Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Virol ; 94(20)2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32727874

RESUMEN

To ensure productive infection, herpesviruses utilize tegument proteins and nonstructural regulatory proteins to counteract cellular defense mechanisms and to reprogram cellular pathways. The M25 proteins of mouse cytomegalovirus (MCMV) belong to the betaherpesvirus UL25 gene family that encodes viral proteins implicated with regulatory functions. Through affinity purification and mass spectrometric analysis, we discovered the tumor suppressor protein p53 as a host factor interacting with the M25 proteins. M25-p53 interaction in infected and transfected cells was confirmed by coimmunoprecipitation. Moreover, the proteins colocalized in nuclear dot-like structures upon both infection and inducible expression of the two M25 isoforms. p53 accumulated in wild-type MCMV-infected cells, while this did not occur upon infection with a mutant lacking the M25 gene. Both M25 proteins were able to mediate the effect, identifying them as the first CMV proteins responsible for p53 accumulation during infection. Interaction with M25 proteins led to substantial prolongation of the half-life of p53. In contrast to the higher abundance of the p53 protein in wild-type MCMV-infected cells, the transcript levels of the prominent p53 target genes Cdkn1a and Mdm2 were diminished compared to cells infected with the ΔM25 mutant, and this was associated with reduced binding of p53 to responsive elements within the respective promoters. Notably, the productivity of the M25 deletion mutant was partially rescued on p53-negative fibroblasts. We propose that the MCMV M25 proteins sequester p53 molecules in the nucleus of infected cells, reducing their availability for activating a subset of p53-regulated genes, thereby dampening the antiviral role of p53.IMPORTANCE Host cells use a number of factors to defend against viral infection. Viruses are, however, in an arms race with their host cells to overcome these defense mechanisms. The tumor suppressor protein p53 is an important sensor of cell stress induced by oncogenic insults or viral infections, which upon activation induces various pathways to ensure the integrity of cells. Viruses have to counteract many functions of p53, but complex DNA viruses such as cytomegaloviruses may also utilize some p53 functions for their own benefit. In this study, we discovered that the M25 proteins of mouse cytomegalovirus interact with p53 and mediate its accumulation during infection. Interaction with the M25 proteins sequesters p53 molecules in nuclear dot-like structures, limiting their availability for activation of a subset of p53-regulated target genes. Understanding the interaction between viral proteins and p53 may allow to develop new therapeutic strategies against cytomegalovirus and other viruses.


Asunto(s)
Núcleo Celular/metabolismo , Infecciones por Herpesviridae/metabolismo , Muromegalovirus/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Virales/metabolismo , Animales , Núcleo Celular/genética , Núcleo Celular/virología , Células HCT116 , Células HEK293 , Infecciones por Herpesviridae/genética , Humanos , Ratones , Muromegalovirus/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Virales/genética
2.
Cell Mol Life Sci ; 74(2): 373-392, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27604867

RESUMEN

The zonula occludens (ZO)-2 protein links tight junctional transmembrane proteins to the actin cytoskeleton and associates with splicing and transcription factors in the nucleus. Multiple posttranslational modifications control the intracellular distribution of ZO-2. Here, we report that ZO-2 is a target of the SUMOylation machinery and provide evidence on how this modification may affect its cellular distribution and function. We show that ZO-2 associates with the E2 SUMO-conjugating enzyme Ubc9 and with SUMO-deconjugating proteases SENP1 and SENP3. In line with this, modification of ZO-2 by endogenous SUMO1 was detectable. Ubc9 fusion-directed SUMOylation confirmed SUMOylation of ZO-2 and was inhibited in the presence of SENP1 but not by an enzymatic-dead SENP1 protein. Moreover, lysine 730 in human ZO-2 was identified as a potential modification site. Mutation of this site to arginine resulted in prolonged nuclear localization of ZO-2 in nuclear recruitment assays. In contrast, a construct mimicking constitutive SUMOylation of ZO-2 (SUMO1ΔGG-ZO-2) was preferentially localized in the cytoplasm. Based on previous findings the differential localization of these ZO-2 constructs may affect glycogen-synthase-kinase-3ß (GSK3ß) activity and ß-catenin/TCF-4-mediated transcription. In this context we observed that ZO-2 directly binds to GSK3ß and SUMO1ΔGG-ZO-2 modulates its kinase activity. Moreover, we show that ZO-2 forms a complex with ß-catenin. Wild-type ZO-2 and ZO-2-K730R inhibited transcriptional activity in reporter gene assays, whereas the cytosolic SUMO1ΔGG-ZO-2 did not. From these data we conclude that SUMOylation affects the intracellular localization of ZO-2 and its regulatory role on GSK3ß and ß-catenin signaling activity.


Asunto(s)
Espacio Intracelular/metabolismo , Sumoilación , Proteína de la Zonula Occludens-2/metabolismo , Secuencia de Aminoácidos , Animales , Cisteína Endopeptidasas/metabolismo , Perros , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HEK293 , Humanos , Lisina/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Fosforilación , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transfección , Enzimas Ubiquitina-Conjugadoras/metabolismo , Proteína de la Zonula Occludens-2/química , beta Catenina/metabolismo
3.
Biochim Biophys Acta ; 1853(1): 136-43, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25409927

RESUMEN

The non-canonical IKK kinase TBK1 serves as an important signal transmitter of the antiviral interferon response, but is also involved in the regulation of further processes such as autophagy. The activity of TBK1 is regulated by posttranslational modifications comprising phosphorylation and ubiquitination. This study identifies SUMOylation as a novel posttranslational TBK1 modification. TBK1 kinase activity is required to allow the attachment of SUMO1 or SUMO2/3 proteins. Since TBK1 does not bind to the E2 enzyme Ubc9, this modification most likely proceeds via trans-SUMOylation. Mass spectrometry allowed identifying K694 as the SUMO acceptor site, a residue located in the C-terminal coiled-coil domain which is exclusively responsible for the association with the adaptor proteins NAP1, Sintbad and TANK. SUMO modification at K694 contributes to the antiviral function of TBK1 and accordingly the viral protein Gam1 antagonizes this posttranslational modification.


Asunto(s)
Antivirales/farmacología , Proteínas Serina-Treonina Quinasas/química , Sumoilación , Células HEK293 , Humanos , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/farmacología , Estructura Terciaria de Proteína
4.
J Cell Biochem ; 115(8): 1430-40, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24616021

RESUMEN

The NF-κB subunit RelB is known to act either as an activator or repressor of NF-κB-dependent gene expression. The RelB-p52 heterodimer, for instance, is the key element of the alternative NF-κB signaling pathway supporting the expression of a subset of NF-κB target genes. By contrast, RelB is crucial for the repression of important pro-inflammatory cytokines like TNFα or interleukin 1ß. Despite accumulating reports describing the functional variability of RelB, the molecular mechanisms underlying these divergent functions are still unknown. One potential explanation could be a functional reprogramming of RelB by different post-translational modifications. Here, we demonstrate that SUMOylation of RelB might be one of these post-translational modifications rendering the function of the NF-κB transcription factor RelB. In vivo SUMOylation analyses using either the UBC9-fusion-directed SUMOylation method or endogenous proteins from Namalwa B cells revealed that RelB is modified by either SUMO1 or SUMO2 attachment at various sites. Functional studies suggest that SUMOylation converts RelB into a transcriptional repressor. For instance, a SUMO1-RelB fusion protein mimicking RelB-SUMOylation displayed a reduced transcriptional activity in comparison to wild type RelB. Consistently, inactivation of specific SUMOylation sites in the central part of RelB augmented the transcription activity of the corresponding RelB mutant. Taken together, our data suggest that SUMOylation might be a potential molecular mechanism involved in reprogramming RelB, thus contributing to its functional diversity.


Asunto(s)
FN-kappa B/metabolismo , Procesamiento Proteico-Postraduccional/genética , Sumoilación/genética , Factor de Transcripción ReIB/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Regiones Promotoras Genéticas , Proteína SUMO-1/metabolismo , Transducción de Señal/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Transcripción Genética , Factor de Necrosis Tumoral alfa
5.
Hum Mol Genet ; 20(24): 4865-78, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21920940

RESUMEN

Spinal muscular atrophy (SMA), a frequent neurodegenerative disease, is caused by reduced levels of functional survival of motoneuron (SMN) protein. SMN is involved in multiple pathways, including RNA metabolism and splicing as well as motoneuron development and function. Here we provide evidence for a major contribution of the Rho-kinase (ROCK) pathway in SMA pathogenesis. Using an in vivo protein interaction system based on SUMOylation of proteins, we found that SMN is directly interacting with profilin2a. Profilin2a binds to a stretch of proline residues in SMN, which is heavily impaired by a novel SMN2 missense mutation (S230L) derived from a SMA patient. In different SMA models, we identified differential phosphorylation of the ROCK-downstream targets cofilin, myosin-light chain phosphatase and profilin2a. We suggest that hyper-phosphorylation of profilin2a is the molecular link between SMN and the ROCK pathway repressing neurite outgrowth in neuronal cells. Finally, we found a neuron-specific increase in the F-/G-actin ratio that further support the role of actin dynamics in SMA pathogenesis.


Asunto(s)
Atrofia Muscular Espinal/metabolismo , Profilinas/metabolismo , Transducción de Señal , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Quinasas Asociadas a rho/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Conos de Crecimiento/metabolismo , Conos de Crecimiento/patología , Humanos , Ratones , Modelos Biológicos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/patología , Proteínas Mutantes/metabolismo , Mutación Missense/genética , Neuritas/metabolismo , Fosforilación , Unión Proteica , Ratas , Proteína 1 para la Supervivencia de la Neurona Motora/genética
6.
Cell Death Discov ; 9(1): 14, 2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36658119

RESUMEN

The TNF receptor-interacting protein kinases (RIPK)-1 and 3 are regulators of extrinsic cell death response pathways, where RIPK1 makes the cell survival or death decisions by associating with distinct complexes mediating survival signaling, caspase activation or RIPK3-dependent necroptotic cell death in a context-dependent manner. Using a mass spectrometry-based screen to find new components of the ripoptosome/necrosome, we discovered the protein-arginine methyltransferase (PRMT)-5 as a direct interaction partner of RIPK1. Interestingly, RIPK3 but not RIPK1 was then found to be a target of PRMT5-mediated symmetric arginine dimethylation. A conserved arginine residue in RIPK3 (R486 in human, R415 in mouse) was identified as the evolutionarily conserved target for PRMT5-mediated symmetric dimethylation and the mutations R486A and R486K in human RIPK3 almost completely abrogated its methylation. Rescue experiments using these non-methylatable mutants of RIPK3 demonstrated PRMT5-mediated RIPK3 methylation to act as an efficient mechanism of RIPK3-mediated feedback control on RIPK1 activity and function. Therefore, this study reveals PRMT5-mediated RIPK3 methylation as a novel modulator of RIPK1-dependent signaling.

7.
Biochem J ; 438(3): 495-503, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21675959

RESUMEN

Protein-protein interactions are essential for almost all cellular processes, hence understanding these processes mainly depends on the identification and characterization of the relevant protein-protein interactions. In the present paper, we introduce the concept of TRS (trans-SUMOylation), a new method developed to identify and verify protein-protein interactions in mammalian cells in vivo. TRS utilizes Ubc9-fusion proteins that trans-SUMOylate co-expressed interacting proteins. Using TRS, we analysed interactions of 65 protein pairs co-expressed in HEK (human embryonic kidney)-293 cells. We identified seven new and confirmed 16 known protein interactions, which were determined via endogenous SUMOylation sites of the binding partners or by using SUMOylation-site tags respectively. Four of the new protein interactions were confirmed by GST (glutathione transferase) pull-down and the p38α-Edr2 interaction was verified by co-localization analysis. Functionally, this p38α-Edr2 interaction could possibly be involved in the recruitment of p38α to the polycomb chromatin-remodelling complex to phosphorylate Bmi1. We also used TRS to characterize protein-interaction domains of the protein kinase pairs p38α-MK2 [MK is MAPK (mitogen-activated protein kinase)-activated protein kinase] and ERK3 (extracellular-signal-regulated kinase 3)-MK5 and of the p38α-p53 complex. The ability of TRS to monitor protein interactions in mammalian cells in vivo at levels similar to endogenous expression makes it an excellent new tool that can help in defining the protein interactome of mammalian cells.


Asunto(s)
Mapeo de Interacción de Proteínas/métodos , Sumoilación , Animales , Sitios de Unión , Cromatina/metabolismo , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Proteína Quinasa 6 Activada por Mitógenos/genética , Proteína Quinasa 6 Activada por Mitógenos/metabolismo , Fosforilación , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Nucleic Acids Res ; 37(4): e30, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19174562

RESUMEN

Post-translational modifications control the physiological activity of the signal transducer and activator of transcription STAT1. While phosphorylation at tyrosine Y701 is a prerequisite for STAT1 dimerization, its SUMOylation represses the transcriptional activity. Recently, we have demonstrated that SUMOylation at lysine K703 inhibits the phosphorylation of nearby localized Y701 of STAT1. Here, we analysed the influence of phosphorylation of Y701 on SUMOylation of K703 in vivo. For that reason, an Ubc9/substrate dimerization-dependent SUMOylation (USDDS) system was developed, which consists of fusions of the SUMOylation substrate and of the SUMO-conjugating enzyme Ubc9 to the chemically activatable heterodimerization domains FKBP and FRB, respectively. When FKBP fusion proteins of STAT1, p53, CRSP9, FOS, CSNK2B, HES1, TCF21 and MYF6 are coexpressed with Ubc9-FRB, treatment of HEK293 cells with the rapamycin-related dimerizer compound AP21967 induces SUMOylation of these proteins in vivo. For STAT1-FKBP and p53-FKBP we show that this SUMOylation takes place at their specific SUMOylation sites in vivo. Using USDDS, we then demonstrate that STAT1 phosphorylation at Y701 induced by interferon-beta treatment inhibits SUMOylation of K703 in vivo. Thus, pY701 and SUMO-K703 of STAT1 represent mutually exclusive modifications, which prevent signal integration at this molecule and probably ensure the existence of differentially modified subpopulations of STAT1 necessary for its regulated nuclear cytoplasmic activation/inactivation cycle.


Asunto(s)
Procesamiento Proteico-Postraduccional , Factor de Transcripción STAT1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Línea Celular , Dimerización , Humanos , Factor de Transcripción STAT1/química , Sirolimus/análogos & derivados , Sirolimus/farmacología , Tirosina/metabolismo
9.
Methods Mol Biol ; 497: 63-79, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19107411

RESUMEN

The bottleneck in studying protein sumoylation-the conjugation of the small ubiquitin-like modifier (SUMO)-is the detection of the low level of in vivo sumoylated proteins. The Ubc9 fusion-directed sumoylation (UFDS) system strongly enhances the in vivo sumoylation of a substrate protein at its specific sumoylation site. UFDS utilizes an expression plasmid for the protein of interest fused to the SUMO-conjugating enzyme Ubc9. When expressed in HEK293, COS-7, HeLa, or CHO cells, the fused target protein is conjugated with endogenous or coexpressed SUMO at its native sumoylation sites. This sumoylation requires neither SUMO ligase nor any extracellular stimulation and is easily detectable by fusion protein- or Ubc9-specific Western blotting with commercially available antibodies.


Asunto(s)
Técnicas de Laboratorio Clínico , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Cricetinae , Eficiencia , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Enzimas Ubiquitina-Conjugadoras/genética
10.
Nucleic Acids Res ; 35(17): e109, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17709345

RESUMEN

Constitutive and induced protein SUMOylation is involved in the regulation of a variety of cellular processes, such as regulation of gene expression and protein transport, and proceeds mainly in the nucleus of the cell. So far, several hundred SUMOylation targets have been identified, but presumably they represent only a part of the total of proteins which are regulated by SUMOylation. Here, we used the Ubc9 fusion-dependent SUMOylation system (UFDS) to screen for constitutive and induced SUMOylation of 46 randomly chosen proteins with proven or potential nuclear localization. Fourteen new UFDS-substrate proteins were identified of which eight could be demonstrated to be SUMOylated in a UFDS-independent manner in vivo. Of these, three were constitutively SUMOylated (FOS, CRSP9 and CDC37) while the remaining five substrates (CSNK2B, TAF10, HSF2BP, PSMC3 and DRG1) showed a stimulation-dependent SUMOylation induced by the MAP3 kinase MEKK1. Hence, UFDS is appropriate for the identification and characterization of constitutive and, more importantly, induced protein SUMOylation in vivo.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Línea Celular , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética
11.
FEBS J ; 275(7): 1411-1426, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18331345

RESUMEN

Hepatoma-derived growth factor is a nuclear targeted mitogen containing a PWWP domain that mediates binding to DNA. To date, almost nothing is known about the molecular mechanisms of the functions of hepatoma-derived growth factor, its routes of secretion and internalization or post-translational modifications. In the present study, we show for the first time that hepatoma-derived growth factor is modified by the covalent attachment of small ubiquitin-related modifier 1 (SUMO-1), a post-translational modification with regulatory functions for an increasing number of proteins. Using a basal SUMOylation system in Escherichia coli followed by a MALDI-TOF-MS based peptide analysis, we identified the lysine residue SUMOylated located in the N-terminal part of the protein adjacent to the PWWP domain. Surprisingly, this lysine residue is not part of the consensus motif described for SUMOylation. With a series of hepatoma-derived growth factor mutants, we then confirmed that this unusual location is also used in mammalian cells and that SUMOylation of hepatoma-derived growth factor takes place in the nucleus. Finally, we demonstrate that SUMOylated hepatoma-derived growth factor is not binding to chromatin, in contrast to its unSUMOylated form. These observations potentially provide new perspectives for a better understanding of the functions of hepatoma-derived growth factor.


Asunto(s)
Cromatina/metabolismo , Regulación hacia Abajo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína SUMO-1/metabolismo , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células COS , Chlorocebus aethiops , Secuencia de Consenso , Regulación hacia Abajo/genética , Escherichia coli , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Datos de Secuencia Molecular , Unión Proteica , Procesamiento Proteico-Postraduccional/genética , Estructura Terciaria de Proteína , Proteína SUMO-1/fisiología
12.
Cell Signal ; 16(11): 1291-8, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15337528

RESUMEN

The p75 neurotrophin receptor (p75NTR) has dual functions in cell survival and cell death but its intracellular signalling pathways are not understood. Here we describe that in rat brain and in pervanadate-stimulated PCNA and HEK293 cells p75NTR is phosphorylated at a single tyrosine residue within the cytosolic C-terminus. Phosphorylated tyrosine 308 constitutes a binding site for the ubiquitin ligase c-Cbl. This interaction is a prerequisite for ubiquitination of p75NTR. Our data suggest a c-Cbl-dependent ubiquitination of p75NTR involved in the regulation of p75NTR signalling.


Asunto(s)
Proteínas Proto-Oncogénicas/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/fisiología , Tirosina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Secuencia de Aminoácidos/fisiología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Línea Celular , Humanos , Fosforilación , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Proteínas Proto-Oncogénicas c-cbl , Receptor de Factor de Crecimiento Nervioso , Vanadatos/farmacología
13.
J Steroid Biochem Mol Biol ; 138: 162-73, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23727127

RESUMEN

The 20-hydroxyecdysone receptor (EcR) is a transcription factor belonging to the nuclear receptor superfamily. Together with the ultraspiracle nuclear receptor (Usp) it coordinates critical biological processes in insects such as development and reproduction. EcR and its ligands are used in commercially available ecdysone-inducible expression systems and are considered to be artificial gene switches with potential therapeutic applications. However, the regulation of EcR action is still unclear, especially in mammals and as far as posttranslational modifications are concerned. Up until now, there has been no study on EcR sumoylation. Using bioinformatic predictors, a Ubc9 fusion-directed sumoylation system and mutagenesis experiments, we present EcR as a new target of SUMO1 and SUMO3 modification. Our research revealed that EcR undergoes isoform-specific multisumoylation. The pattern of modification remains unchanged in the presence of the ligand and the dimerization partner. The SUMO acceptor sites are located in the DNA-binding domain and the ligand-binding domain that both exhibit structural plasticity. We also demonstrated the existence of a sumoylation site in the F region and EcRA-A/B region, both revealing characteristics of intrinsically disordered regions. The consequences of modification and the resulting impact on conformation and function may be especially crucial for the disordered sequences in these two areas. The isoform-specificity of sumoylation may explain the differences in the transcriptional activity of EcR isoforms.


Asunto(s)
Proteínas de Drosophila/metabolismo , Receptores de Esteroides/metabolismo , Animales , Western Blotting , Línea Celular , Biología Computacional , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Modelos Biológicos , Receptores de Esteroides/química , Receptores de Esteroides/genética , Sumoilación
14.
J Steroid Biochem Mol Biol ; 132(3-5): 227-38, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22676916

RESUMEN

The ultraspiracle protein (Usp), together with an ecdysone receptor (EcR) forms a heterodimeric ecdysteroid receptor complex, which controls metamorphosis in Drosophila melanogaster. Although the ecdysteroid receptor is considered to be a source of elements for ecdysteroid inducible gene switches in mammals, nothing is known about posttranslational modifications of the receptor constituents in mammalian cells. Up until now there has been no study about Usp sumoylation. Using Ubc9 fusion-directed sumoylation system, we identified Usp as a new target of SUMO1 and SUMO3 modification. Mutagenesis studies on the fragments of Usp indicated that sumoylation can occur alternatively on several defined Lys residues, i.e. three (Lys16, Lys20, Lys37) in A/B region, one (Lys424) in E region and one (Lys506) in F region. However, sumoylation of one Lys residue within A/B region prevents modification of other residues in this region. This was also observed for Lys residues in carboxyl-terminal fragment of Usp, i.e. comprising E and F regions. Mass spectrometry analysis of the full-length Usp indicated that the main SUMO attachment site is at Lys20. EcR, the heterodimerization partner of Usp, and muristerone A, the EcR ligand, do not influence sumoylation patterns of Usp. Another heterodimerization partner of Usp - HR38 fused with Ubc9 interacts with Usp in HEK293 cells and allows sumoylation of Usp independent of the direct fusion to Ubc9. Taken together, we propose that sumoylation of DmUsp can be an important factor in modulating its activity by changing molecular interactions.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ecdisterona/análogos & derivados , Ecdisterona/metabolismo , Humanos , Ligandos , Lisina/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sumoilación , Factores de Transcripción/genética , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
15.
Mol Cell Biol ; 32(6): 1068-79, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22203040

RESUMEN

Podocytes are highly differentiated and polarized epithelial cells located on the visceral side of the glomerulus. They form an indispensable component of the glomerular filter, the slit diaphragm, formed by several transmembrane proteins and adaptor molecules. Disruption of the slit diaphragm can lead to massive proteinuria and nephrotic syndrome in mice and humans. CD2AP is an adaptor protein that is important for the maintenance of the slit diaphragm. Together with its paralogue, CIN85, CD2AP belongs to a family of adaptor proteins that are primarily described as being involved in endocytosis and downregulation of receptor tyrosine kinase activity. We have shown that full-length CIN85 is upregulated in podocytes in the absence of CD2AP, whereas in wild-type cells, full-length CIN85 is not detectable. In this study, we show that full-length CIN85 is postranslationally modified by SUMOylation in wild-type podocytes. We can demonstrate that CIN85 is SUMOylated by SUMO-1, -2, and -3 and that SUMOylation is enhanced in the presence of CD2AP. Conversion of lysine 598 to arginine completely abolishes SUMOylation and leads to increased binding of CIN85 to nephrin. Our results indicate a novel role for CD2AP in regulating posttranslational modification of CIN85.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Podocitos/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Proteínas del Citoesqueleto/genética , Eliminación de Gen , Regulación de la Expresión Génica , Humanos , Ratones , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/genética , Filogenia
17.
Nat Methods ; 4(3): 245-50, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17277783

RESUMEN

Although small ubiquitin-like modifier (SUMO) is conjugated to proteins involved in diverse cellular processes, the functional analysis of SUMOylated proteins is often hampered by low levels of specific SUMOylated proteins in the cell. Here we describe a SUMO-conjugating enzyme (Ubc9) fusion-directed SUMOylation (UFDS) system, which allows efficient and selective in vivo SUMOylation of proteins. Although SUMOylation of overexpressed p53 and STAT1 was difficult to detect in HEK293 cells, up to 40% of p53 and STAT1 were conjugated with endogenous SUMO when fused to Ubc9. We verified the specificity of UFDS using SUMOylation-site mutants and showed that the method is not dependent on SUMO ligases. Using UFDS we demonstrated that SUMOylation of STAT1 inhibits its phosphorylation at Tyr701 and discovered p53 multi-SUMOylation in vivo. We propose that UFDS will be useful for the analysis of function of SUMOylation in protein interactions, subcellular localization as well as enzymatic activity.


Asunto(s)
Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/metabolismo , Proteína SUMO-1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Línea Celular , Humanos , Riñón , Factor de Transcripción STAT1/metabolismo , Proteína SUMO-1/genética , Enzimas Ubiquitina-Conjugadoras/genética
18.
EMBO Rep ; 7(8): 838-44, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16845370

RESUMEN

The mechanism by which YopP simultaneously inhibits mitogen-activated protein kinase (MAPK) and nuclear factor-kappaB pathways has been elusive. Ectopic expression of YopP inhibits the activity and ubiquitination of a complex consisting of overexpressed TGF-beta-activated kinase 1 (TAK1) and its subunit TAK1-binding protein (TAB)1, but not of MEK kinase 1. YopP, but not the catalytically inactive mutant YopP(C172A), also suppresses basal and interleukin-1-inducible activation of endogenous TAK1, TAB1 and TAB2. YopP does not affect the interaction of TAK1, TAB1 and TAB2 but inhibits autophosphorylation of TAK1 at Thr 187 and phosphorylation of TAB1 at Ser 438. Glutathione S-transferase-tagged YopP (GST-YopP) binds to MAPK kinase (MAPKK)4 and TAB1 but not to TAK1 or TAB2 in vitro. Furthermore, YopP in synergy with a previously described negative regulatory feedback loop inhibits TAK1 by MAPKK6-p38-mediated TAB1 phosphorylation. Taken together, these data strongly suggest that YopP binds to TAB1 and directly inhibits TAK1 activity by affecting constitutive TAK1 and TAB1 ubiquitination that is required for autoactivation of TAK1.


Asunto(s)
Proteínas Bacterianas/fisiología , Interleucina-1/fisiología , Quinasas Quinasa Quinasa PAM/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas Bacterianas/genética , Western Blotting , Línea Celular , Células Cultivadas , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Interleucina-1/genética , Interleucina-1/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/fisiología , Ratones , Plásmidos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/genética , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA