Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cells ; 11(23)2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36497121

RESUMEN

CEP55 is a central regulator of late cytokinesis and is overexpressed in numerous cancers. Its post-translationally controlled recruitment to the midbody is crucial to the structural coordination of the abscission sequence. Our recent evidence that CEP55 contains two ubiquitin-binding domains was the first structural and functional link between ubiquitin signaling and ESCRT-mediated severing of the intercellular bridge. So far, high-content screens focusing on cytokinesis have used multinucleation as the endpoint readout. Here, we report an automated image-based detection method of intercellular bridges, which we applied to further our understanding of late cytokinetic signaling by performing an RNAi screen of ubiquitin ligases and deubiquitinases. A secondary validation confirmed four candidate genes, i.e., LNX2, NEURL, UCHL1 and RNF157, whose downregulation variably affects interconnected phenotypes related to CEP55 and its UBDs, as follows: decreased recruitment of CEP55 to the midbody, increased number of midbody remnants per cell, and increased frequency of intercellular bridges or multinucleation events. This brings into question the Notch-dependent or independent contributions of LNX2 and NEURL proteins to late cytokinesis. Similarly, the role of UCHL1 in autophagy could link its function with the fate of midbody remnants. Beyond the biological interest, this high-content screening approach could also be used to isolate anticancer drugs that act by impairing cytokinesis and CEP55 functions.


Asunto(s)
Proteínas Nucleares , Ubiquitina , Humanos , Ubiquitina/metabolismo , Proteínas Nucleares/metabolismo , Citocinesis/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Unión Proteica
2.
RSC Chem Biol ; 3(4): 456-467, 2022 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-35441144

RESUMEN

Epigenetic regulation is a dynamic and reversible process that controls gene expression. Abnormal function results in human diseases such as cancer, thus the enzymes that establish epigenetic marks, such as histone methyltransferases (HMTs), are potentially therapeutic targets. Noteworthily, HMTs form multiprotein complexes that in concert regulate gene expression. To probe epigenetic protein complexes regulation in cells, we developed a reliable chemical biology high-content imaging strategy to screen compound libraries simultaneously on multiple histone marks inside cells. By this approach, we identified that compound 4, a published CARM1 inhibitor, inhibits both histone mark H3R2me2a, regulated also by CARM1, and H3K79me2, regulated only by DOT1L, pointing out a crosstalk between CARM1 and DOT1L. Based on this interaction, we combined compound 4 and DOT1L inhibitor EPZ-5676 resulting in a stronger inhibition of cell proliferation and increase in apoptosis, indicating that our approach identifies possible effective synergistic drug combinations.

3.
Nat Commun ; 7: 12629, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27586688

RESUMEN

The NF-κB pathway has critical roles in cancer, immunity and inflammatory responses. Understanding the mechanism(s) by which mutations in genes involved in the pathway cause disease has provided valuable insight into its regulation, yet many aspects remain unexplained. Several lines of evidence have led to the hypothesis that the regulatory/sensor protein NEMO acts as a biological binary switch. This hypothesis depends on the formation of a higher-order structure, which has yet to be identified using traditional molecular techniques. Here we use super-resolution microscopy to reveal the existence of higher-order NEMO lattice structures dependent on the presence of polyubiquitin chains before NF-κB activation. Such structures may permit proximity-based trans-autophosphorylation, leading to cooperative activation of the signalling cascade. We further show that NF-κB activation results in modification of these structures. Finally, we demonstrate that these structures are abrogated in cells derived from incontinentia pigmenti patients.


Asunto(s)
Quinasa I-kappa B/ultraestructura , Incontinencia Pigmentaria/patología , Microscopía/métodos , FN-kappa B/metabolismo , Línea Celular Tumoral , Activación Enzimática , Humanos , Quinasa I-kappa B/metabolismo , Quinasa I-kappa B/fisiología , Unión Proteica , Estructura Secundaria de Proteína , Ubiquitina/metabolismo
4.
Methods Mol Biol ; 1278: 391-405, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25859965

RESUMEN

In the cell, homo- and hetero-associations of polypeptide chains evolve and take place within subcellular compartments that are crowded with many other cellular macromolecules. In vivo chemical cross-linking of proteins is a powerful method to examine changes in protein oligomerization and protein-protein interactions upon cellular events such as signal transduction. This chapter is intended to provide a guide for the selection of cell membrane permeable cross-linkers, the optimization of in vivo cross-linking conditions, and the identification of specific cross-links in a cellular context where the frequency of random collisions is high. By combining the chemoselectivity of the homo-bifunctional cross-linker and the length of its spacer arm with knowledge on the protein structure, we show that selective cross-links can be introduced specifically on either the dimer or the hexamer form of the same polypeptide in vitro as well as in vivo, using the human type B nucleoside diphosphate kinase as a protein model.


Asunto(s)
Nucleósido-Difosfato Quinasa/química , Péptidos/metabolismo , Mapeo de Interacción de Proteínas/métodos , Proteínas/metabolismo , Secuencia de Aminoácidos , Permeabilidad de la Membrana Celular , Humanos , Péptidos/química , Proteínas/química
5.
J Cell Biol ; 204(2): 231-45, 2014 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-24446482

RESUMEN

Nuclear factor κB (NF-κB) essential modulator (NEMO), a regulatory component of the IκB kinase (IKK) complex, controls NF-κB activation through its interaction with ubiquitin chains. We show here that stimulation with interleukin-1 (IL-1) and TNF induces a rapid and transient recruitment of NEMO into punctate structures that are anchored at the cell periphery. These structures are enriched in activated IKK kinases and ubiquitinated NEMO molecules, which suggests that they serve as organizing centers for the activation of NF-κB. These NEMO-containing structures colocalize with activated TNF receptors but not with activated IL-1 receptors. We investigated the involvement of nondegradative ubiquitination in the formation of these structures, using cells deficient in K63 ubiquitin chains or linear ubiquitin chain assembly complex (LUBAC)-mediated linear ubiquitination. Our results indicate that, unlike TNF, IL-1 requires K63-linked and linear ubiquitin chains to recruit NEMO into higher-order complexes. Thus, different mechanisms are involved in the recruitment of NEMO into supramolecular complexes, which appear to be essential for NF-κB activation.


Asunto(s)
Quinasa I-kappa B/metabolismo , Interleucina-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Línea Celular Tumoral , Células HeLa , Humanos , Quinasa I-kappa B/análisis , Interleucina-1/análisis , Interleucina-1/fisiología , Quinasas Asociadas a Receptores de Interleucina-1/análisis , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , FN-kappa B/análisis , FN-kappa B/metabolismo , Receptores de Interleucina-1/análisis , Receptores de Interleucina-1/metabolismo , Receptores del Factor de Necrosis Tumoral/análisis , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/fisiología , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina/fisiología , Ubiquitinación
6.
J Biol Chem ; 288(47): 33722-33737, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24100029

RESUMEN

Hypomorphic mutations in the X-linked human NEMO gene result in various forms of anhidrotic ectodermal dysplasia with immunodeficiency. NEMO function is mediated by two distal ubiquitin binding domains located in the regulatory C-terminal domain of the protein: the coiled-coil 2-leucine zipper (CC2-LZ) domain and the zinc finger (ZF) domain. Here, we investigated the effect of the D406V mutation found in the NEMO ZF of an ectodermal dysplasia with immunodeficiency patients. This point mutation does not impair the folding of NEMO ZF or mono-ubiquitin binding but is sufficient to alter NEMO function, as NEMO-deficient fibroblasts and Jurkat T lymphocytes reconstituted with full-length D406V NEMO lead to partial and strong defects in NF-κB activation, respectively. To further characterize the ubiquitin binding properties of NEMO ZF, we employed di-ubiquitin (di-Ub) chains composed of several different linkages (Lys-48, Lys-63, and linear (Met-1-linked)). We showed that the pathogenic mutation preferentially impairs the interaction with Lys-63 and Met-1-linked di-Ub, which correlates with its ubiquitin binding defect in vivo. Furthermore, sedimentation velocity and gel filtration showed that NEMO ZF, like other NEMO related-ZFs, binds mono-Ub and di-Ub with distinct stoichiometries, indicating the presence of a new Ub site within the NEMO ZF. Extensive mutagenesis was then performed on NEMO ZF and characterization of mutants allowed the proposal of a structural model of NEMO ZF in interaction with a Lys-63 di-Ub chain.


Asunto(s)
Displasia Ectodérmica/metabolismo , Quinasa I-kappa B/metabolismo , Síndromes de Inmunodeficiencia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mutación Missense , FN-kappa B/metabolismo , Ubiquitina/metabolismo , Sustitución de Aminoácidos , Animales , Displasia Ectodérmica/genética , Humanos , Quinasa I-kappa B/química , Quinasa I-kappa B/genética , Síndromes de Inmunodeficiencia/genética , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Células Jurkat , Ratones , Ratones Mutantes , Modelos Moleculares , FN-kappa B/química , FN-kappa B/genética , Unión Proteica/genética , Estructura Terciaria de Proteína , Ubiquitina/genética , Dedos de Zinc
7.
Biochem Pharmacol ; 82(9): 1163-74, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21803029

RESUMEN

Aberrant and constitutive NF-κB activation are frequently reported in numerous tumor types, making its inhibition an attractive target for the treatment of certain cancers. NEMO (NF-κB essential modulator) is the crucial component of the canonical NF-κB pathway that mediates IκB kinase (IKK) complex activation. IKK activation resides in the ability of the C-terminal domain of NEMO to properly dimerize and interact with linear and K63-linked polyubiquitin chains. Here, we have identified a new NEMO peptide inhibitor, termed UBI (ubiquitin binding inhibitor) that derives from the NOA/NUB/UBAN ubiquitin binding site located in the CC2-LZ domain of NEMO. UBI specifically inhibits the NF-κB pathway at the IKK level in different cell types stimulated by a variety of NF-κB signals. Circular dichroïsm and fluorescence studies showed that UBI exhibits an increased α-helix character and direct, good-affinity binding to the NOA-LZ region of NEMO. We also showed that UBI targets NEMO in cells but its mode of inhibition is completely different from the previously reported LZ peptide (herein denoted NOA-LZ). UBI does not promote dissociation of NEMO subunits in cells but impairs the interaction between the NOA UBD of NEMO and polyubiquitin chains. Importantly, we showed that UBI efficiently competes with the in vitro binding of K63-linked chains, but not with linear chains. The identification of this new NEMO inhibitor emphasizes the important contribution of K63-linked chains for IKK activation in NF-κB signaling and would provide a new tool for studying the complex role of NF-κB in inflammation and cancer.


Asunto(s)
Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Ubiquitina/metabolismo , Sitios de Unión , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Quinasa I-kappa B/genética , Modelos Moleculares , FN-kappa B/antagonistas & inhibidores , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína
8.
Blood ; 118(4): 926-35, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21622647

RESUMEN

Nuclear factor-κB essential modulator (NEMO), the regulatory subunit of the IκB kinase complex, is a critical component of the NF-κB pathway. Hypomorphic mutations in the X-linked human NEMO gene cause various forms of anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID). All known X-linked EDA-ID-causing mutations impair NEMO protein expression, folding, or both. We describe here 2 EDA-ID-causing missense mutations that affect the same residue in the CC2-LZ domain (D311N and D311G) that do not impair NEMO production or folding. Structural studies based on pull-down experiments showed a defect in noncovalent interaction with K63-linked and linear polyubiquitin chains for these mutant proteins. Functional studies on the patients' cells showed an impairment of the classic NF-κB signaling pathways after activation of 2 NEMO ubiquitin-binding-dependent receptors, the TNF and IL-1ß receptors, and in the CD40-dependent NF-κB pathway. We report the first human NEMO mutations responsible for X-linked EDA-ID found to affect the polyubiquitin binding of NEMO rather than its expression and folding. These experiments demonstrate that the binding of human NEMO to polyubiquitin is essential for NF-κB activation. They also demonstrate that the normal expression and folding of NEMO do not exclude a pathogenic role for NEMO mutations in patients with EDA-ID.


Asunto(s)
Displasia Ectodermal Anhidrótica Tipo 1/genética , Quinasa I-kappa B/genética , Síndromes de Inmunodeficiencia/genética , Ubiquitina/metabolismo , Western Blotting , Displasia Ectodermal Anhidrótica Tipo 1/metabolismo , Activación Enzimática/genética , Femenino , Humanos , Quinasa I-kappa B/metabolismo , Síndromes de Inmunodeficiencia/metabolismo , Masculino , Mutación Missense , FN-kappa B/metabolismo , Linaje , Unión Proteica , Pliegue de Proteína , Transducción de Señal/genética , Adulto Joven
9.
Blood ; 116(20): 4240-50, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-20671123

RESUMEN

The FOXO transcription factors are involved in multiple signaling pathways and have tumor-suppressor functions. In acute myeloid leukemia (AML), deregulation of oncogenic kinases, including Akt, extra-signal-regulated kinase, or IκB kinase, is frequently observed, which may potentially inactivate FOXO activity. We therefore investigated the mechanism underlying the regulation of FOXO3a, the only FOXO protein constantly expressed in AML blast cells. We show that in both primary AML samples and in a MV4-11/FOXO3a-GFP cell line, FOXO3a is in a constant inactive state due to its cytoplasmic localization, and that neither PI3K/Akt nor extra-signal-regulated kinase-specific inhibition resulted in its nuclear translocation. In contrast, the anti-Nemo peptide that specifically inhibits IKK activity was found to induce FOXO3a nuclear localization in leukemic cells. Furthermore, an IKK-insensitive FOXO3a protein mutated at S644 translocated into the nucleus and activated the transcription of the Fas-L and p21(Cip1) genes. This, in turn, inhibited leukemic cell proliferation and induced apoptosis. These results thus indicate that IKK activity maintains FOXO3a in the cytoplasm and establishes an important role of FOXO3a inactivation in the proliferation and survival of AML cells. The restoration of FOXO3a activity by interacting with its subcellular distribution may thus represent a new attractive therapeutic strategy for AML.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Quinasa I-kappa B/metabolismo , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Apoptosis/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proteína Forkhead Box O3 , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Mutantes/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Serina/metabolismo , Relación Estructura-Actividad
10.
J Mol Biol ; 395(1): 89-104, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19854204

RESUMEN

NEMO is an integral part of the IkappaB kinase complex and serves as a molecular switch by which the NF-kappaB signaling pathway can be regulated. Oligomerization and polyubiquitin (poly-Ub) binding, mediated through the regulatory CC2-LZ domain, were shown to be key features governing NEMO function, but the relationship between these two activities remains unclear. In this study, we solved the structure of this domain in complex with a designed ankyrin repeat protein, which helps its crystallization. We generated several NEMO mutants in this domain, including those associated with human diseases incontinentia pigmenti and immunodeficiency with or without anhidrotic ectodermal dysplasia. Analytical ultracentrifugation and thermal denaturation experiments were used to evaluate the dimerization properties of these mutants. A fluorescence-based assay was developed, as well, to quantify the interaction to monoubiquitin and poly-Ub chains. Moreover, the effect of these mutations was investigated for the full-length protein. We show that a proper folding of the ubiquitin-binding domain, termed NOA/UBAN/NUB, into a stable coiled-coil dimer is required but not sufficient for efficient interaction with poly-Ub. In addition, we show that binding to poly-Ub and, to a lesser extent, to monoubiquitin increases the stability of the NOA coiled-coil dimer. Collectively, these data provide structural insights into how several pathological mutations within and outside of the CC2-LZ's NOA ubiquitin binding site affect IkappaB kinase activation in the NF-kappaB signaling pathway.


Asunto(s)
Repetición de Anquirina , Quinasa I-kappa B/química , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Multimerización de Proteína , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Cristalografía por Rayos X , Humanos , Lisina/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , FN-kappa B/metabolismo , Poliubiquitina/metabolismo , Unión Proteica/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/farmacología
11.
J Biol Chem ; 284(5): 2902-2907, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19033441

RESUMEN

NEMO (NF-kappaB essential modulator) is a regulatory protein essential to the canonical NF-kappaB signaling pathway, notably involved in immune and inflammatory responses, apoptosis, and oncogenesis. Here, we report that the zinc finger (ZF) motif, located in the regulatory C-terminal half of NEMO, forms a specific complex with ubiquitin. We have investigated the NEMO ZF-ubiquitin interaction and proposed a structural model of the complex based on NMR, fluorescence, and mutagenesis data and on the sequence homology with the polymerase eta ubiquitin-binding zinc finger involved in DNA repair. Functional complementation assays and in vivo pull-down experiments further show that ZF residues involved in ubiquitin binding are functionally important and required for NF-kappaB signaling in response to tumor necrosis factor-alpha. Thus, our findings indicate that NEMOZFisa bona fide ubiquitin-binding domain of the ubiquitin-binding zinc finger type.


Asunto(s)
Ubiquitina/metabolismo , Dedos de Zinc , Secuencia de Aminoácidos , Sitios de Unión , Western Blotting , Línea Celular , Prueba de Complementación Genética , Humanos , Inmunoprecipitación , Células Jurkat , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Homología de Secuencia de Aminoácido , Ubiquitina/química
12.
J Mol Biol ; 377(5): 1419-32, 2008 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-18313693

RESUMEN

The regulatory NEMO (NF-kappaB essential modulator) protein has a crucial role in the canonical NF-kappaB signaling pathway notably involved in immune and inflammatory responses, apoptosis and oncogenesis. The regulatory domain is located in the C-terminal half of NEMO and contains a classical CCHC-type zinc finger (ZF). We have investigated the structural and functional effects of a cysteine to phenylalanine point mutation (C417F) in the ZF motif, identified in patients with anhidrotic ectodermal dysplasia with immunodeficiency. The solution structures of the wild type and mutant ZF were determined by NMR. Remarkably, the mutant adopts a global betabetaalpha fold similar to that of the wild type and retains thermodynamic stability, i.e., the ability to bind zinc with a native-like affinity, although the last zinc-chelating residue is missing. However, the mutation induces enhanced dynamics in the motif and leads to an important loss of stability. A detailed analysis of the wild type solution structure and experimental evidences led to the identification of two possible protein-binding surfaces that are largely destabilized in the mutant. This is sufficient to alter NEMO function, since functional complementation assays using NEMO-deficient pre-B and T lymphocytes show that full-length C417F pathogenic NEMO leads to a partial to strong defect in LPS, IL-1beta and TNF-alpha-induced NF-kappaB activation, respectively, as compared to wild type NEMO. Altogether, these results shed light onto the role of NEMO ZF as a protein-binding motif and show that a precise structural integrity of the ZF should be preserved to lead to a functional protein-recognition motif triggering full NF-kappaB activation.


Asunto(s)
Sitios de Unión , Quinasa I-kappa B/química , Quinasa I-kappa B/genética , Mutación Puntual , Dedos de Zinc , Dicroismo Circular , Displasia Ectodérmica Hipohidrótica Autosómica Recesiva/genética , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Quinasa I-kappa B/metabolismo , Síndromes de Inmunodeficiencia/genética , Células Jurkat , Espectroscopía de Resonancia Magnética , Modelos Moleculares , FN-kappa B/genética , FN-kappa B/metabolismo , Unión Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Soluciones , Espectrometría de Fluorescencia , Espectrometría de Masa por Ionización de Electrospray , Tirosina/química , Tirosina/genética , Tirosina/metabolismo , Zinc/metabolismo
13.
Protein Sci ; 16(9): 2013-22, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17766391

RESUMEN

The link between the NF-kappaB signal transduction pathway and cancer is now well established. Inhibiting this pathway is therefore a promising approach in the treatment of certain cancers through a pro-apoptotic effect in malignant cells. Owing to its central role in the pathway, the IkappaB kinase (IKK) complex is a privileged target for designing inhibitors. Previously, we showed that oligomerization of NEMO is necessary for IKK activation and defined a minimal oligomerization domain (CC2-LZ) for NEMO, and we developed NEMO peptides inhibiting NF-kappaB activation at the level of the IKK complex. To improve the low-affinity inhibitors, we used ribosome display to select small and stable proteins with high affinity against the individual CC2-LZ because the entire NEMO protein is poorly soluble. Several binders with affinities in the low nanomolar range were obtained. When expressed in human cells, some of the selected molecules, despite their partial degradation, inhibited TNF-alpha-mediated NF-kappaB activation while having no effect on the basal activity. Controls with a naive library member or null plasmid had no effect. Furthermore, we could show that this NF-kappaB inhibition occurs through a specific interaction between the binders and the endogenous NEMO, resulting in decreased IKK activation. These results indicate that in vitro selections with the NEMO subdomain alone as a target may be sufficient to lead to interesting compounds that are able to inhibit NF-kappaB activation.


Asunto(s)
Repetición de Anquirina , Quinasa I-kappa B/química , FN-kappa B/antagonistas & inhibidores , FN-kappa B/química , Ubiquitina/metabolismo , Línea Celular , ADN Complementario , Escherichia coli/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Riñón/citología , Luciferasas/análisis , Luciferasas/metabolismo , Modelos Moleculares , FN-kappa B/genética , FN-kappa B/metabolismo , Plásmidos , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/farmacología , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
14.
J Exp Med ; 203(7): 1745-59, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16818673

RESUMEN

Germline mutations in five autosomal genes involved in interleukin (IL)-12-dependent, interferon (IFN)-gamma-mediated immunity cause Mendelian susceptibility to mycobacterial diseases (MSMD). The molecular basis of X-linked recessive (XR)-MSMD remains unknown. We report here mutations in the leucine zipper (LZ) domain of the NF-kappaB essential modulator (NEMO) gene in three unrelated kindreds with XR-MSMD. The mutant proteins were produced in normal amounts in blood and fibroblastic cells. However, the patients' monocytes presented an intrinsic defect in T cell-dependent IL-12 production, resulting in defective IFN-gamma secretion by T cells. IL-12 production was also impaired as the result of a specific defect in NEMO- and NF-kappaB/c-Rel-mediated CD40 signaling after the stimulation of monocytes and dendritic cells by CD40L-expressing T cells and fibroblasts, respectively. However, the CD40-dependent up-regulation of costimulatory molecules of dendritic cells and the proliferation and immunoglobulin class switch of B cells were normal. Moreover, the patients' blood and fibroblastic cells responded to other NF-kappaB activators, such as tumor necrosis factor-alpha, IL-1beta, and lipopolysaccharide. These two mutations in the NEMO LZ domain provide the first genetic etiology of XR-MSMD. They also demonstrate the importance of the T cell- and CD40L-triggered, CD40-, and NEMO/NF-kappaB/c-Rel-mediated induction of IL-12 by monocyte-derived cells for protective immunity to mycobacteria in humans.


Asunto(s)
Antígenos CD40/fisiología , Genes Ligados a X , Predisposición Genética a la Enfermedad , Quinasa I-kappa B/genética , Interleucina-12/biosíntesis , Infecciones por Mycobacterium/genética , Infecciones por Mycobacterium/inmunología , Cromosoma X , Adolescente , Adulto , Animales , Línea Celular Transformada , Células Cultivadas , Niño , Preescolar , Femenino , Humanos , Lactante , Células L , Masculino , Ratones , Linaje
15.
J Biol Chem ; 281(10): 6334-48, 2006 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-16379012

RESUMEN

The NEMO (NF-kappaB essential modulator) protein plays a crucial role in the canonical NF-kappaB pathway as the regulatory component of the IKK (IkappaB kinase) complex. The human disease anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) has been recently linked to mutations in NEMO. We investigated the effect of an alanine to glycine substitution found in the NEMO polypeptide of an EDA-ID patient. This pathogenic mutation is located within the minimal oligomerization domain of the protein, which is required for the IKK activation in response to diverse stimuli. The mutation does not dramatically change the native-like state of the trimer, but temperature-induced unfolding studied by circular dichroism showed that it leads to an important loss in the oligomer stability. Furthermore, fluorescence studies showed that the tyrosine located in the adjacent zinc finger domain, which is possibly required for NEMO ubiquitination, exhibits an alteration in its spectral properties. This is probably due to a conformational change of this domain, providing evidence for a close interaction between the oligomerization domain and the zinc finger. In addition, functional complementation assays using NEMO-deficient pre-B and T lymphocytes showed that the pathogenic mutation reduced TNF-alpha and LPS-induced NF-kappaB activation by altering the assembly of the IKK complex. Altogether, our findings provide understanding as to how a single point mutation in NEMO leads to the observed EDA-ID phenotype in relation to the NEMO-dependent mechanism of IKK activation.


Asunto(s)
Displasia Ectodérmica/genética , Quinasa I-kappa B/genética , Síndromes de Inmunodeficiencia/genética , Lipopolisacáridos/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Mutación Puntual , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Dicroismo Circular , Displasia Ectodérmica/patología , Humanos , Quinasa I-kappa B/química , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/metabolismo , Síndromes de Inmunodeficiencia/patología , Lipopolisacáridos/farmacología , Ratones , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Temperatura , Tirosina/genética , Dedos de Zinc/genética
16.
J Biol Chem ; 279(52): 54248-57, 2004 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-15466857

RESUMEN

NF-kappa B essential modulator/IKK-gamma (NEMO/IKK-gamma) plays a key role in the activation of the NF-kappa B pathway in response to proinflammatory stimuli. Previous studies suggested that the signal-dependent activation of the IKK complex involves the trimerization of NEMO. The minimal oligomerization domain of this protein consists of two coiled-coil subdomains named Coiled-coil 2 (CC2) and leucine zipper (LZ) (Agou, F., Traincard, F., Vinolo, E., Courtois, G., Yamaoka, S., Israel, A., and Veron, M. (2004) J. Biol. Chem. 279, 27861-27869). To search for drugs inhibiting NF-kappa B activation, we have rationally designed cell-permeable peptides corresponding to the CC2 and LZ subdomains that mimic the contact areas between NEMO subunits. The peptides were tagged with the Antennapedia/Penetratin motif and delivered to cells prior to stimulation with lipopolysaccharide. Peptide transduction was monitored by fluorescence-activated cell sorter, and their effect on lipopolysaccharide-induced NF-kappa B activation was quantified using an NF-kappa B-dependent beta-galactosidase assay in stably transfected pre-B 70Z/3 lymphocytes. We show that the peptides corresponding to the LZ and CC2 subdomains inhibit NF-kappa B activation with an IC(50) in the mum range. Control peptides, including mutated CC2 and LZ peptides and a heterologous coiled-coil peptide, had no inhibitory effect. The designed peptides are able to induce cell death in human retinoblastoma Y79 cells exhibiting constitutive NF-kappa B activity. Our results provide the "proof of concept" for a new and promising strategy for the inhibition of NF-kappa B pathway activation through targeting the oligomerization state of the NEMO protein.


Asunto(s)
FN-kappa B/antagonistas & inhibidores , FN-kappa B/química , Péptidos/farmacología , Proteínas Serina-Treonina Quinasas/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Muerte Celular/efectos de los fármacos , Línea Celular , Fenómenos Químicos , Química Física , ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/farmacología , Diseño de Fármacos , Complejos de Clasificación Endosomal Requeridos para el Transporte , Citometría de Flujo , Humanos , Quinasa I-kappa B , Interleucina-2/genética , Células Jurkat , Leucina Zippers/genética , Leucina Zippers/fisiología , Lipopolisacáridos/farmacología , Ratones , Datos de Secuencia Molecular , Mutagénesis , FN-kappa B/metabolismo , Péptidos/química , Péptidos/genética , Regiones Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinasas/genética , Subunidades de Proteína/química , Proteínas Recombinantes de Fusión , Retinoblastoma , Relación Estructura-Actividad , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/farmacología , Transfección , Células Tumorales Cultivadas , beta-Galactosidasa/genética
17.
J Biol Chem ; 279(27): 27861-9, 2004 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-15107419

RESUMEN

NEMO (NF-kappaB essential modulator) plays a key role in the canonical NF-kappaB pathway as the scaffold/regulatory component of the IkappaB kinase (IKK) complex. The self-association of NEMO involves the C-terminal halves of the polypeptide chains containing two putative coiled-coil motifs (a CC2 and a LZ leucine zipper), a proline-rich region, and a ZF zinc finger motif. Using purified truncation mutants, we showed that the minimal oligomerization domain of NEMO is the CC2-LZ segment and that both CC2 and LZ subdomains are necessary to restore the LPS-dependent activation of the NF-kappaB pathway in a NEMO-deficient cell line. We confirmed the association of the oligomerization domain in a trimer and investigated the specific role of CC2 and LZ subdomains in the building of the oligomer. Whereas a recombinant CC2-LZ polypeptide self-associated into a trimer with an association constant close to that of the wild-type protein, the isolated CC2 and LZ peptides, respectively, formed trimers and dimers with weaker association constants. Upon mixing, isolated CC2 and LZ peptides associated to form a stable hetero-hexamer as shown by gel filtration and fluorescence anisotropy experiments. We propose a structural model for the organization of the oligomerization domain of activated NEMO in which three C-terminal domains associate into a pseudo-hexamer forming a six-helix bundle. This model is discussed in relation to the mechanism of activation of the IKK complex by upstream activators.


Asunto(s)
Proteínas Serina-Treonina Quinasas/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Anisotropía , Línea Celular , Cromatografía , Cromatografía en Gel , ADN Complementario/metabolismo , Dimerización , Relación Dosis-Respuesta a Droga , Prueba de Complementación Genética , Vectores Genéticos , Quinasa I-kappa B , Cinética , Lipopolisacáridos/metabolismo , Ratones , Microscopía Fluorescente , Modelos Genéticos , Datos de Secuencia Molecular , Mutación , FN-kappa B/metabolismo , Péptidos/química , Pruebas de Precipitina , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Transfección , Dedos de Zinc
18.
Methods Mol Biol ; 261: 427-42, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15064474

RESUMEN

In the cell, homo- and heteroassociations of polypeptide chains evolve and take place within subcellular compartments that are crowded with many other cellular macromolecules. In vivo chemical cross-linking of proteins is a powerful method to examine changes in protein oligomerization and protein-protein interactions upon cellular events such as signal transduction. This chapter is intended to provide a guide to the selection of the cell-membrane-permeable cross-linkers, the optimization of in vivo cross-linking conditions, and the identification of specific cross-links in a cellular context where the frequency of random collisions is high. By combining the chemoselectivity of the homo-bifunctional cross-linker and the length of its spacer arm with knowledge on the protein structure, we show that selective cross-links can be introduced specifically on either the dimer or the hexamer form of the same polypeptide in vitro as well as in vivo, using the human type B nucleoside diphosphate kinase as a protein model.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Proteínas/química , Células HeLa , Humanos , Modelos Moleculares , Nucleósido-Difosfato Quinasa/química , Nucleósido-Difosfato Quinasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA