Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 41(21): e110372, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36124865

RESUMO

In a genome-wide screening for components of the dsDNA-break-induced IKK-NF-κB pathway, we identified scores of regulators, including tumor susceptibility gene TSG101. TSG101 is essential for DNA damage-induced formation of cellular poly(ADP-ribose) (PAR). TSG101 binds to PARP1 and is required for PARP1 activation. This function of TSG101 is independent of its role in the ESCRT-I endosomal sorting complex. In the absence of TSG101, the PAR-dependent formation of a nuclear PARP1-IKKγ signalosome, which triggers IKK activation, is impaired. According to its requirement for PARP1 and NF-κB activation, TSG101-deficient cells are defective in DNA repair and apoptosis protection. Loss of TSG101 results in PARP1 trapping at damage sites and mimics the effect of pharmacological PARP inhibition. We also show that the loss of TSG101 in connection with inactivated tumor suppressors BRCA1/2 in breast cancer cells is lethal. Our results imply TSG101 as a therapeutic target to achieve synthetic lethality in cancer treatment.


Assuntos
NF-kappa B , Poli ADP Ribosilação , NF-kappa B/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Dano ao DNA , Reparo do DNA , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo
2.
EMBO J ; 40(6): e104296, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33459422

RESUMO

The IκB kinase (IKK)-NF-κB pathway is activated as part of the DNA damage response and controls both inflammation and resistance to apoptosis. How these distinct functions are achieved remained unknown. We demonstrate here that DNA double-strand breaks elicit two subsequent phases of NF-κB activation in vivo and in vitro, which are mechanistically and functionally distinct. RNA-sequencing reveals that the first-phase controls anti-apoptotic gene expression, while the second drives expression of senescence-associated secretory phenotype (SASP) genes. The rapidly activated first phase is driven by the ATM-PARP1-TRAF6-IKK cascade, which triggers proteasomal destruction of inhibitory IκBα, and is terminated through IκBα re-expression from the NFKBIA gene. The second phase, which is activated days later in senescent cells, is on the other hand independent of IKK and the proteasome. An altered phosphorylation status of NF-κB family member p65/RelA, in part mediated by GSK3ß, results in transcriptional silencing of NFKBIA and IKK-independent, constitutive activation of NF-κB in senescence. Collectively, our study reveals a novel physiological mechanism of NF-κB activation with important implications for genotoxic cancer treatment.


Assuntos
Senescência Celular/fisiologia , Quinase I-kappa B/metabolismo , Inibidor de NF-kappaB alfa/biossíntese , Fator de Transcrição RelA/metabolismo , Transcrição Gênica/genética , Animais , Apoptose/genética , Linhagem Celular , Proliferação de Células/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Feminino , Inativação Gênica/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Inibidor de NF-kappaB alfa/genética , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo
3.
Development ; 148(21)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34751748

RESUMO

Although the role of the transcription factor NF-κB in intestinal inflammation and tumor formation has been investigated extensively, a physiological function of NF-κB in sustaining intestinal epithelial homeostasis beyond inflammation has not been demonstrated. Using NF-κB reporter mice, we detected strong NF-κB activity in Paneth cells, in '+4/+5' secretory progenitors and in scattered Lgr5+ crypt base columnar stem cells of small intestinal (SI) crypts. To examine NF-κB functions in SI epithelial self-renewal, mice or SI crypt organoids ('mini-guts') with ubiquitously suppressed NF-κB activity were used. We show that NF-κB activity is dispensable for maintaining SI epithelial proliferation, but is essential for ex vivo organoid growth. Furthermore, we demonstrate a dramatic reduction of Paneth cells in the absence of NF-κB activity, concomitant with a significant increase in goblet cells and immature intermediate cells. This indicates that NF-κB is required for proper Paneth versus goblet cell differentiation and for SI epithelial homeostasis, which occurs via regulation of Wnt signaling and Sox9 expression downstream of NF-κB. The current study thus presents evidence for an important role for NF-κB in intestinal epithelial self-renewal.


Assuntos
Células Caliciformes/citologia , Intestino Delgado/citologia , NF-kappa B/metabolismo , Celulas de Paneth/citologia , Animais , Diferenciação Celular , Autorrenovação Celular , Células Caliciformes/metabolismo , Homeostase , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Camundongos , NF-kappa B/genética , Organoides/citologia , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Celulas de Paneth/metabolismo , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt
4.
EMBO J ; 37(24)2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30467221

RESUMO

The IκB kinase (IKK) is considered to control gene expression primarily through activation of the transcription factor NF-κB. However, we show here that IKK additionally regulates gene expression on post-transcriptional level. IKK interacted with several mRNA-binding proteins, including a Processing (P) body scaffold protein, termed enhancer of decapping 4 (EDC4). IKK bound to and phosphorylated EDC4 in a stimulus-sensitive manner, leading to co-recruitment of P body components, mRNA decapping proteins 1a and 2 (DCP1a and DCP2) and to an increase in P body numbers. Using RNA sequencing, we identified scores of transcripts whose stability was regulated via the IKK-EDC4 axis. Strikingly, in the absence of stimulus, IKK-EDC4 promoted destabilization of pro-inflammatory cytokines and regulators of apoptosis. Our findings expand the reach of IKK beyond its canonical role as a regulator of transcription.


Assuntos
Quinase I-kappa B/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Endorribonucleases/genética , Endorribonucleases/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Quinase I-kappa B/genética , Complexos Multiproteicos/genética , Proteínas/genética , RNA Mensageiro/genética , Transativadores/genética , Transativadores/metabolismo
5.
Blood ; 133(13): 1489-1494, 2019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30696620

RESUMO

Persistent NF-κB activation is a hallmark of the malignant Hodgkin/Reed-Sternberg (HRS) cells in classical Hodgkin lymphoma (cHL). Genomic lesions, Epstein-Barr virus infection, soluble factors, and tumor-microenvironment interactions contribute to this activation. Here, in an unbiased approach to identify the cHL cell-secreted key factors for NF-κB activation, we have dissected the secretome of cultured cHL cells by chromatography and subsequent mass spectrometry. We identified lymphotoxin-α (LTA) as the causative factor for autocrine and paracrine activation of canonical and noncanonical NF-κB in cHL cell lines. In addition to inducing NF-κB, LTA promotes JAK2/STAT6 signaling. LTA and its receptor TNFRSF14 are transcriptionally activated by noncanonical NF-κB, creating a continuous feedback loop. Furthermore, LTA shapes the expression of cytokines, receptors, immune checkpoint ligands and adhesion molecules, including CSF2, CD40, PD-L1/PD-L2, and VCAM1. Comparison with single-cell gene-activity profiles of human hematopoietic cells showed that LTA induces genes restricted to the lymphoid lineage, as well as those largely restricted to the myeloid lineage. Thus, LTA sustains autocrine NF-κB activation, impacts activation of several signaling pathways, and drives expression of genes essential for microenvironmental interactions and lineage ambiguity. These data provide a robust rationale for targeting LTA as a treatment strategy for cHL patients.


Assuntos
Doença de Hodgkin/imunologia , Janus Quinase 2/imunologia , Linfotoxina-alfa/imunologia , NF-kappa B/imunologia , Fator de Transcrição STAT6/imunologia , Linhagem Celular , Regulação Neoplásica da Expressão Gênica , Doença de Hodgkin/genética , Humanos , Linfotoxina-alfa/genética , Células de Reed-Sternberg/imunologia , Células de Reed-Sternberg/metabolismo , Transdução de Sinais , Ativação Transcricional
6.
J Pathol ; 251(2): 160-174, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32222043

RESUMO

The IκB kinase (IKK)-NF-κB signaling pathway plays a multifaceted role in inflammatory bowel disease (IBD): on the one hand, it protects from apoptosis; on the other, it activates transcription of numerous inflammatory cytokines and chemokines. Although several murine models of IBD rely on disruption of IKK-NF-κB signaling, these involve either knockouts of a single family member of NF-κB or of upstream kinases that are known to have additional, NF-κB-independent, functions. This has made the distinct contribution of NF-κB to homeostasis in intestinal epithelium cells difficult to assess. To examine the role of constitutive NF-κB activation in intestinal epithelial cells, we generated a mouse model with a tissue-specific knockout of the direct inhibitor of NF-κB, Nfkbia/IκBα. We demonstrate that constitutive activation of NF-κB in intestinal epithelial cells induces several hallmarks of IBD including increased apoptosis, mucosal inflammation in both the small intestine and the colon, crypt hyperplasia, and depletion of Paneth cells, concomitant with aberrant Wnt signaling. To determine which NF-κB-driven phenotypes are cell-intrinsic, and which are extrinsic and thus require the immune compartment, we established a long-term organoid culture. Constitutive NF-κB promoted stem-cell proliferation, mis-localization of Paneth cells, and sensitization of intestinal epithelial cells to apoptosis in a cell-intrinsic manner. Increased number of stem cells was accompanied by a net increase in Wnt activity in organoids. Because aberrant Wnt signaling is associated with increased risk of cancer in IBD patients and because NFKBIA has recently emerged as a risk locus for IBD, our findings have critical implications for the clinic. In a context of constitutive NF-κB, our findings imply that general anti-inflammatory or immunosuppressive therapies should be supplemented with direct targeting of NF-κB within the epithelial compartment in order to attenuate apoptosis, inflammation, and hyperproliferation. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Assuntos
Apoptose , Doenças Inflamatórias Intestinais/metabolismo , Intestino Delgado/metabolismo , Inibidor de NF-kappaB alfa/deficiência , Celulas de Paneth/metabolismo , Células-Tronco/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Intestino Delgado/patologia , Camundongos Knockout , Inibidor de NF-kappaB alfa/genética , Organoides/metabolismo , Organoides/patologia , Celulas de Paneth/patologia , Células-Tronco/patologia , Fator de Transcrição RelA/metabolismo , Via de Sinalização Wnt
7.
Development ; 143(9): 1512-22, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26952977

RESUMO

In the epidermis of mice lacking transcription factor nuclear factor-kappa B (NF-κB) activity, primary hair follicle (HF) pre-placode formation is initiated without progression to proper placodes. NF-κB modulates WNT and SHH signaling at early stages of HF development, but this does not fully account for the phenotypes observed upon NF-κB inhibition. To identify additional NF-κB target genes, we developed a novel method to isolate and transcriptionally profile primary HF placodes with active NF-κB signaling. In parallel, we compared gene expression at the same developmental stage in NF-κB-deficient embryos and controls. This uncovered novel NF-κB target genes with potential roles in priming HF placodes for down-growth. Importantly, we identify Lhx2 (encoding a LIM/homeobox transcription factor) as a direct NF-κB target gene, loss of which replicates a subset of phenotypes seen in NF-κB-deficient embryos. Lhx2 and Tgfb2 knockout embryos exhibit very similar abnormalities in HF development, including failure of the E-cadherin suppression required for follicle down-growth. We show that TGFß2 signaling is impaired in NF-κB-deficient and Lhx2 knockout embryos and that exogenous TGFß2 rescues the HF phenotypes in Lhx2 knockout skin explants, indicating that it operates downstream of LHX2. These findings identify a novel NF-κB/LHX2/TGFß2 signaling axis that is crucial for primary HF morphogenesis, which may also function more broadly in development and disease.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Folículo Piloso/embriologia , Proteínas com Homeodomínio LIM/genética , Organogênese/genética , Fator de Transcrição RelA/genética , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta2/genética , Animais , Caderinas/metabolismo , Diferenciação Celular/genética , Movimento Celular/genética , Embrião de Mamíferos/metabolismo , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
8.
Genes Dev ; 25(20): 2137-46, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21979374

RESUMO

In malignancies, enhanced nuclear factor-κB (NF-κB) activity is largely viewed as an oncogenic property that also confers resistance to chemotherapy. Recently, NF-κB has been postulated to participate in a senescence-associated and possibly senescence-reinforcing cytokine response, thereby suggesting a tumor-restraining role for NF-κB. Using a mouse lymphoma model and analyzing transcriptome and clinical data from lymphoma patients, we show here that therapy-induced senescence presents with and depends on active NF-κB signaling, whereas NF-κB simultaneously promotes resistance to apoptosis. Further characterization and genetic engineering of primary mouse lymphomas according to distinct NF-κB-related oncogenic networks reminiscent of diffuse large B-cell lymphoma (DLBCL) subtypes guided us to identify Bcl2-overexpressing germinal center B-cell-like (GCB) DLBCL as a clinically relevant subgroup with significantly superior outcome when NF-κB is hyperactive. Our data illustrate the power of cross-species investigations to functionally test genetic mechanisms in transgenic mouse tumors that recapitulate distinct features of the corresponding human entity, and to ultimately use the mouse model-derived genetic information to redefine novel, clinically relevant patient subcohorts.


Assuntos
Antineoplásicos/farmacologia , Senescência Celular/efeitos dos fármacos , Senescência Celular/fisiologia , NF-kappa B/metabolismo , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/metabolismo , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma não Hodgkin/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
9.
Mol Cell ; 40(1): 63-74, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20932475

RESUMO

As part of the genotoxic stress response, cells activate the transcription factor NF-κB. The DNA strand break sensor poly(ADP-ribose)-polymerase-1 (PARP-1) and the kinase ataxia telangiectasia mutated (ATM) act as proximal signal mediators. PARP-1 assembles a nucleoplasmic signalosome, which triggers PIASy-mediated IKKγ SUMOylation. ATM-dependent IKKγ phosphorylation and subsequent ubiquitination were implicated to activate the cytoplasmic IκB kinase (IKK) complex by unknown mechanisms. We show that activated ATM translocates in a calcium-dependent manner to cytosol and membrane fractions. Through a TRAF-binding motif, ATM activates TRAF6, resulting in Ubc13-mediated K63-linked polyubiquitin synthesis and cIAP1 recruitment. The ATM-TRAF6-cIAP1 module stimulates TAB2-dependent TAK1 phosphorylation. Both nuclear PARP-1- and cytoplasmic ATM-driven signaling branches converge at the IKK complex to catalyze monoubiquitination of IKKγ at K285. Our data indicate that exported SUMOylated IKKγ acts as a substrate. IKKγ monoubiquitination is a prerequisite for genotoxic IKK and NF-κB activation, but also promotes cytokine signaling.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Citosol/enzimologia , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Núcleo Celular/enzimologia , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/genética , Células HeLa , Células Hep G2 , Humanos , Quinase I-kappa B/metabolismo , Proteínas Inibidoras de Apoptose/genética , MAP Quinase Quinase Quinases/metabolismo , Mutação , NF-kappa B/genética , Fosforilação , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Interferência de RNA , Proteínas Recombinantes/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Supressoras de Tumor/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação
10.
J Am Soc Nephrol ; 28(11): 3191-3204, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28687535

RESUMO

ANCA-associated vasculitis (AAV) is a highly inflammatory condition in which ANCA-activated neutrophils interact with the endothelium, resulting in necrotizing vasculitis. We tested the hypothesis that endothelial NF-κB mediates necrotizing crescentic GN (NCGN) and provides a specific treatment target. Reanalysis of kidneys from previously examined murine NCGN disease models revealed NF-κB activation in affected kidneys, mostly as a p50/p65 heterodimer, and increased renal expression of NF-κB-dependent tumor necrosis factor α (TNF-α). NF-κB activation positively correlated with crescent formation, and nuclear phospho-p65 staining showed NF-κB activation within CD31-expressing endothelial cells (ECs) in affected glomeruli. Therefore, we studied the effect of ANCA on NF-κB activation in neutrophil/EC cocultures in vitro ANCA did not activate NF-κB in primed human neutrophils, but ANCA-stimulated primed neutrophils activated NF-κB in ECs, at least in part via TNF-α release. This effect increased endothelial gene transcription and protein production of NF-κB-regulated interleukin-8. Moreover, upregulation of endothelial NF-κB promoted neutrophil adhesion to EC monolayers, an effect that was inhibited by a specific IKKß inhibitor. In a murine NCGN model, prophylactic application of E-selectin-targeted immunoliposomes packed with p65 siRNA to downregulate endothelial NF-κB significantly reduced urine abnormalities, renal myeloid cell influx, and NCGN. Increased glomerular endothelial phospho-p65 staining in patients with AAV indicated that NF-κB is activated in human NCGN also. We suggest that ANCA-stimulated neutrophils activate endothelial NF-κB, which contributes to NCGN and provides a potential therapeutic target in AAV.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/etiologia , Glomerulonefrite/etiologia , NF-kappa B/fisiologia , Animais , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/prevenção & controle , Células Cultivadas , Endotélio Vascular , Glomerulonefrite/patologia , Glomérulos Renais/patologia , Camundongos , NF-kappa B/antagonistas & inibidores , Necrose , Ativação de Neutrófilo
11.
Mol Cell ; 36(3): 365-78, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19917246

RESUMO

Upon genotoxic stresses, cells activate IkappaB kinases (IKKs) and the transcription factor NF-kappaB to modulate apoptotic responses. The SUMO-1 ligase PIASy and the kinase ataxia talengiectasia mutated (ATM) have been implicated to SUMOylate and phosphorylate nuclear IKKgamma (NEMO) in a consecutive mode of action, which in turn results in activation of cytoplasmic IKK holocomplexes. However, the nuclear signals and scaffold structures that initiate IKKgamma recruitment and activation are unknown. Here, we show that poly(ADP-ribose)-polymerase-1 (PARP-1) is the DNA proximal regulator, which senses DNA strand breaks and, through poly(ADP-ribose) (PAR) synthesis, assembles IKKgamma, PIASy, and ATM in a dynamic manner. Signalosome formation involves direct protein-protein interactions and binding to ADP-ribose polymers through PAR binding motifs (PARBM). Activated PARP-1 and a PARBM in PIASy are required to trigger IKKgamma SUMOylation, which in turn permits IKK and NF-kappaB activation, as well as NF-kappaB-regulated resistance to apoptosis.


Assuntos
Dano ao DNA , Quinase I-kappa B/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Humanos , Quinase I-kappa B/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Dados de Sequência Molecular , NF-kappa B/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Homologia de Sequência de Aminoácidos , Proteínas Supressoras de Tumor/metabolismo
12.
Proc Natl Acad Sci U S A ; 111(42): E4513-22, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25288773

RESUMO

Deregulated transcription factor (TF) activities are commonly observed in hematopoietic malignancies. Understanding tumorigenesis therefore requires determining the function and hierarchical role of individual TFs. To identify TFs central to lymphomagenesis, we identified lymphoma type-specific accessible chromatin by global mapping of DNaseI hypersensitive sites and analyzed enriched TF-binding motifs in these regions. Applying this unbiased approach to classical Hodgkin lymphoma (HL), a common B-cell-derived lymphoma with a complex pattern of deregulated TFs, we discovered interferon regulatory factor (IRF) sites among the top enriched motifs. High-level expression of the proinflammatory TF IRF5 was specific to HL cells and crucial for their survival. Furthermore, IRF5 initiated a regulatory cascade in human non-Hodgkin B-cell lines and primary murine B cells by inducing the TF AP-1 and cooperating with NF-κB to activate essential characteristic features of HL. Our strategy efficiently identified a lymphoma type-specific key regulator and uncovered a tumor promoting role of IRF5.


Assuntos
Cromatina/metabolismo , Doença de Hodgkin/genética , Doença de Hodgkin/metabolismo , Fatores Reguladores de Interferon/metabolismo , Fator de Transcrição AP-1/metabolismo , Motivos de Aminoácidos , Animais , Linfócitos B/citologia , Linhagem Celular Tumoral , Linhagem da Célula , Quimiocinas/metabolismo , Quimiotaxia , Citocinas/metabolismo , Desoxirribonuclease I/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Inflamação , Leucócitos Mononucleares/citologia , Linfoma/metabolismo , Linfoma não Hodgkin/metabolismo , Camundongos , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos/metabolismo , Baço/citologia
13.
J Am Soc Nephrol ; 27(9): 2658-69, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26823548

RESUMO

NF-κB is a key regulator of innate and adaptive immunity and is implicated in the pathogenesis of AKI. The cell type-specific functions of NF-κB in the kidney are unknown; however, the pathway serves distinct functions in immune and tissue parenchymal cells. We analyzed tubular epithelial-specific NF-κB signaling in a mouse model of ischemia-reperfusion injury (IRI)-induced AKI. NF-κB reporter activity and nuclear localization of phosphorylated NF-κB subunit p65 analyses in mice revealed that IRI induced widespread NF-κB activation in renal tubular epithelia and in interstitial cells that peaked 2-3 days after injury. To genetically antagonize tubular epithelial NF-κB activity, we generated mice expressing the human NF-κB super-repressor IκBαΔN in renal proximal, distal, and collecting duct epithelial cells. Compared with control mice, these mice exhibited improved renal function, reduced tubular apoptosis, and attenuated neutrophil and macrophage infiltration after IRI-induced AKI. Furthermore, tubular NF-κB-dependent gene expression profiles revealed temporally distinct functional gene clusters for apoptosis, chemotaxis, and morphogenesis. Primary proximal tubular cells isolated from IκBαΔN-expressing mice and exposed to hypoxia-mimetic agent cobalt chloride exhibited less apoptosis and expressed lower levels of chemokines than cells from control mice did. Our results indicate that postischemic NF-κB activation in renal tubular epithelia aggravates tubular injury and exacerbates a maladaptive inflammatory response.


Assuntos
Injúria Renal Aguda/etiologia , NF-kappa B/fisiologia , Animais , Apoptose , Modelos Animais de Doenças , Túbulos Renais , Masculino , Camundongos , Traumatismo por Reperfusão , Transdução de Sinais , Urotélio
14.
EMBO Rep ; 15(1): 46-61, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24375677

RESUMO

The IκB kinase (IKK) complex is the signal integration hub for NF-κB activation. Composed of two serine-threonine kinases (IKKα and IKKß) and the regulatory subunit NEMO (also known as IKKγ), the IKK complex integrates signals from all NF-κB activating stimuli to catalyze the phosphorylation of various IκB and NF-κB proteins, as well as of other substrates. Since the discovery of the IKK complex components about 15 years ago, tremendous progress has been made in the understanding of the IKK architecture and its integration into signaling networks. In addition to the control of NF-κB, IKK subunits mediate the crosstalk with other pathways, thereby extending the complexity of their biological function. This review summarizes recent advances in IKK biology and focuses on emerging aspects of IKK structure, regulation and function.


Assuntos
Quinase I-kappa B/fisiologia , NF-kappa B/metabolismo , Ubiquitinação , Animais , Retroalimentação Fisiológica , Humanos , Fosforilação , Mapas de Interação de Proteínas , Multimerização Proteica , Transdução de Sinais , Ubiquitina/fisiologia
15.
Immunol Rev ; 246(1): 59-76, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22435547

RESUMO

The inhibitory IκB proteins have been discovered as fundamental regulators of the inducible transcription factor nuclear factor-κB (NF-κB). As a generally excepted model, stimulus-dependent destruction of inhibitory IκBs and processing of precursor molecules, both promoted by components of the signal integrating IκB kinase complex, are the key events for the release of various NF-κB/Rel dimers and subsequent transcriptional activation. Intense research of more than 20 years provides evidence that the extending family of IκBs act not simply as reversible inhibitors of NF-κB activation but rather as a complex regulatory module, which assures feedback regulation of the NF-κB system and either can inhibit or promote transcriptional activity in a stimulus-dependent manner. Thus, IκB and NF-κB/Rel family proteins establish a complex interrelationship that allows modulated NF-κB-dependent transcription, tailored to the physiological environment.


Assuntos
Proteínas I-kappa B/metabolismo , NF-kappa B/metabolismo , Animais , Regulação da Expressão Gênica , Humanos , Proteínas I-kappa B/genética , NF-kappa B/genética , Ligação Proteica , Transdução de Sinais
16.
Blood ; 122(13): 2242-50, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-23869088

RESUMO

Constitutive activation of the nuclear factor-κ B (NF-κB) pathway is a hallmark of the activated B-cell-like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL). Recurrent mutations of NF-κB regulators that cause constitutive activity of this oncogenic pathway have been identified. However, it remains unclear how specific target genes are regulated. We identified the atypical nuclear IκB protein IκB-ζ to be upregulated in ABC compared with germinal center B-cell-like (GCB) DLBCL primary patient samples. Knockdown of IκB-ζ by RNA interference was toxic to ABC but not to GCB DLBCL cell lines. Gene expression profiling after IκB-ζ knockdown demonstrated a significant downregulation of a large number of known NF-κB target genes, indicating an essential role of IκB-ζ in regulating a specific set of NF-κB target genes. To further investigate how IκB-ζ mediates NF-κB activity, we performed immunoprecipitations and detected a physical interaction of IκB-ζ with both p50 and p52 NF-κB subunits, indicating that IκB-ζ interacts with components of both the canonical and the noncanonical NF-κB pathway in ABC DLBCL. Collectively, our data demonstrate that IκB-ζ is essential for nuclear NF-κB activity in ABC DLBCL, and thus might represent a promising molecular target for future therapies.


Assuntos
Redes Reguladoras de Genes , Linfoma Difuso de Grandes Células B/metabolismo , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Western Blotting , Ensaio de Imunoadsorção Enzimática , Técnicas de Silenciamento de Genes , Humanos , Proteínas I-kappa B , Imunoprecipitação , Linfoma Difuso de Grandes Células B/genética , NF-kappa B/genética , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Transdução de Sinais/fisiologia , Transcriptoma , Transdução Genética
17.
iScience ; 26(10): 107917, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37817938

RESUMO

The activation of IKK/NF-κB by genotoxic stress is a crucial process in the DNA damage response. Due to the anti-apoptotic impact of NF-κB, it can affect cell-fate decisions upon DNA damage and therefore interfere with tumor therapy-induced cell death. Here, we developed a dynamical model describing IKK/NF-κB signaling that faithfully reproduces quantitative time course data and enables a detailed analysis of pathway regulation. The approach elucidates a pathway topology with two hubs, where the first integrates signals from two DNA damage sensors and the second forms a coherent feedforward loop. The analyses reveal a critical role of the sensor protein PARP-1 in the pathway regulation. Introducing a method for calculating the impact of changes in individual components on pathway activity in a time-resolved manner, we show how irradiation dose influences pathway activation. Our results give a mechanistic understanding relevant for the interpretation of experimental and clinical studies.

18.
Cell Chem Biol ; 30(10): 1303-1312.e3, 2023 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-37506701

RESUMO

Transcription factor NF-κB potently activates anti-apoptotic genes, and its inactivation significantly reduces tumor cell survival following genotoxic stresses. We identified two structurally distinct lead compounds that selectively inhibit NF-κB activation by DNA double-strand breaks, but not by other stimuli, such as TNFα. Our compounds do not directly inhibit previously identified regulators of this pathway, most critically including IκB kinase (IKK), but inhibit signal transmission in-between ATM, PARP1, and IKKγ. Deconvolution strategies, including derivatization and in vitro testing in multi-kinase panels, yielded shared targets, cdc-like kinase (CLK) 2 and 4, as essential regulators of DNA damage-induced IKK and NF-κB activity. Both leads sensitize to DNA damaging agents by increasing p53-induced apoptosis, thereby reducing cancer cell viability. We propose that our lead compounds and derivatives can be used in context of genotoxic therapy-induced or ongoing DNA damage to increase tumor cell apoptosis, which may be beneficial in cancer treatment.


Assuntos
NF-kappa B , Transdução de Sinais , NF-kappa B/metabolismo , Dano ao DNA , Regulação da Expressão Gênica , DNA
19.
Nat Commun ; 14(1): 6947, 2023 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-37935654

RESUMO

Disease-causing mutations in genes encoding transcription factors (TFs) can affect TF interactions with their cognate DNA-binding motifs. Whether and how TF mutations impact upon the binding to TF composite elements (CE) and the interaction with other TFs is unclear. Here, we report a distinct mechanism of TF alteration in human lymphomas with perturbed B cell identity, in particular classic Hodgkin lymphoma. It is caused by a recurrent somatic missense mutation c.295 T > C (p.Cys99Arg; p.C99R) targeting the center of the DNA-binding domain of Interferon Regulatory Factor 4 (IRF4), a key TF in immune cells. IRF4-C99R fundamentally alters IRF4 DNA-binding, with loss-of-binding to canonical IRF motifs and neomorphic gain-of-binding to canonical and non-canonical IRF CEs. IRF4-C99R thoroughly modifies IRF4 function by blocking IRF4-dependent plasma cell induction, and up-regulates disease-specific genes in a non-canonical Activator Protein-1 (AP-1)-IRF-CE (AICE)-dependent manner. Our data explain how a single mutation causes a complex switch of TF specificity and gene regulation and open the perspective to specifically block the neomorphic DNA-binding activities of a mutant TF.


Assuntos
Fatores Reguladores de Interferon , Linfoma , Humanos , Linfócitos B/metabolismo , DNA , Regulação da Expressão Gênica , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Linfoma/genética
20.
EMBO J ; 26(22): 4634-45, 2007 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-17948050

RESUMO

Triggering of antigen receptors on lymphocytes is critical for initiating adaptive immune response against pathogens. T-cell receptor (TCR) engagement induces the formation of the Carma1-Bcl10-Malt1 (CBM) complex that is essential for activation of the IkappaB kinase (IKK)/NF-kappaB pathway. However, the molecular mechanisms that link CBM complex formation to IKK activation remain unclear. Here we report that Malt1 is polyubiquitinated upon T-cell activation. Ubiquitin chains on Malt1 provide a docking surface for the recruitment of the IKK regulatory subunit NEMO/IKKgamma. TRAF6 associates with Malt1 in response to T-cell activation and can function as an E3 ligase for Malt1 in vitro and in vivo, mediating lysine 63-linked ubiquitination of Malt1. Multiple lysine residues in the C-terminus of Malt1 serve as acceptor sites for the assembly of polyubiquitin chains. Malt1 mutants that lack C-terminal ubiquitin acceptor lysines are impaired in rescuing NF-kappaB signaling and IL-2 production in Malt1-/- T cells. Thus, our data demonstrate that induced Malt1 ubiquitination is critical for the engagement of CBM and IKK complexes, thereby directing TCR signals to the canonical NF-kappaB pathway.


Assuntos
Caspases/metabolismo , NF-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 10 de Linfoma CCL de Células B , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Linhagem Celular , Guanilato Ciclase/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Interleucina-2/metabolismo , Células Jurkat , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa , Linfócitos T/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA