and define a new distal Ca(2+)-dependent checkpoint in TCR-induced NF-κB signaling that has broad implications for the control of immune cell development and T cell functional specificity.


Assuntos
Canais de Cálcio/biossíntese , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Neoplasias/biossíntese , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Fator de Transcrição RelA/metabolismo , Ativação Transcricional/fisiologia , Canais de Cálcio/genética , Humanos , Células Jurkat , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Proteína ORAI1 , Fosforilação/fisiologia , Receptores de Antígenos de Linfócitos T/genética , Molécula 1 de Interação Estromal , Fator de Transcrição RelA/genética
4.
Proc Natl Acad Sci U S A ; 110(41): 16556-61, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-24062461

RESUMO

Activation of the nuclear transcription factor κB (NF-κB) regulates the expression of inflammatory genes crucially involved in the pathogenesis of inflammatory diseases. NF-κB governs the expression of adhesion molecules that play a pivotal role in leukocyte-endothelium interactions. We uncovered the crucial role of NF-κB activation within endothelial cells in models of immune-mediated diseases using a "sneaking ligand construct" (SLC) selectively inhibiting NF-κB in the activated endothelium. The recombinant SLC1 consists of three modules: (i) an E-selectin targeting domain, (ii) a Pseudomonas exotoxin A translocation domain, and (iii) a NF-κB Essential Modifier-binding effector domain interfering with NF-κB activation. The E-selectin-specific SLC1 inhibited NF-κB by interfering with endothelial IκB kinase 2 activity in vitro and in vivo. In murine experimental peritonitis, the application of SLC1 drastically reduced the extravasation of inflammatory cells. Furthermore, SLC1 treatment significantly ameliorated the disease course in murine models of rheumatoid arthritis. Our data establish that endothelial NF-κB activation is critically involved in the pathogenesis of arthritis and can be selectively inhibited in a cell type- and activation stage-dependent manner by the SLC approach. Moreover, our strategy is applicable to delineating other pathogenic signaling pathways in a cell type-specific manner and enables selective targeting of distinct cell populations to improve effectiveness and risk-benefit ratios of therapeutic interventions.


Assuntos
Artrite/tratamento farmacológico , Artrite/imunologia , Células Endoteliais/imunologia , Regulação da Expressão Gênica/imunologia , NF-kappa B/antagonistas & inibidores , Proteínas Recombinantes de Fusão/imunologia , Animais , Proteínas de Bactérias/metabolismo , Clonagem Molecular , Selectina E/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Células Endoteliais/efeitos dos fármacos , Escherichia coli , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/imunologia
5.
Biochem Biophys Res Commun ; 450(1): 341-6, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24942881

RESUMO

Non-canonical NF-κB signaling is controlled by the precise regulation of NF-κB inducing kinase (NIK) stability. NIK is constitutively ubiquitylated by cellular inhibitor of apoptosis (cIAP) proteins 1 and 2, leading to its complete proteasomal degradation in resting cells. Following stimulation, cIAP-mediated ubiquitylation of NIK ceases and NIK is stabilized, allowing for inhibitor of κB kinase (IKK)α activation and non-canonical NF-κB signaling. Non-canonical NF-κB signaling is terminated by feedback phosphorylation of NIK by IKKα that promotes NIK degradation; however, the mechanism of active NIK protein turnover remains unknown. To address this question, we established a strategy to precisely distinguish between basal degradation of newly synthesized endogenous NIK and induced active NIK in stimulated cells. Using this approach, we found that IKKα-mediated degradation of signal-induced activated NIK occurs through the proteasome. To determine whether cIAP1 or cIAP2 play a role in active NIK turnover, we utilized a Smac mimetic (GT13072), which promotes degradation of these E3 ubiquitin ligases. As expected, GT13072 stabilized NIK in resting cells. However, loss of the cIAPs did not inhibit proteasome-dependent turnover of signal-induced NIK showing that unlike the basal regulatory mechanism, active NIK turnover is independent of cIAP1 and cIAP2. Our results therefore establish that the negative feedback control of IKKα-mediated NIK turnover occurs via a novel proteasome-dependent and cIAP-independent mechanism.


Assuntos
Retroalimentação Fisiológica/fisiologia , Regulação da Expressão Gênica/fisiologia , Quinase I-kappa B/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células 3T3 , Animais , Ativação Enzimática , Células HeLa , Humanos , Camundongos , Quinase Induzida por NF-kappaB
6.
Nature ; 456(7223): 819-23, 2008 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-18849970

RESUMO

DNA double-strand breaks are generated by genotoxic agents and by cellular endonucleases as intermediates of several important physiological processes. The cellular response to genotoxic DNA breaks includes the activation of transcriptional programs known primarily to regulate cell-cycle checkpoints and cell survival. DNA double-strand breaks are generated in all developing lymphocytes during the assembly of antigen receptor genes, a process that is essential for normal lymphocyte development. Here we show that in murine lymphocytes these physiological DNA breaks activate a broad transcriptional program. This program transcends the canonical DNA double-strand break response and includes many genes that regulate diverse cellular processes important for lymphocyte development. Moreover, the expression of several of these genes is regulated similarly in response to genotoxic DNA damage. Thus, physiological DNA double-strand breaks provide cues that can regulate cell-type-specific processes not directly involved in maintaining the integrity of the genome, and genotoxic DNA breaks could disrupt normal cellular functions by corrupting these processes.


Assuntos
Linfócitos B/metabolismo , Quebras de DNA de Cadeia Dupla , Regulação da Expressão Gênica no Desenvolvimento/genética , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Linfócitos B/efeitos dos fármacos , Proteínas de Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proteínas de Ligação a DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Knockout , Camundongos SCID , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Supressoras de Tumor/efeitos dos fármacos
7.
bioRxiv ; 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38405871

RESUMO

X Chromosome Inactivation (XCI) is a female-specific process which balances X-linked gene dosage between sexes. Unstimulated T cells lack cytological enrichment of Xist RNA and heterochromatic modifications on the inactive X chromosome (Xi), and these modifications become enriched at the Xi after cell stimulation. Here, we examined allele-specific gene expression and the epigenomic profiles of the Xi following T cell stimulation. We found that the Xi in unstimulated T cells is largely dosage compensated and is enriched with the repressive H3K27me3 modification, but not the H2AK119-ubiquitin (Ub) mark, even at promoters of XCI escape genes. Upon CD3/CD28-mediated T cell stimulation, the Xi accumulates H2AK119-Ub and H3K27me3 across the Xi. Next, we examined the T cell signaling pathways responsible for Xist RNA localization to the Xi and found that T cell receptor (TCR) engagement, specifically NF-κB signaling downstream of TCR, is required. Disruption of NF-κB signaling, using inhibitors or genetic deletions, in mice and patients with immunodeficiencies prevents Xist/XIST RNA accumulation at the Xi and alters expression of some X-linked genes. Our findings reveal a novel connection between NF-κB signaling pathways which impact XCI maintenance in female T cells.

8.
Nature ; 446(7135): 552-6, 2007 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-17322906

RESUMO

Intestinal epithelial cells (IECs) provide a primary physical barrier against commensal and pathogenic microorganisms in the gastrointestinal (GI) tract, but the influence of IECs on the development and regulation of immunity to infection is unknown. Here we show that IEC-intrinsic IkappaB kinase (IKK)-beta-dependent gene expression is a critical regulator of responses of dendritic cells and CD4+ T cells in the GI tract. Mice with an IEC-specific deletion of IKK-beta show a reduced expression of the epithelial-cell-restricted cytokine thymic stromal lymphopoietin in the intestine and, after infection with the gut-dwelling parasite Trichuris, fail to develop a pathogen-specific CD4+ T helper type 2 (T(H)2) response and are unable to eradicate infection. Further, these animals show exacerbated production of dendritic-cell-derived interleukin-12/23p40 and tumour necrosis factor-alpha, increased levels of CD4+ T-cell-derived interferon-gamma and interleukin-17, and develop severe intestinal inflammation. Blockade of proinflammatory cytokines during Trichuris infection ablates the requirement for IKK-beta in IECs to promote CD4+ T(H)2 cell-dependent immunity, identifying an essential function for IECs in tissue-specific conditioning of dendritic cells and limiting type 1 cytokine production in the GI tract. These results indicate that the balance of IKK-beta-dependent gene expression in the intestinal epithelium is crucial in intestinal immune homeostasis by promoting mucosal immunity and limiting chronic inflammation.


Assuntos
Células Epiteliais/enzimologia , Regulação Enzimológica da Expressão Gênica , Homeostase , Quinase I-kappa B/metabolismo , Intestinos/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Citocinas/deficiência , Citocinas/imunologia , Células Dendríticas/imunologia , Células Epiteliais/metabolismo , Quinase I-kappa B/genética , Imunidade nas Mucosas/imunologia , Interferon gama/imunologia , Interleucina-17/imunologia , Intestinos/citologia , Intestinos/parasitologia , Camundongos , NF-kappa B/metabolismo , Trichuris/imunologia , Trichuris/fisiologia , Linfopoietina do Estroma do Timo
9.
J Immunol ; 185(5): 2665-9, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20693425

RESUMO

The signaling and adaptor protein Homer3 plays a role in controlling immune homeostasis and self-reactivity. Homer3 is recruited to the immune synapse (IS) following TCR ligation, although the mechanisms regulating this subcellular localization are unknown. We show that Homer3 specifically associates with a novel ubiquitin-like domain in the IkappaB kinase (IKK) beta subunit of the IKK complex. Homer3 associates with IKKbeta in T cells and colocalizes with the IKK complex at the IS. However, Homer3 is not required for IKK activation, as NF-kappaB signaling is intact in Homer3-deficient T cells. Instead, the IKK complex recruits Homer3 to the IS following TCR engagement, and we present evidence that this association regulates actin dynamics in T cells. These findings identify a novel interaction between two major signaling proteins and reveal an unexpected NF-kappaB-independent function for the IKK complex in regulating the subcellular localization of Homer3.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas I-kappa B/metabolismo , Proteínas de Transporte/fisiologia , Linhagem Celular , Células HeLa , Proteínas de Arcabouço Homer , Humanos , Proteínas I-kappa B/fisiologia , Sinapses Imunológicas/enzimologia , Sinapses Imunológicas/imunologia , Células Jurkat , Estrutura Terciária de Proteína , Receptores de Antígenos de Linfócitos T/fisiologia , Transdução de Sinais/imunologia , Frações Subcelulares/imunologia , Frações Subcelulares/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo
10.
J Biol Chem ; 285(49): 38069-77, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20923761

RESUMO

Ligation of the lymphotoxin-ß receptor (LTßR) by LIGHT (lymphotoxin-related inducible ligand that competes for glycoprotein D binding to herpes virus entry mediator on T cells (TNFSF14)) activates the noncanonical (NC) NF-κB (nuclear factor-κB) pathway and up-regulates CXCL12 gene expression by human umbilical vein endothelial cells (HUVEC). In contrast, TNF only activates classical NF-κB signaling and does not up-regulate CXCL12. To determine whether cross-talk between the classical and NC pathways affects CXCL12 expression, we investigated the effects of TNF on LIGHT signaling in HUVEC. We show here that TNF inhibits both basal and LIGHT-induced CXCL12 expression. Negative regulation by TNF requires the classical NF-κB pathway as inhibition of basal and induced CXCL12 was reversed in HUVEC-expressing dominant negative IκB (inhibitor of NF-κB) kinase (IKK)ß (IKKß(K44M)). TNF did not inhibit the NC NF-κB pathway activation as LIGHT-induced p100 processing to p52 was intact; however, TNF either alone or together with LIGHT up-regulated p100 and RelB expression and induced the nuclear localization of p100-RelB complexes. Enhanced p100 and RelB expression was inhibited by IKKß(K44M), which led us to question whether the IκB function of elevated p100 mediates the inhibition of CXCL12 expression by TNF. We retrovirally transduced HUVEC to express p100 at a level similar to that up-regulated by TNF; however, basal and LIGHT-induced CXCL12 expression was normal in the transduced cells. In contrast, ectopic RelB expression recapitulated the effects of TNF on NC signaling and inhibited basal and LIGHT-induced CXCL12 expression by HUVEC. Our findings therefore demonstrate that TNF-induced classical NF-κB signaling up-regulates RelB expression that inhibits both basal and NC NF-κB-dependent CXCL12 expression.


Assuntos
Núcleo Celular/metabolismo , Quimiocina CXCL12/biossíntese , Células Endoteliais/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição RelB/metabolismo , Veias Umbilicais/metabolismo , Regulação para Cima/fisiologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/fisiologia , Linhagem Celular , Núcleo Celular/genética , Quimiocina CXCL12/genética , Células Endoteliais/citologia , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Mutação de Sentido Incorreto , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelB/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Veias Umbilicais/citologia , Regulação para Cima/efeitos dos fármacos
11.
Nat Med ; 10(6): 617-24, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15156202

RESUMO

Bone destruction is a pathological hallmark of several chronic inflammatory diseases, including rheumatoid arthritis and periodontitis. Inflammation-induced bone loss of this sort results from elevated numbers of bone-resorbing osteoclasts. Gene targeting studies have shown that the transcription factor nuclear factor-kappa B (NF-kappa B) has a crucial role in osteoclast differentiation, and blocking NF-kappa B is a potential strategy for preventing inflammatory bone resorption. We tested this approach using a cell-permeable peptide inhibitor of the I kappa B-kinase complex, a crucial component of signal transduction pathways to NF-kappa B. The peptide inhibited RANKL-stimulated NF-kappa B activation and osteoclastogenesis both in vitro and in vivo. In addition, this peptide significantly reduced the severity of collagen-induced arthritis in mice by reducing levels of tumor necrosis factor-alpha and interleukin-1 beta, abrogating joint swelling and reducing destruction of bone and cartilage. Therefore, selective inhibition of NF-kappa B activation offers an effective therapeutic approach for inhibiting chronic inflammatory diseases involving bone resorption.


Assuntos
Reabsorção Óssea/metabolismo , Osso e Ossos/metabolismo , Proteínas I-kappa B/antagonistas & inibidores , Inflamação/metabolismo , NF-kappa B/antagonistas & inibidores , Osteoclastos/fisiologia , Peptídeos/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Reabsorção Óssea/imunologia , Osso e Ossos/citologia , Osso e Ossos/patologia , Proteínas de Transporte/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Inflamação/imunologia , Interleucina-1/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , NF-kappa B/metabolismo , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
12.
Methods Mol Biol ; 2366: 145-164, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34236637

RESUMO

The central role of calcium (Ca2+) signaling in lymphocyte development and acquisition of functional immunity and tolerance is well established. Ca2+ signals are initiated upon antigen binding to cognate receptors on lymphocytes that trigger store operated Ca2+ entry (SOCE). The underlying mechanism of SOCE in lymphocytes involves TCR and BCR mediated activation of Stromal Interaction Molecule 1 and 2 (STIM1/2) embedded in the ER membrane. Once activated, STIM proteins oligomerize and re-localize to ER domains juxtaposed to the plasma membrane where they activate Orai channels to allow Ca2+ to enter the cell across the plasma membrane. Importantly, STIM/Orai-dependent Ca2+ signals guide antigen induced lymphocyte development and function principally by regulating the activity of transcription factors.The most widely studied of these transcription factors is the Nuclear Factor of Activated T cells (NFAT). NFAT is expressed ubiquitously and the mechanism by which Ca2+ regulates NFAT activation and signaling is well known. By contrast, a mechanistic understanding of how Ca2+ signals also shape the activation and specificity of NF-κB to control the expression of pro-inflammatory genes has lagged. Here we discuss the methodology used to investigate Ca2+ dependent mechanisms of NF-κB activation in lymphocytes. Our approach focuses on three main areas of signal transduction and signaling: (1) antigen receptor engagement and Ca2+ dependent initiation of NF-kB signaling, (2) Ca2+ dependent induction of NF-κB heterodimer activation and nuclear localization, and (3) and how Ca2+ regulates NF-κB dependent expression of target genes and proteins.


Assuntos
Linfócitos B , Linfócitos B/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/genética , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Molécula 2 de Interação Estromal
13.
J Biol Chem ; 284(40): 27596-608, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19666475

RESUMO

Proinflammatory NF-kappaB activation requires the IkappaB (inhibitor of NF-kappaB) kinase (IKK) complex that contains two catalytic subunits named IKKalpha and IKKbeta and a regulatory subunit named NF-kappaB essential modulator (NEMO). NEMO and IKKbeta are essential for tumor necrosis factor (TNF)-induced NF-kappaB activation, and we recently demonstrated that NEMO and IKKalpha are sufficient for interleukin (IL)-1-induced signaling. IKKalpha and IKKbeta both contain a functional NEMO-binding domain (NBD); however, the role of NEMO association with each kinase in NF-kappaB signaling and IKK complex formation remains unclear. To address this question, we stably reconstituted IKKalpha(-/-) and IKKbeta(-/-) murine embryonic fibroblasts (MEFs) with wild-type (WT) or NBD-deficient (DeltaNBD) versions of IKKalpha and IKKbeta, respectively. TNF-induced classical NF-kappaB activation in IKKbeta(-/-) MEFs was rescued by IKKbeta(WT) but not IKKbeta(DeltaNBD), whereas neither IKKbeta(WT) nor IKKbeta(DeltaNBD) affected IL-1-induced NF-kappaB signaling. As previously described, classical NF-kappaB transcriptional activity was absent in IKKalpha(-/-) cells. Reconstitution with either IKKalpha(WT) or IKKalpha(DeltaNBD) rescued both IL-1 and TNF-induced transcription, demonstrating that NEMO association is not required for IKKalpha-dependent regulation of NF-kappaB-dependent transcription. Stably expressed IKKalpha(WT) or IKKbeta(WT) associated with endogenous IKKs and NEMO in IKKalpha(-/-) or IKKbeta(-/-) MEFs, respectively, resulting in formation of the heterotrimeric IKKalpha-IKKbeta-NEMO complex. In contrast, although the IKKalpha(DeltaNBD) and IKKbeta(DeltaNBD) mutants associated with endogenous IKKs containing an NBD, these dimeric endogenous IKK-IKK(DeltaNBD) complexes did not associate with NEMO. These findings therefore demonstrate that formation of the heterotrimeric IKKalpha-IKKbeta-NEMO holocomplex absolutely requires two intact NEMO-binding domains.


Assuntos
Quinase I-kappa B/química , Quinase I-kappa B/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , NF-kappa B/metabolismo , Animais , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Quinase I-kappa B/genética , Interleucina-1/farmacologia , Camundongos , Estrutura Terciária de Proteína , Deleção de Sequência , Transdução de Sinais , Fator de Necrose Tumoral alfa/farmacologia
14.
Mol Cell Biol ; 27(21): 7582-93, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17724077

RESUMO

Receptor-mediated signaling is commonly associated with multiple functions, including the production of reactive oxygen species. However, whether mitochondrion-derived superoxide (mROS) contributes directly to physiological signaling is controversial. Here we demonstrate a previously unknown mechanism in which physiologic Ca(2+)-evoked mROS production plays a pivotal role in endothelial cell (EC) activation and leukocyte firm adhesion. G protein-coupled receptor (GPCR) and tyrosine kinase-mediated inositol 1,4,5-trisphosphate-dependent mitochondrial Ca(2+) uptake resulted in NADPH oxidase-independent mROS production. However, GPCR-linked mROS production did not alter mitochondrial function or trigger cell death but rather contributed to activation of NF-kappaB and leukocyte adhesion via the EC induction of intercellular adhesion molecule 1. Dismutation of mROS by manganese superoxide dismutase overexpression and a cell-permeative superoxide dismutase mimetic ablated NF-kappaB transcriptional activity and facilitated leukocyte detachment from the endothelium under simulated circulation following GPCR- but not cytokine-induced activation. These results demonstrate that mROS is the downstream effector molecule that translates receptor-mediated Ca(2+) signals into proinflammatory signaling and leukocyte/EC firm adhesion.


Assuntos
Cálcio/metabolismo , Células Endoteliais/citologia , Leucócitos/citologia , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Galinhas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/ultraestrutura , Humanos , Inflamação , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/efeitos dos fármacos , Camundongos , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , NADPH Oxidases/metabolismo , NF-kappa B/metabolismo , Receptor PAR-1/metabolismo , Superóxidos/metabolismo , Trombina/farmacologia
15.
Sci Signal ; 13(625)2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32234960

RESUMO

Clinical response to chimeric antigen receptor (CAR) T cell therapy is correlated with CAR T cell persistence, especially for CAR T cells that target CD19+ hematologic malignancies. 4-1BB-costimulated CAR (BBζ) T cells exhibit longer persistence after adoptive transfer than do CD28-costimulated CAR (28ζ) T cells. 4-1BB signaling improves T cell persistence even in the context of 28ζ CAR activation, which indicates distinct prosurvival signals mediated by the 4-1BB cytoplasmic domain. To specifically study signal transduction by CARs, we developed a cell-free, ligand-based activation and ex vivo culture system for CD19-specific CAR T cells. We observed greater ex vivo survival and subsequent expansion of BBζ CAR T cells when compared to 28ζ CAR T cells. We showed that only BBζ CARs activated noncanonical nuclear factor κB (ncNF-κB) signaling in T cells basally and that the anti-CD19 BBζ CAR further enhanced ncNF-κB signaling after ligand engagement. Reducing ncNF-κB signaling reduced the expansion and survival of anti-CD19 BBζ T cells and was associated with a substantial increase in the abundance of the most pro-apoptotic isoforms of Bim. Although our findings do not exclude the importance of other signaling differences between BBζ and 28ζ CARs, they demonstrate the necessary and nonredundant role of ncNF-κB signaling in promoting the survival of BBζ CAR T cells, which likely underlies the engraftment persistence observed with this CAR design.


Assuntos
NF-kappa B/imunologia , Receptores de Antígenos Quiméricos/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Linhagem Celular , Humanos , Receptores de Antígenos Quiméricos/genética , Transdução de Sinais/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética
16.
Cell Rep ; 31(2): 107474, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32294437

RESUMO

B cell receptor (BCR) engagement induces naive B cells to differentiate and perform critical immune-regulatory functions. Acquisition of functional specificity requires that a cell survive, enter the cell cycle, and proliferate. We establish that quantitatively distinct Ca2+ signals triggered by variations in the extent of BCR engagement dynamically regulate these transitions by controlling nuclear factor κB (NF-κB), NFAT, and mTORC1 activity. Weak BCR engagement induces apoptosis by failing to activate NF-κB-driven anti-apoptotic gene expression. Stronger signals that trigger more robust Ca2+ signals promote NF-κB-dependent survival and NFAT-, mTORC1-, and c-Myc-dependent cell-cycle entry and proliferation. Finally, we establish that CD40 or TLR9 costimulation circumvents these Ca2+-regulated checkpoints of B cell activation and proliferation. As altered BCR signaling is linked to autoimmunity and B cell malignancies, these results have important implications for understanding the pathogenesis of aberrant B cell activation and differentiation and therapeutic approaches to target these responses.


Assuntos
Cálcio/metabolismo , Células Precursoras de Linfócitos B/metabolismo , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Apoptose/imunologia , Linfócitos B/imunologia , Ciclo Celular/imunologia , Diferenciação Celular/imunologia , Proliferação de Células/fisiologia , Sobrevivência Celular/imunologia , Ativação Linfocitária/imunologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/imunologia , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/imunologia , Fatores de Transcrição NFATC/metabolismo , Células Precursoras de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais/imunologia
17.
Methods Mol Biol ; 512: 209-32, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19347279

RESUMO

Nuclear factor kappa B (NF-kappaB) is an inducible transcription factor that regulates the expression of many genes involved in normal immune and inflammatory responses. NF-kappaB activation is normally a rapid and transient response to pro-inflammatory stimuli however dysregulated constitutively active NF-kappaB signaling leads to chronic inflammation and provides a cell survival signal in many types of cancer. NF-kappaB signaling is therefore an important target for the development of novel anti-inflammatory or anti-cancer drugs. We previously identified and characterized a cell-permeable peptide that blocks NF-kappaB signaling by disrupting the critical upstream IkappaB kinase (IKK) complex. We describe in this chapter three separate methods to determine the effects of this NEMO-binding domain (NBD) peptide on pro-inflammatory NF-kappaB signaling in response to tumor necrosis factor (TNF).


Assuntos
Quinase I-kappa B/farmacologia , NF-kappa B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Células HeLa/efeitos dos fármacos , Células HeLa/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Immunoblotting , Luciferases/metabolismo , Estrutura Terciária de Proteína , Fator de Necrose Tumoral alfa/farmacologia
18.
J Allergy Clin Immunol ; 122(6): 1169-1177.e16, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18851874

RESUMO

BACKGROUND: Human hypomorphic nuclear factor-kappaB essential modulator (NEMO) mutations cause diverse clinical and immunologic phenotypes, but understanding of their scope and mechanistic links to immune function and genotype is incomplete. OBJECTIVE: We created and analyzed a database of hypomorphic NEMO mutations to determine the spectrum of phenotypes and their associated genotypes and sought to establish a standardized NEMO reconstitution system to obtain mechanistic insights. METHODS: Phenotypes of 72 individuals with NEMO mutations were compiled. NEMO L153R and C417R were investigated further in a reconstitution system. TNF-alpha or Toll-like receptor (TLR)-5 signals were evaluated for nuclear factor-kappaB activation, programmed cell death, and A20 gene expression. RESULTS: Thirty-two different mutations were identified; 53% affect the zinc finger domain. Seventy-seven percent were associated with ectodermal dysplasia, 86% with serious pyogenic infection, 39% with mycobacterial infection, 19% with serious viral infection, and 23% with inflammatory diseases. Thirty-six percent of individuals died at a mean age of 6.4 years. CD40, IL-1, TNF-alpha, TLR, and T-cell receptor signals were impaired in 15 of 16 (94%), 6 of 7 (86%), 9 of 11 (82%), 9 of 14 (64%), and 7 of 18 (39%), respectively. Hypomorphism-reconstituted NEMO-deficient cells demonstrated partial restoration of NEMO functions. Although both L153R and C417R impaired TLR and TNF-alpha-induced NF-kappaB activation, L153R also increased TNF-alpha-induced programmed cell death with decreased A20 expression. CONCLUSION: Distinct NEMO hypomorphs define specific disease and genetic characteristics. A reconstitution system can identify attributes of hypomorphisms independent of an individual's genetic background. Apoptosis susceptibility in L153R reconstituted cells defines a specific phenotype of this mutation that likely contributes to the excessive inflammation with which it is clinically associated.


Assuntos
Bases de Dados Genéticas , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/metabolismo , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Apoptose/genética , Antígenos CD40/metabolismo , Proteínas de Ligação a DNA , Regulação da Expressão Gênica/genética , Teste de Complementação Genética/métodos , Genótipo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Células Jurkat , NF-kappa B/metabolismo , Proteínas Nucleares/biossíntese , Fenótipo , Estrutura Terciária de Proteína/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/genética , Receptor 5 Toll-Like/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Fator de Necrose Tumoral alfa/metabolismo
19.
Stroke ; 39(9): 2578-86, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18420947

RESUMO

BACKGROUND AND PURPOSE: NF-kappaB is a transcription factor that regulates inflammatory and apoptotic pathways. We described previously that intraperitoneal administration of the NF-kappaB inhibitor TAT-NBD at 0 and 3 hours after neonatal hypoxia-ischemia (HI) markedly reduced brain damage. We hypothesize that timing and duration of NF-kappaB inhibition will be a major factor in determining outcome. METHODS: HI was induced in P7 rats by unilateral carotid artery occlusion and hypoxia. In vivo TAT-NBD effects were determined on cerebral damage, NF-kappaB activity, cytokine expression, and pro- and antiapoptotic molecules. In vitro effects of TAT-NBD were determined using primary neurons and cell lines. RESULTS: HI induced 2 peaks of cerebral NF-kappaB activity at 3 to 6 and 24 hours after HI. Neuroprotective 0/3-hour TAT-NBD treatment only inhibited early NF-kappaB activity. However, inhibition of both early and late NF-kappaB-activity by 0/6/12-hour TAT-NBD or only late NF-kappaB activity by 18/21-hour TAT-NBD aggravated damage. 0/6/12-hour TAT-NBD did not prevent HI-induced upregulation of cytokines at 24 hours after HI. Protective 0/3-hour TAT-NBD treatment prevented nuclear accumulation of p53 at 24 hours after HI. Nuclear p53 was not reduced after 0/6/12-hour TAT-NBD. Prolonged TAT-NBD increased the proapoptotic factor PUMA and reduced the antiapoptotic factors Bcl-2 and Bcl-xL. Also in neuronal cultures prolonged TAT-NBD exposure overruled protective short-term TAT-NBD treatment. CONCLUSIONS: Early NF-kappaB activation contributes to neonatal HI brain damage. Late NF-kappaB provides endogenous neuroprotection and upregulates antiapoptotic molecules. Inhibition of early NF-kappaB activity is neuroprotective only when late NF-kappaB activity is maintained. Moreover, cerebral cytokine production can occur independently of NF-kappaB.


Assuntos
Encéfalo/metabolismo , Infarto Cerebral/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , NF-kappa B/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/metabolismo , Encéfalo/fisiopatologia , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/fisiopatologia , Citocinas/metabolismo , Citoproteção/efeitos dos fármacos , Citoproteção/fisiologia , Feminino , Humanos , Hipóxia-Isquemia Encefálica/fisiopatologia , Masculino , NF-kappa B/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo
20.
Stroke ; 39(7): 2129-37, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18420952

RESUMO

BACKGROUND AND PURPOSE: Interactions between excitotoxic, inflammatory, and apoptotic pathways determine outcome in hypoxic-ischemic brain damage. The transcription factor NF-kappaB has been suggested to enhance brain damage via stimulation of cytokine production. There is also evidence that NF-kappaB activity is required for neuronal survival. We used the NF-kappaB inhibitor NBD, coupled to TAT to facilitate cerebral uptake, to determine the neuroprotective capacity of NF-kappaB inhibition in neonatal hypoxia-ischemia (HI) and to identify its contribution to cerebral inflammation and damage. METHODS: Brain damage was induced in neonatal rats by unilateral carotid artery occlusion and hypoxia and analyzed immunohistochemically; NF-kappaB activity was analyzed by EMSA. We analyzed cytokine mRNA levels and activation of apoptotic pathways by Western blotting. In vitro effects of TAT-NBD were determined in a neuronal cell line. RESULTS: Inhibition of cerebral NF-kappaB activity by TAT-NBD had a significant neuroprotective effect; brain damage was reduced by more than 80% with a therapeutic window of at least 6 hours. In contrast to earlier suggestions, the protective effect of TAT-NBD did not involve suppression of early cytokine upregulation after HI. Moreover, NF-kappaB inhibition prevented HI-induced upregulation and nuclear as well as mitochondrial accumulation of p53, prevented mitochondrial cytochrome-c release and activation of caspase-3. Finally, TAT-NBD could directly increase neuronal survival because TAT-NBD was sufficient to inhibit death in a neuronal cell line. A nonactive mutant peptide did not have any effect. CONCLUSIONS: Inhibition of NF-kappaB has strong neuroprotective effects that involve downregulation of apoptotic molecules but are independent of inhibition of cytokine production.


Assuntos
Apoptose , Citocinas/biossíntese , Hipóxia-Isquemia Encefálica/patologia , NF-kappa B/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Encéfalo/patologia , Inflamação , Cinética , Modelos Biológicos , NF-kappa B/metabolismo , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar , Transdução de Sinais , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA