Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Virol ; 92(13)2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29695423

RESUMEN

Human adenovirus (HAdV) E1B-55K is a multifunctional regulator of productive viral replication and oncogenic transformation in nonpermissive mammalian cells. These functions depend on E1B-55K's posttranslational modification with the SUMO protein and its binding to HAdV E4orf6. Both early viral proteins recruit specific host factors to form an E3 ubiquitin ligase complex that targets antiviral host substrates for proteasomal degradation. Recently, we reported that the PML-NB-associated factor Daxx represses efficient HAdV productive infection and is proteasomally degraded via a SUMO-E1B-55K-dependent, E4orf6-independent pathway, the details of which remained to be established. RNF4, a cellular SUMO-targeted ubiquitin ligase (STUbL), induces ubiquitinylation of specific SUMOylated proteins and plays an essential role during DNA repair. Here, we show that E1B-55K recruits RNF4 to the insoluble nuclear matrix fraction of the infected cell to support RNF4/Daxx association, promoting Daxx PTM and thus inhibiting this antiviral factor. Removing RNF4 from infected cells using RNA interference resulted in blocking the proper establishment of viral replication centers and significantly diminished viral gene expression. These results provide a model for how HAdV antagonize the antiviral host responses by exploiting the functional capacity of cellular STUbLs. Thus, RNF4 and its STUbL function represent a positive factor during lytic infection and a novel candidate for future therapeutic antiviral intervention strategies.IMPORTANCE Daxx is a PML-NB-associated transcription factor that was recently shown to repress efficient HAdV productive infection. To counteract this antiviral measurement during infection, Daxx is degraded via a novel pathway including viral E1B-55K and host proteasomes. This virus-mediated degradation is independent of the classical HAdV E3 ubiquitin ligase complex, which is essential during viral infection to target other host antiviral substrates. To maintain a productive viral life cycle, HAdV E1B-55K early viral protein inhibits the chromatin-remodeling factor Daxx in a SUMO-dependent manner. In addition, viral E1B-55K protein recruits the STUbL RNF4 and sequesters it into the insoluble fraction of the infected cell. E1B-55K promotes complex formation between RNF4- and E1B-55K-targeted Daxx protein, supporting Daxx posttranslational modification prior to functional inhibition. Hence, RNF4 represents a novel host factor that is beneficial for HAdV gene expression by supporting Daxx counteraction. In this regard, RNF4 and other STUbL proteins might represent novel targets for therapeutic intervention.


Asunto(s)
Proteínas E1B de Adenovirus/metabolismo , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/fisiología , Proteínas Nucleares/metabolismo , Procesamiento Proteico-Postraduccional , Proteína SUMO-1/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas E1B de Adenovirus/genética , Infecciones por Adenovirus Humanos/metabolismo , Proteínas Co-Represoras , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Cuerpos de Inclusión Intranucleares , Chaperonas Moleculares , Proteínas Nucleares/genética , Proteína SUMO-1/genética , Sumoilación , Factores de Transcripción/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Replicación Viral
2.
EMBO J ; 32(9): 1225-37, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23481255

RESUMEN

Adenosine diphosphate (ADP)-ribosylation is a post-translational protein modification implicated in the regulation of a range of cellular processes. A family of proteins that catalyse ADP-ribosylation reactions are the poly(ADP-ribose) (PAR) polymerases (PARPs). PARPs covalently attach an ADP-ribose nucleotide to target proteins and some PARP family members can subsequently add additional ADP-ribose units to generate a PAR chain. The hydrolysis of PAR chains is catalysed by PAR glycohydrolase (PARG). PARG is unable to cleave the mono(ADP-ribose) unit directly linked to the protein and although the enzymatic activity that catalyses this reaction has been detected in mammalian cell extracts, the protein(s) responsible remain unknown. Here, we report the homozygous mutation of the c6orf130 gene in patients with severe neurodegeneration, and identify C6orf130 as a PARP-interacting protein that removes mono(ADP-ribosyl)ation on glutamate amino acid residues in PARP-modified proteins. X-ray structures and biochemical analysis of C6orf130 suggest a mechanism of catalytic reversal involving a transient C6orf130 lysyl-(ADP-ribose) intermediate. Furthermore, depletion of C6orf130 protein in cells leads to proliferation and DNA repair defects. Collectively, our data suggest that C6orf130 enzymatic activity has a role in the turnover and recycling of protein ADP-ribosylation, and we have implicated the importance of this protein in supporting normal cellular function in humans.


Asunto(s)
Glicósido Hidrolasas/fisiología , Enfermedades Neurodegenerativas/enzimología , Poli Adenosina Difosfato Ribosa/fisiología , Tioléster Hidrolasas/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Células Cultivadas , Niño , Preescolar , Familia , Femenino , Glicósido Hidrolasas/genética , Células HEK293 , Células HeLa , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Enfermedades Neurodegenerativas/genética , Linaje , Poli Adenosina Difosfato Ribosa/genética , Procesamiento Proteico-Postraduccional/genética , Homología de Secuencia de Aminoácido , Tioléster Hidrolasas/genética
3.
Nat Methods ; 9(3): 245-53, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22373911

RESUMEN

Data-intensive research depends on tools that manage multidimensional, heterogeneous datasets. We built OME Remote Objects (OMERO), a software platform that enables access to and use of a wide range of biological data. OMERO uses a server-based middleware application to provide a unified interface for images, matrices and tables. OMERO's design and flexibility have enabled its use for light-microscopy, high-content-screening, electron-microscopy and even non-image-genotype data. OMERO is open-source software, available at http://openmicroscopy.org/.


Asunto(s)
Sistemas de Administración de Bases de Datos , Bases de Datos Factuales , Interpretación de Imagen Asistida por Computador/métodos , Almacenamiento y Recuperación de la Información/métodos , Modelos Biológicos , Programas Informáticos , Interfaz Usuario-Computador , Animales , Biología/métodos , Simulación por Computador , Humanos
4.
J Virol ; 88(11): 6076-92, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24623443

RESUMEN

UNLABELLED: Promyelocytic leukemia nuclear bodies (PML-NBs) are nuclear structures that accumulate intrinsic host factors to restrict viral infections. To ensure viral replication, these must be limited by expression of viral early regulatory proteins that functionally inhibit PML-NB-associated antiviral effects. To benefit from the activating capabilities of Sp100A and simultaneously limit repression by Sp100B, -C, and -HMG, adenoviruses (Ads) employ several features to selectively and individually target these isoforms. Ads induce relocalization of Sp100B, -C, and -HMG from PML-NBs prior to association with viral replication centers. In contrast, Sp100A is kept at the PML tracks that surround the newly formed viral replication centers as designated sites of active transcription. We concluded that the host restriction factors Sp100B, -C, and -HMG are potentially inactivated by active displacement from these sites, whereas Sp100A is retained to amplify Ad gene expression. Ad-dependent loss of Sp100 SUMOylation is another crucial part of the virus repertoire to counteract intrinsic immunity by circumventing Sp100 association with HP1, therefore limiting chromatin condensation. We provide evidence that Ad selectively counteracts antiviral responses and, at the same time, benefits from PML-NB-associated components which support viral gene expression by actively recruiting them to PML track-like structures. Our findings provide insights into novel strategies for manipulating transcriptional regulation to either inactivate or amplify viral gene expression. IMPORTANCE: We describe an adenoviral evasion strategy that involves isoform-specific and active manipulation of the PML-associated restriction factor Sp100. Recently, we reported that the adenoviral transactivator E1A targets PML-II to efficiently activate viral transcription. In contrast, the PML-associated proteins Daxx and ATRX are inhibited by early viral factors. We show that this concept is more intricate and significant than originally believed, since adenoviruses apparently take advantage of specific PML-NB-associated proteins and simultaneously inhibit antiviral measures to maintain the viral infectious program. Specifically, we observed Ad-induced relocalization of the Sp100 isoforms B, C, and HMG from PML-NBs juxtaposed with viral replication centers. In contrast, Sp100A is retained at Ad-induced PML tracks that surround the newly formed viral replication centers, acting as designated sites of active transcription. The host restriction factors Sp100B, -C, and -HMG are potentially inactivated by active displacement from these sites, whereas Sp100A is retained to amplify Ad gene expression.


Asunto(s)
Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/metabolismo , Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Regulación Viral de la Expresión Génica/genética , Inmunidad Innata/inmunología , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adenovirus Humanos/genética , Línea Celular , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Immunoblotting , Hibridación in Situ , Luciferasas , Proteína de la Leucemia Promielocítica , Isoformas de Proteínas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Sumoilación
6.
Biochem J ; 428(1): 55-65, 2010 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-20196770

RESUMEN

Enzymes from the PDE (phosphodiesterase) 4 cAMP-specific PDE family are crucial for the maintenance of compartmentalized cAMP responses in many cell types. Regulation of PDE activity can be achieved via post-translational modification such as phosphorylation by ERK (extracellular-signal-regulated kinase) MAPKs (mitogen-activated protein kinases) and PKA (protein kinase A). In the present paper, we report for the first time that PDE4 isoforms from the PDE4A and PDE4D subfamilies can be selectively modified by SUMO (small ubiquitin-related modifier). We have identified a single SUMO site within a consensus tetrapeptide motif, PsiKXE (where Psi represents a hydrophobic residue), which lies in the catalytic unit of these enzymes. SUMO modification of PDE4 at this site was observed upon overexpression of the SUMO E3 ligase PIASy [protein inhibitor of activated STAT (signal transducer and activator of transcription) Y] in HEK (human embryonic kidney)-293 cells and we identify PIASy as a novel binding partner for long PDE4 isoforms. Site-directed mutagenesis of the acceptor lysine residue ablated conjugation of PDE4 with SUMO, suggesting the presence of a single SUMO site in the first subdomain of the conserved PDE4 catalytic unit. This observation was supported by both cell-free in vitro SUMOylation assays and analysis of SUMOylated spot-immobilized peptide arrays. SUMO modification of long PDE4 isoforms serves to augment their activation by PKA phosphorylation and repress their inhibition by ERK phosphorylation. Following ligation of beta-adrenergic receptors, SUMOylation of PDE4 isoforms sufficiently amplified PKA-stimulated PDE4 activity to reduce markedly the PKA phosphorylation status of the beta2-adrenergic receptor. These results highlight a new means whereby cells might achieve the selective regulation of the activity of cAMP-specific PDE4 enyzmes.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína SUMO-1/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Humanos , Datos de Secuencia Molecular , Fosforilación , Conformación Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal
7.
mBio ; 11(2)2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32184235

RESUMEN

Human adenoviruses (HAdVs) have developed mechanisms to manipulate cellular antiviral measures to ensure proper DNA replication, with detailed processes far from being understood. Host cells repress incoming viral genomes through a network of transcriptional regulators that normally control cellular homeostasis. The nuclear domains involved are promyelocytic leukemia protein nuclear bodies (PML-NBs), interferon-inducible, dot-like nuclear structures and hot spots of SUMO posttranslational modification (PTM). In HAdV-infected cells, such SUMO factories are found in close proximity to newly established viral replication centers (RCs) marked by the adenoviral DNA binding protein (DBP) E2A. Here, we show that E2A is a novel target of host SUMOylation, leading to PTMs supporting E2A function in promoting productive infection. Our data show that SUMOylated E2A interacts with PML. Decreasing SUMO-E2A protein levels by generating HAdV variants mutated in the three main SUMO conjugation motifs (SCMs) led to lower numbers of viral RCs and PML-NBs, and these two structures were no longer next to each other. Our data further indicate that SUMOylated E2A binds the host transcription factor Sp100A, promoting HAdV gene expression, and represents the molecular bridge between PML tracks and adjacent viral RCs. Consequently, E2A SCM mutations repressed late viral gene expression and progeny production. These data highlight a novel mechanism used by the virus to benefit from host antiviral responses by exploiting the cellular SUMO conjugation machinery.IMPORTANCE PML nuclear bodies (PML-NBs) are implicated in general antiviral defense based on recruiting host restriction factors; however, it is not understood so far why viruses would establish viral replication centers (RCs) juxtaposed to such "antiviral" compartments. To understand this enigma, we investigate the cross talk between PML-NB components and viral RCs to find the missing link connecting both compartments to promote efficient viral replication and gene expression. Taken together, the current concept is more intricate than originally believed, since viruses apparently take advantage of several specific PML-NB-associated proteins to promote productive infection. Simultaneously, they efficiently inhibit antiviral measures to maintain the viral infectious program. Our data provide evidence that SUMOylation of the viral RC marker protein E2A represents the basis of this virus-host interface and regulates various downstream events to support HAdV productive infection. These results are the basis of our current attempts to generate and screen for specific E2A SUMOylation inhibitors to constitute novel therapeutic approaches to limit and prevent HAdV-mediated diseases and mortality of immunosuppressed patients.


Asunto(s)
Proteínas E2 de Adenovirus/metabolismo , Interacciones Huésped-Patógeno , Proteína de la Leucemia Promielocítica/metabolismo , Sumoilación , Proteínas Virales/metabolismo , Replicación Viral , Proteínas E2 de Adenovirus/genética , Adenovirus Humanos/fisiología , Línea Celular , Humanos , Mutación , Proteína de la Leucemia Promielocítica/genética , Procesamiento Proteico-Postraduccional , Proteínas Virales/genética
8.
Int J Cancer ; 125(6): 1334-42, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19507254

RESUMEN

A consistent feature of the Hodgkin and Reed-Sternberg (HRS) cells in classical Hodgkin lymphoma (cHL) is the constitutive activation of NF-kappaB transcription factors. In Epstein-Barr virus (EBV)-associated cases of cHL, expression of viral antigens most probably leads to NF-kappaB activation but for non-EBV-associated cases, the mechanism is not clear. Previous small studies have demonstrated deleterious mutations of NFKBIA, the gene encoding IkappaB alpha, in HRS cells. In the present study, we aimed to establish the frequency of NFKBIA mutation in cHL by investigating a larger series of cases and to determine whether these mutations are a characteristic feature of non-EBV-associated cHL. Single HRS cells from 20 cases of cHL were analysed by PCRs covering all 6 exons of the gene. Clonal deleterious mutations were detected in 3 cases and in 1 case both alleles of the gene were shown to harbour mutations. NFKBIA mutations were detected only in non-EBV-associated cases but the majority of these cases had wild-type NFKBIA. It remains possible that defects in genes encoding other inhibitors of NF-kappaB, such as TNFAIP3 (A20) and CYLD, are involved in the latter cases, as described for one case in this series.


Asunto(s)
Proteínas de Unión al ADN/genética , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/fisiología , Enfermedad de Hodgkin/genética , Proteínas I-kappa B/genética , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Adolescente , Adulto , Anciano , Niño , Hibridación Genómica Comparativa , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Femenino , Perfilación de la Expresión Génica , Enfermedad de Hodgkin/patología , Enfermedad de Hodgkin/virología , Humanos , Masculino , Persona de Mediana Edad , Inhibidor NF-kappaB alfa , Análisis de Secuencia por Matrices de Oligonucleótidos , Adulto Joven
9.
J Neurosci ; 27(36): 9664-9, 2007 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-17804627

RESUMEN

The cytokine ciliary neurotrophic factor (CNTF) promotes the growth of neural processes from many kinds of neurons in the developing and regenerating adult nervous system, but the intracellular signaling mechanisms mediating this important function of CNTF are poorly understood. Here, we show that CNTF activates the nuclear factor-kappaB (NF-kappaB) transcriptional system in neonatal sensory neurons and that blocking NF-kappaB-dependent transcription inhibits CNTF-promoted neurite growth. Selectively blocking NF-kappaB activation by the noncanonical pathway that requires tyrosine phosphorylation of inhibitor kappaB-alpha (IkappaB-alpha), but not by the canonical pathway that requires serine phosphorylation of IkappaB-alpha, also effectively inhibits CNTF-promoted neurite growth. CNTF treatment activates spleen tyrosine kinase (SYK) whose substrates include IkappaB-alpha. CNTF-induced SYK phosphorylation is rapidly followed by increased tyrosine phosphorylation of IkappaB-alpha, and blocking SYK activation or tyrosine phosphorylation of IkappaB-alpha prevents CNTF-induced NF-kappaB activation and CNTF-promoted neurite growth. These findings demonstrate that NF-kappaB signaling by an unusual activation mechanism is essential for the ability of CNTF to promote the growth of neural processes in the developing nervous system.


Asunto(s)
Factor Neurotrófico Ciliar/fisiología , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Neuritas/fisiología , Neuronas Aferentes/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Factor Neurotrófico Ciliar/farmacología , Activación Enzimática/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Inhibidor NF-kappaB alfa , Neuritas/efectos de los fármacos , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/ultraestructura , Ganglio Nudoso/citología , Oligonucleótidos/farmacología , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Quinasa Syk , Transcripción Genética/efectos de los fármacos , Tirosina/metabolismo
10.
Mol Biol Cell ; 16(11): 5115-26, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16120648

RESUMEN

We identify ADAR1, an RNA-editing enzyme with transient nucleolar localization, as a novel substrate for sumoylation. We show that ADAR1 colocalizes with SUMO-1 in a subnucleolar region that is distinct from the fibrillar center, the dense fibrillar component, and the granular component. Our results further show that human ADAR1 is modified by SUMO-1 on lysine residue 418. An arginine substitution of K418 abolishes SUMO-1 conjugation and although it does not interfere with ADAR1 proper localization, it stimulates the ability of the enzyme to edit RNA both in vivo and in vitro. Moreover, modification of wild-type recombinant ADAR1 by SUMO-1 reduces the editing activity of the enzyme in vitro. Taken together these data suggest a novel role for sumoylation in regulating RNA-editing activity.


Asunto(s)
Adenosina Desaminasa/metabolismo , Nucléolo Celular/metabolismo , Edición de ARN , Proteína SUMO-1/fisiología , Animales , Células COS , Chlorocebus aethiops , Células HeLa , Humanos , Lisina/metabolismo , Mutagénesis Sitio-Dirigida , Procesamiento Proteico-Postraduccional , Proteínas de Unión al ARN , Proteína SUMO-1/metabolismo
11.
Mol Cell Biol ; 22(10): 3373-88, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11971970

RESUMEN

The androgen receptor (AR) is a nuclear hormone receptor superfamily member that conveys both trans repression and ligand-dependent trans-activation function. Activation of the AR by dihydrotestosterone (DHT) regulates diverse physiological functions including secondary sexual differentiation in the male and the induction of apoptosis by the JNK kinase, MEKK1. The AR is posttranslationally modified on lysine residues by acetylation and sumoylation. The histone acetylases p300 and P/CAF directly acetylate the AR in vitro at a conserved KLKK motif. To determine the functional properties governed by AR acetylation, point mutations of the KLKK motif that abrogated acetylation were engineered and examined in vitro and in vivo. The AR acetylation site point mutants showed wild-type trans repression of NF-kappa B, AP-1, and Sp1 activity; wild-type sumoylation in vitro; wild-type ligand binding; and ligand-induced conformational changes. However, acetylation-deficient AR mutants were selectively defective in DHT-induced trans activation of androgen-responsive reporter genes and coactivation by SRC1, Ubc9, TIP60, and p300. The AR acetylation site mutant showed 10-fold increased binding of the N-CoR corepressor compared with the AR wild type in the presence of ligand. Furthermore, histone deacetylase 1 (HDAC1) bound the AR both in vivo and in cultured cells and HDAC1 binding to the AR was disengaged in a DHT-dependent manner. MEKK1 induced AR-dependent apoptosis in prostate cancer cells. The AR acetylation mutant was defective in MEKK1-induced apoptosis, suggesting that the conserved AR acetylation site contributes to a pathway governing prostate cancer cellular survival. As AR lysine residue mutations that abrogate acetylation correlate with enhanced binding of the N-CoR repressor in cultured cells, the conserved AR motif may directly or indirectly regulate ligand-dependent corepressor disengagement and, thereby, ligand-dependent trans activation.


Asunto(s)
Apoptosis/fisiología , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Androgénicos/metabolismo , Proteína SUMO-1/metabolismo , Activación Transcripcional , Acetilación , Secuencias de Aminoácidos , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dihidrotestosterona/farmacología , Inhibidores Enzimáticos/metabolismo , Genes Reporteros , Histona Desacetilasa 1 , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/metabolismo , Técnicas In Vitro , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutación Puntual , Receptores Androgénicos/genética , Proteína SUMO-1/genética , Proteína smad3 , Ligando Inductor de Apoptosis Relacionado con TNF , Transactivadores/genética , Transactivadores/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
12.
Cell Cycle ; 2(6): 528-30, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14504467

RESUMEN

SUMO modification of proteins is being increasingly linked with transcriptional repression. We recently demonstrated that SUMO modification also downregulates the transcriptional activity of the ETS-domain transcription factor Elk-1. However, as Elk-1 becomes activated through MAP kinase-mediated phosphorylation, the SUMO modification is lost, providing an elegant molecular switch that promotes the loss of repressive activities at the same time as permitting the recruitment of coactivator proteins. However, the mechanism by which SUMO promotes transcriptional repression remains enigmatic.


Asunto(s)
Proteínas de Unión al ADN , Regulación de la Expresión Génica , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína SUMO-1/metabolismo , Factores de Transcripción , Transcripción Genética , Animales , Humanos , Proteínas Proto-Oncogénicas/metabolismo , Proteína Elk-1 con Dominio ets
13.
PLoS One ; 7(12): e51672, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23284737

RESUMEN

The NF-κB pathway is regulated by SUMOylation at least at three levels: the inhibitory molecule IκBα, the IKK subunit γ/NEMO and the p52 precursor p100. Here we investigate the role of SUMO-2/3 in the degradation of IκBα and activation of NF-κB mediated by TNFα. We found that under conditions of deficient SUMOylation, an important delay in both TNFα-mediated proteolysis of IκBα and NF-κB dependent transcription occurs. In vitro and ex vivo approaches, including the use of ubiquitin-traps (TUBEs), revealed the formation of chains on IκBα containing SUMO-2/3 and ubiquitin after TNFα stimulation. The integration of SUMO-2/3 appears to promote the formation of ubiquitin chains on IκBα after activation of the TNFα signalling pathway. Furthermore, heterologous chains of SUMO-2/3 and ubiquitin promote a more efficient degradation of IκBα by the 26S proteasome in vitro compared to chains of either SUMO-2/3 or ubiquitin alone. Consistently, Ubc9 silencing reduced the capture of IκBα modified with SUMO-ubiquitin hybrid chains that display a defective proteasome-mediated degradation. Thus, hybrid SUMO-2/3-ubiquitin chains increase the susceptibility of modified IκBα to the action of 26S proteasome, contributing to the optimal control of NF-κB activity after TNFα-stimulation.


Asunto(s)
Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Citoplasma/metabolismo , Células HeLa , Humanos , Inhibidor NF-kappaB alfa , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Transducción de Señal/efectos de los fármacos , Sumoilación , Factor de Necrosis Tumoral alfa/farmacología
14.
Mol Biol Cell ; 21(9): 1462-9, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20237157

RESUMEN

Key components of the miRNA-mediated gene regulation pathway are localized in cytoplasmic processing bodies (P-bodies). Mounting evidence suggests that the presence of microscopic P-bodies are not always required for miRNA-mediated gene regulation. Here we have shown that geldanamycin, a well-characterized HSP90 inhibitor, abolishes P-bodies and significantly reduces Argonaute and GW182 protein levels but does not affect the miRNA level and the efficiency of miRNA-mediated gene repression; however, it significantly impairs siRNA loading and the efficacy of exogenous siRNA. Our data suggests that HSP90 protein chaperones Argonautes before binding RNA and may facilitate efficient loading of small RNA.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas Argonautas , Autoantígenos/genética , Autoantígenos/metabolismo , Benzoquinonas/farmacología , Northern Blotting , Western Blotting , Línea Celular , Línea Celular Tumoral , Gránulos Citoplasmáticos/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/genética , Expresión Génica , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Células HeLa , Humanos , Lactamas Macrocíclicas/farmacología , Luciferasas/genética , Luciferasas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Microscopía Fluorescente , Unión Proteica , Interferencia de ARN , Proteínas de Unión al ARN , Transducción de Señal/efectos de los fármacos , Transfección
15.
J Biol Chem ; 280(11): 9937-45, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15637059

RESUMEN

The TFIID complex is composed of the TATA-binding protein (TBP) and TBP-associated factors (TAFs) and is the only component of the general RNA polymerase II (RNAP II) transcription machinery with intrinsic sequence-specific DNA-binding activity. Binding of transcription factor (TF) IID to the core promoter region of protein-coding genes is a key event in RNAP II transcription activation and is the first and rate-limiting step of transcription initiation complex assembly. Intense research efforts in the past have established that TFIID promoter-binding activity as well as the function of TFIID-promoter complexes is tightly regulated through dynamic TFIID interactions with positive- and negative-acting transcription regulatory proteins. However, very little is known about the role of post-translational modifications in the regulation of TFIID. Here we show that the human TFIID subunits hsTAF5 and hsTAF12 are modified by the small ubiquitin-related modifier SUMO-1 in vitro and in human cells. We identify Lys-14 in hsTAF5 and Lys-19 in hsTAF12 as the primary SUMO-1 acceptor sites and show that SUMO conjugation has no detectable effect on nuclear import or intranuclear distribution of hsTAF5 and hsTAF12. Finally, we demonstrate that purified human TFIID complex can be SUMO-1-modified in vitro at both hsTAF5 and hsTAF12. We find that SUMO-1 conjugation at hsTAF5 interferes with binding of TFIID to promoter DNA, whereas modification of hsTAF12 has no detectable effect on TFIID promoter-binding activity. Our observations suggest that reversible SUMO modification at hsTAF5 contributes to the dynamic regulation of TFIID promoter-binding activity in human cells.


Asunto(s)
Proteína SUMO-1/metabolismo , Factor de Transcripción TFIID/química , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , ADN/química , Electroforesis en Gel de Poliacrilamida , Epítopos/química , Células HeLa , Humanos , Immunoblotting , Lisina/química , Modelos Biológicos , Modelos Genéticos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , ARN Polimerasa II/química , Proteínas Recombinantes/química , Homología de Secuencia de Aminoácido , Factores Asociados con la Proteína de Unión a TATA/química , Transcripción Genética , Transfección
16.
J Biol Chem ; 280(26): 24330-8, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15857832

RESUMEN

A variety of transcription factors are post-translationally modified by SUMO, a 97-residue ubiquitin-like protein bound covalently to the targeted lysine. Here we describe SUMO modification of the Ets family member ERM at positions 89, 263, 293, and 350. To investigate how SUMO modification affects the function of ERM, Ets-responsive intercellular adhesion molecule 1 (ICAM-1) and E74 reporter plasmids were employed to demonstrate that SUMO modification causes inhibition of ERM-dependent transcription without affecting the subcellular localization, stability, or DNA-binding capacity of the protein. When the adenoviral protein Gam1 or the SUMO protease SENP1 was used to inhibit the SUMO modification pathway, ERM-dependent transcription was de-repressed. These results demonstrate that ERM is subject to SUMO modification and that this post-translational modification causes inhibition of transcription-enhancing activity.


Asunto(s)
Proteínas de Unión al ADN/química , Proteína SUMO-1/fisiología , Factores de Transcripción/química , Transcripción Genética , Adenoviridae/genética , Animales , Arginina/química , Sitios de Unión , Western Blotting , Células COS , Cisteína Endopeptidasas , ADN/química , ADN Complementario/metabolismo , Proteínas de Unión al ADN/genética , Endopeptidasas/metabolismo , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Biblioteca de Genes , Genes Reporteros , Células HeLa , Humanos , Inmunoprecipitación , Molécula 1 de Adhesión Intercelular/metabolismo , Lisina/química , Microscopía Fluorescente , Modelos Genéticos , Plásmidos/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Conejos , Proteína SUMO-1/metabolismo , Factores de Transcripción/genética , Activación Transcripcional , Técnicas del Sistema de Dos Híbridos , Ubiquitina/química
17.
Mol Cell ; 12(1): 63-74, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12887893

RESUMEN

The ETS domain transcription factor Elk-1 is a direct target of the MAP kinase pathways. Phosphorylation of the Elk-1 transcriptional activation domain by MAP kinases triggers its activation. However, Elk-1 also contains two domains with repressive activities. One of these, the R motif, appears to function by suppressing the activity of the activation domain. Here, we demonstrate that SUMO modification of the R motif is required for this repressive activity. A dynamic interplay exists between the activating ERK MAP kinase pathway and the repressive SUMO pathway. ERK pathway activation leads to both phosphorylation of Elk-1 and loss of SUMO conjugation and, hence, to the loss of the repressive activity of the R motif. Thus, the reciprocal regulation of the activation and repressive activities are coupled by MAP kinase modification of Elk-1.


Asunto(s)
Proteínas de Unión al ADN , Células Eucariotas/enzimología , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Proteína SUMO-1/genética , Factores de Transcripción , Transcripción Genética/genética , Animales , Línea Celular , Genes Reguladores/genética , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estructura Terciaria de Proteína/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Proteína SUMO-1/metabolismo , Proteína Elk-1 con Dominio ets
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA