Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Biol Chem ; 291(16): 8440-52, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26826124

RESUMEN

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.


Asunto(s)
Canales de Calcio/biosíntesis , Señalización del Calcio/fisiología , Calcio/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de Neoplasias/biosíntesis , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Factor de Transcripción ReIA/metabolismo , Activación Transcripcional/fisiología , Canales de Calcio/genética , Humanos , Células Jurkat , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Proteína ORAI1 , Fosforilación/fisiología , Receptores de Antígenos de Linfocitos T/genética , Molécula de Interacción Estromal 1 , Factor de Transcripción ReIA/genética
2.
J Immunol ; 185(5): 2665-9, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20693425

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas I-kappa B/metabolismo , Proteínas Portadoras/fisiología , Línea Celular , Células HeLa , Proteínas de Andamiaje Homer , Humanos , Proteínas I-kappa B/fisiología , Sinapsis Inmunológicas/enzimología , Sinapsis Inmunológicas/inmunología , Células Jurkat , Estructura Terciaria de Proteína , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/inmunología , Fracciones Subcelulares/inmunología , Fracciones Subcelulares/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo
3.
J Biol Chem ; 285(49): 38069-77, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-20923761

RESUMEN

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.


Asunto(s)
Núcleo Celular/metabolismo , Quimiocina CXCL12/biosíntesis , Células Endoteliales/metabolismo , Transducción de Señal/fisiología , Factor de Transcripción ReIB/metabolismo , Venas Umbilicales/metabolismo , Regulación hacia Arriba/fisiología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Línea Celular , Núcleo Celular/genética , Quimiocina CXCL12/genética , Células Endoteliales/citología , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Mutación Missense , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIB/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Venas Umbilicales/citología , Regulación hacia Arriba/efectos de los fármacos
4.
J Biol Chem ; 284(40): 27596-608, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-19666475

RESUMEN

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.


Asunto(s)
Quinasa I-kappa B/química , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , FN-kappa B/metabolismo , Animales , Línea Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Quinasa I-kappa B/genética , Interleucina-1/farmacología , Ratones , Estructura Terciaria de Proteína , Eliminación de Secuencia , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología
5.
J Med Chem ; 63(6): 2915-2929, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32134643

RESUMEN

To identify Janus kinase (JAK) inhibitors that selectively target gastrointestinal tissues with limited systemic exposures, a class of imidazopyrrolopyridines with a range of physical properties was prepared and evaluated. We identified compounds with low intrinsic permeability and determined a correlation between permeability and physicochemical properties, clogP and tPSA, for a subset of compounds. This low intrinsic permeability translated into compounds displaying high colonic exposure and low systemic exposure after oral dosing at 25 mg/kg in mouse. In a mouse PK/PD model, oral dosing of lead compound 2 demonstrated dose-dependent inhibition of pSTAT phosphorylation in colonic explants post-oral dose but low systemic exposure and no measurable systemic pharmacodynamic activity. We thus demonstrate the utility of JAK inhibitors with low intrinsic permeability as a feasible approach to develop gut-restricted, pharmacologically active molecules with a potential advantage over systemically available compounds that are limited by systemic on-target adverse events.


Asunto(s)
Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Inhibidores de las Cinasas Janus/farmacología , Inhibidores de las Cinasas Janus/farmacocinética , Piridinas/farmacología , Piridinas/farmacocinética , Administración Oral , Animales , Perros , Descubrimiento de Drogas , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Inhibidores de las Cinasas Janus/administración & dosificación , Inhibidores de las Cinasas Janus/química , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , Permeabilidad , Fosforilación/efectos de los fármacos , Piridinas/administración & dosificación , Piridinas/química
6.
Methods Mol Biol ; 512: 209-32, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19347279

RESUMEN

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).


Asunto(s)
Quinasa I-kappa B/farmacología , FN-kappa B/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Células HeLa/efectos de los fármacos , Células HeLa/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Immunoblotting , Luciferasas/metabolismo , Estructura Terciaria de Proteína , Factor de Necrosis Tumoral alfa/farmacología
7.
J Allergy Clin Immunol ; 122(6): 1169-1177.e16, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18851874

RESUMEN

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.


Asunto(s)
Bases de Datos Genéticas , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Mutación Missense , Sustitución de Aminoácidos , Apoptosis/genética , Antígenos CD40/metabolismo , Proteínas de Unión al ADN , Regulación de la Expresión Génica/genética , Prueba de Complementación Genética/métodos , Genotipo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , FN-kappa B/metabolismo , Proteínas Nucleares/biosíntesis , Fenotipo , Estructura Terciaria de Proteína/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/genética , Receptor Toll-Like 5/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Factor de Necrosis Tumoral alfa/metabolismo
8.
J Immunol ; 180(5): 3467-77, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18292573

RESUMEN

Activation of the classical and noncanonical NF-kappaB pathways by ligation of the lymphotoxin (LT)-beta receptor (LTbetaR) plays a crucial role in lymphoid organogenesis and in the generation of ectopic lymphoid tissue at sites of chronic inflammation. Within these microenvironments, LTbetaR signaling regulates the phenotype of the specialized high endothelial cells. However, the direct effects of LTbetaR ligation on endothelial cells remain unclear. We therefore questioned whether LTbetaR ligation could directly activate endothelial cells and regulate classical and noncanonical NF-kappaB-dependent gene expression. We demonstrate that the LTbetaR ligands LIGHT and LTalpha1beta2 activate both NF-kappaB pathways in HUVECs and human dermal microvascular endothelial cells (HDMEC). Classical pathway activation was less robust than TNF-induced signaling; however, only LIGHT and LTalpha1beta2 and not TNF activated the noncanonical pathway. LIGHT and LTalpha1beta2 induced the expression of classical NF-kappaB-dependent genes in HUVEC, including those encoding the adhesion molecules E-selectin, ICAM-1, and VCAM-1. Consistent with this stimulation, LTbetaR ligation up-regulated T cell adhesion to HUVEC. Furthermore, the homeostatic chemokine CXCL12 was up-regulated by LIGHT and LTalpha1beta2 but not TNF in both HUVEC and HDMEC. Using HUVEC retrovirally transduced with dominant negative IkappaB kinase alpha, we demonstrate that CXCL12 expression is regulated by the noncanonical pathway in endothelial cells. Our findings therefore demonstrate that LTbetaR ligation regulates gene expression in endothelial cells via both NF-kappaB pathways and we identify CXCL12 as a bona fide noncanonical NF-kappaB-regulated gene in these cells.


Asunto(s)
Endotelio Vascular/inmunología , Endotelio Vascular/patología , Regulación de la Expresión Génica/inmunología , Mediadores de Inflamación/fisiología , Heterotrímero de Linfotoxina alfa1 y beta2/fisiología , FN-kappa B/fisiología , Transducción de Señal/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Adhesión Celular/genética , Adhesión Celular/inmunología , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Línea Celular , Endotelio Vascular/metabolismo , Humanos , Ligandos , Heterotrímero de Linfotoxina alfa1 y beta2/metabolismo , Receptor beta de Linfotoxina/biosíntesis , Receptor beta de Linfotoxina/metabolismo , Receptor beta de Linfotoxina/fisiología , Linfotoxina-alfa/metabolismo , Linfotoxina-alfa/fisiología , Linfotoxina beta/metabolismo , Linfotoxina beta/fisiología , FN-kappa B/metabolismo , Transducción de Señal/genética , Piel/irrigación sanguínea , Piel/citología , Piel/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
9.
J Biol Chem ; 282(22): 16105-16, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17430892

RESUMEN

Cells lacking functional NF-kappaB die after ligation of some tumor necrosis factor (TNF) receptor family members through failure to express NF-kappaB-dependent anti-apoptotic genes. NF-kappaB activation requires the IkappaB kinase (IKK) complex containing two catalytic subunits named IKKalpha and IKKbeta that regulate distinct NF-kappaB pathways. IKKbeta is critical for classical signaling that induces pro-inflammatory and anti-apoptotic gene profiles, whereas IKKalpha regulates the non-canonical pathway involved in lymphoid organogenesis and B-cell development. To determine whether IKKalpha and IKKbeta differentially function in rescuing cells from death induced by activators of the classical and non-canonical pathways, we analyzed death after ligation of the TNF and lymphotoxin-beta receptors, respectively. Using murine embryonic fibroblasts (MEFs) lacking each of the IKKs, the caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone, and dominant negative Fas-associated death domain protein, we found that deletion of these kinases sensitized MEFs to distinct cell death pathways. MEFs lacking IKKalpha were sensitized to death in response to both cytokines that was entirely caspase-dependent, demonstrating that IKKalpha functions in this process. Surprisingly, death of IKKbeta-/- MEFs was not blocked by caspase inhibition, demonstrating that IKKbeta negatively regulates caspase-independent cell death (CICD). CICD was strongly activated by both TNF and lymphotoxin-beta receptor ligation in IKKbeta-/- MEFs and was accompanied by loss of mitochondrial membrane potential and the generation of reactive oxygen species. CICD was inhibited by the anti-oxidant butylated hydroxyanosole and overexpression of Bcl-2, neither of which blocked caspase-dependent apoptosis. Our findings, therefore, demonstrate that both IKKalpha and IKKbeta regulate cytokine-induced apoptosis, and IKKbeta additionally represses reactive oxygen species- and mitochondrial-dependent CICD.


Asunto(s)
Apoptosis/genética , Caspasas/metabolismo , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Especies Reactivas de Oxígeno/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Linfocitos B/citología , Linfocitos B/metabolismo , Inhibidores de Caspasas , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/citología , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/metabolismo , Receptor beta de Linfotoxina/agonistas , Receptor beta de Linfotoxina/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Organogénesis/efectos de los fármacos , Organogénesis/genética , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Agregación de Receptores/efectos de los fármacos , Agregación de Receptores/genética , Factor de Necrosis Tumoral alfa/farmacología , Quinasa de Factor Nuclear kappa B
10.
J Immunol ; 179(11): 7385-96, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18025182

RESUMEN

Studies of patients with congenital immunodeficiency due to mutation of the NF-kappaB essential modulator (NEMO) gene have demonstrated that NEMO integrity is required for NK cell cytotoxicity. Thus, we have studied the physiology of NF-kappaB activation in NK cells during the cytolytic program. In resting ex vivo human NK cells or cell lines, IkappaB was degraded after 10 min exposure to PMA and ionomycin, or TNF and was maximally degraded by 30 min. Ligation of several NK cell activation receptors including NKp30 induced a similar response and was blocked by pretreatment with the proteosome inhibitor MG132. There was no short-term effect on p100 processing, the signature of noncanonical NF-kappaB activation. NK cell IkappaB degradation corresponded to increases in nuclear NF-kappaB as detected by EMSA. Supershift of stimulated NK cells and fluorescence microscopy of individual NK cells in cytolytic conjugates demonstrated that the p65/p50 heterodimer was the primary NF-kappaB used. NF-kappaB function was evaluated in NK92 cells transduced with a kappaB GFP reporter, and their conjugation with K562 cells or ligation of NKp30 ligation resulted in rapid GFP accumulation. The latter was prevented by the Syk inhibitor piceatannol. Thus, NK cell activation signaling specifically induces transcriptional activation and synthesis of new NF-kappaB dependent proteins during the initiation of cytotoxicity.


Asunto(s)
Células Asesinas Naturales/inmunología , FN-kappa B/metabolismo , Receptores Inmunológicos/inmunología , Línea Celular , Proteínas Fluorescentes Verdes/inmunología , Humanos , Ionomicina/farmacología , Células K562 , Leupeptinas/farmacología , FN-kappa B/antagonistas & inhibidores , Receptor 3 Gatillante de la Citotoxidad Natural , Receptores Inmunológicos/antagonistas & inhibidores , Transducción de Señal/inmunología , Estilbenos/farmacología , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/farmacología
11.
J Biol Chem ; 282(12): 8724-33, 2007 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-17244613

RESUMEN

Activation of NF-kappaB by the pro-inflammatory cytokines tumor necrosis factor (TNF) and interleukin-1 (IL-1) requires the IkappaB kinase (IKK) complex, which contains two kinases named IKKalpha and IKKbeta and a critical regulatory subunit named NEMO. Although we have previously demonstrated that NEMO associates with both IKKs, genetic studies reveal that only its interaction with IKKbeta is required for TNF-induced NF-kappaB activation. To determine whether NEMO and IKKalpha can form a functional IKK complex capable of activating the classical NF-kappaB pathway in the absence of IKKbeta, we utilized a panel of mouse embryonic fibroblasts (MEFs) lacking each of the IKK complex subunits. This confirmed that TNF-induced IkappaBalpha degradation absolutely requires NEMO and IKKbeta. In contrast, we consistently observed intact IkappaBalpha degradation and NF-kappaB activation in response to IL-1 in two separate cell lines lacking IKKbeta. Furthermore, exogenously expressed, catalytically inactive IKKbeta blocked TNF- but not IL-1-induced IkappaBalpha degradation in wild-type MEFs, and reconstitution of IKKalpha/beta double knockout cells with IKKalpha rescued IL-1- but not TNF-induced NF-kappaB activation. Finally, we have shown that incubation of IKKbeta-deficient MEFs with a cell-permeable peptide that blocks the interaction of NEMO with the IKKs inhibits IL-1-induced NF-kappaB activation. Our results therefore demonstrate that NEMO and IKKalpha can form a functional IKK complex that activates the classical NF-kappaB pathway in response to IL-1 but not TNF. These findings further suggest NEMO differentially regulates the fidelity of the IKK subunits activated by distinct upstream signaling pathways.


Asunto(s)
Regulación de la Expresión Génica , Quinasa I-kappa B/metabolismo , Interleucina-1/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , FN-kappa B/metabolismo , Animales , Catálisis , Línea Celular , Activación Enzimática , Humanos , Quinasa I-kappa B/fisiología , Ratones , Péptidos/química , Unión Proteica , Proteínas Recombinantes/química , Transducción de Señal
13.
J Biol Chem ; 279(44): 45528-39, 2004 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-15319427

RESUMEN

Activation of NF-kappaB requires two highly related kinases named IKKalpha and IKKbeta that share identity in the nature and positioning of their structural domains. Despite their similarity, the kinases are functionally divergent, and we therefore sought to identify any structural features specific for IKKalpha or IKKbeta. We performed bioinformatics analysis, and we identified a region resembling a ubiquitin-like domain (UBL) that exists only in IKKbeta and that we named the UBL-like domain (ULD). Deletion of the ULD rendered IKKbeta catalytically inactive and unable to induce NF-kappaB activity, and overexpression of only the ULD dose-dependently inhibited tumor necrosis factor-alpha-induced NF-kappaB activity. The ULD could not be functionally replaced within IKKbeta by ubiquitin or the corresponding region of IKKalpha, whereas deletion of the equivalent section of IKKalpha did not affect its catalytic activity against IkappaBalpha or its activation by NF-kappaB-inducing kinase. We identified five residues conserved among the larger family of UBL-containing proteins and IKKbeta, and alanine scanning revealed that the leucine at position 353 (Leu(353)) is absolutely critical for IKKbeta-induced NF-kappaB activation. Most intriguingly, the L353A mutant was catalytically active but, unlike wild-type IKKbeta, formed a stable complex with the NF-kappaB p65 subunit. Our findings therefore establish the ULD as a critical functional domain specific for IKKbeta that might play a role in dissociating IKKbeta from p65.


Asunto(s)
Proteínas Serina-Treonina Quinasas/química , Ubiquitina/fisiología , Secuencia de Aminoácidos , Animales , Células COS , Catálisis , Células HeLa , Humanos , Quinasa I-kappa B , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Factor de Transcripción ReIA
14.
J Biol Chem ; 278(23): 21295-306, 2003 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-12663669

RESUMEN

In the presence of cycloheximide, tumor necrosis factor or interleukin-1 initiates caspase activation, loss of mitochondrial membrane potential (DeltaPsi), DNA degradation, and nuclear condensation and fragmentation characteristic of apoptotic cell death in human vascular endothelial cells (EC). Inhibition of phosphatidylinositol 3-kinase (PI3K) by LY294002, but not inhibition of Akt by dominant-negative mutation, also sensitizes EC to cytokine-initiated apoptosis. Cytokine-initiated caspase activation is slower and comparatively less with LY294002 than with cycloheximide. Cycloheximide but not LY294002 decreases expression of c-FLIP (cellular FLICE inhibitory protein), an inhibitor of caspase-8 activation. The caspase inhibitor zVADfmk completely blocks caspase activation, DNA degradation, and nuclear fragmentation in both cases but only prevents loss of DeltaPsi and cell death for cytokine plus cycloheximide treatment. In contrast, overexpression of Bcl-2 protects EC treated with cytokine plus LY294002 but not EC treated with cytokine plus cycloheximide. The cathepsin B inhibitor CA-074-Me prevents loss of DeltaPsi, caspase activation, and cell death for EC treated with cytokine plus LY294002 but has no effect on EC treated with cytokine plus cycloheximide. Cathepsin B translocates from lysosomes to cytosol following treatment with LY294002 prior to the activation of caspases. These results suggest that inhibition of PI3K allows cytokines to activate a cathepsin-dependent, mitochondrial death pathway in which caspase activation is secondary, is not inhibited by c-FLIP, and is not essential for cell death.


Asunto(s)
Apoptosis/fisiología , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Interleucina-1/farmacología , Péptidos y Proteínas de Señalización Intracelular , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas , Clorometilcetonas de Aminoácidos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Proteínas Portadoras/genética , Caspasa 3 , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Catepsina B/metabolismo , Núcleo Celular , Células Cultivadas , Cromonas/farmacología , Cicloheximida/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/genética , Retroviridae/genética , Transducción Genética , Factor de Necrosis Tumoral alfa/farmacología , Venas Umbilicales/citología
15.
Am J Pathol ; 161(4): 1485-95, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12368221

RESUMEN

We have examined the effects of interferon (IFN)-gamma on expression and function of CD95 (APO-1/Fas) and associated proteins in cultured human umbilical vein and dermal microvascular endothelial cells (HUVEC and HDMEC, respectively). Unstimulated cells express only low levels of CD95; IFN-gamma produces a time- and concentration-dependent increase of CD95 in both cell types at the mRNA and cell surface protein levels. IFN-gamma also produces an increase in expression of pro-caspase-8 (FLICE/MACH) but does not significantly change expression of either Fas-associated death domain (FADD) protein or cellular FLICE inhibitory protein (cFLIP), other proteins associated with the CD95 death-inducing signaling complex (DISC). Neither resting nor IFN-gamma-treated EC express detectable CD95L mRNA or protein. Untreated HUVEC and HDMEC show minimal apoptosis when transduced to express CD95L. Treatment of CD95L-transduced cells with IFN-gamma causes apoptosis within 24 to 36 hours that can be blocked by antagonistic anti-CD95 antibody or by the caspase-inhibitory peptide zVAD-FMK. The extent of apoptosis is increased by co-treatment with either the protein synthesis inhibitor cycloheximide or the phosphatidylinositol 3-kinase inhibitor LY294002. Untransduced HUVEC treated with IFN-gamma also undergo CD95-initiated apoptosis when mixed with CD95L-transduced HUVEC or when incubated with pharmacologically activated cytolytic T lymphocytes. Overexpression of CD95 in HUVEC confers sensitivity to CD95L in the absence of IFN-gamma-treatment. We conclude that IFN-gamma induces sensitivity of endothelium to CD95L-mediated apoptosis, and that this response may result from increased expression of CD95 and/or pro-caspase-8.


Asunto(s)
Apoptosis , Caspasas/genética , Endotelio Vascular/fisiología , Precursores Enzimáticos/genética , Interferón gamma/farmacología , Receptor fas/genética , Clorometilcetonas de Aminoácidos/farmacología , Antígenos CD/genética , Apoptosis/efectos de los fármacos , Caspasa 8 , Caspasa 9 , Muerte Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos , Humanos , Glicoproteínas de Membrana/genética , Microcirculación , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/irrigación sanguínea , Transfección , Venas Umbilicales , Receptor fas/fisiología
16.
Mol Pharmacol ; 66(3): 572-9, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15322249

RESUMEN

Tumor necrosis factor (TNF) receptor 1-associated death domain protein (TRADD) is an adaptor protein known to be involved in the TNF signaling pathway as well as signaling of other members of the TNF receptor superfamily, including DR3, DR6, p75(NTR), and the Epstein-Barr virus latent membrane protein 1. Current knowledge of the function of the adaptor protein has been derived from studies examining its over-expression in either wild-type or mutated forms. In this study, we analyzed the consequences of antisense oligonucleotide (ASO)-mediated depletion of endogenous TRADD on TNF induction of inflammation-related gene products, such as intercellular adhesion molecule-1, and associated kinase signaling pathways in human umbilical vein endothelial cells. A broader perspective of TRADD's role in TNF signaling was indicated by microarray gene expression analysis, where 20 of 24 genes that showed a 5-fold or greater increase in TNF-induced mRNA expression levels displayed a reduction in TNF-induced expression as a consequence of ASO-mediated knockdown of TRADD. Reduced activation of the nuclear factor-kappaB and c-Jun NH(2)-terminal kinase pathways, as measured by IkappaB-alpha protein levels and the extent of c-Jun phosphorylation, was also observed. These results indicate usage of antisense inhibitors of TRADD expression for modulating diseases associated with TRADD-dependent signal transduction pathways.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Proteínas/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Endotelio Vascular/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor 1 Asociado a Receptor de TNF
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA