Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cytokine ; 120: 264-272, 2019 08.
Article in English | MEDLINE | ID: mdl-31153006

ABSTRACT

ProMyelocytic Leukemia (PML) protein is essential for the formation of nuclear matrix-associated organelles named PML nuclear bodies (NBs) that act as a platform for post-translational modifications and protein degradation. PML NBs harbor transiently and permanently localized proteins and are associated with the regulation of several cellular functions including apoptosis. There are seven PML isoforms, six nuclear (PMLI-VI) and one cytoplasmic (PMLVII), which are encoded by a single gene via alternative RNA splicing. It has been reported that murine PML-null primary cells are resistant to TGF-ß-induced apoptosis and that cytoplasmic PML is an essential activator of TGF-ß signaling. The role and the fate of interferon (IFN)-enhanced PML NBs in response to TGF-ß have not been investigated. Here we show that IFNα potentiated TGF-ß-mediated apoptosis in human cells. IFNα or ectopic expression of PMLIV, but not of PMLIII, enhanced TGF-ß-induced caspase 8 activation. In response to TGF-ß, both PMLIII and PMLIV were conjugated to SUMO and shifted from the nucleoplasm to the nuclear matrix, however only PMLIV, via its specific C-terminal region, interacted with caspase 8 and recruited it within PML NBs. This process was followed by a caspase-dependent PML degradation and PML NB disruption. Taken together, these findings highlight the role of PML NBs in the enhancement by IFN of TGF-ß-induced apoptosis and caspase 8 activation.


Subject(s)
Cell Nucleus/metabolism , Promyelocytic Leukemia Protein/metabolism , Proteolysis , Sumoylation , Transforming Growth Factor beta/pharmacology , Apoptosis/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Enzyme Activation/drug effects , HEK293 Cells , Humans , Interferon-alpha/pharmacology , Nuclear Matrix/metabolism , Promyelocytic Leukemia Protein/chemistry , Protein Binding/drug effects , Proteolysis/drug effects , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation/drug effects
3.
Sci Rep ; 8(1): 1277, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352251

ABSTRACT

Double-stranded RNA (dsRNA)-dependent protein kinase (PKR) is a serine/threonine kinase that exerts its own phosphorylation and the phosphorylation of the α subunit of the protein synthesis initiation factor eIF-2α. PKR was identified as a target of SUMOylation and the triple PKR-SUMO deficient mutant on Lysine residues K60-K150-K440 has reduced PKR activity. We report that SUMO1 and SUMO3 expression exert differential effects on PKR localization, activation and stability. SUMO1 or SUMO3 did not alter the repartition of PKR in the cytoplasm and the nucleus. However, in SUMO3-expressing cells PKR was found more concentrated around the perinuclear membrane and was recruited from small speckles to nuclear dots. Interestingly, SUMO1 expression alone resulted in PKR and eIF-2α activation, whereas SUMO3 reduced PKR and eIF-2α activation upon viral infection or dsRNA transfection. In addition, encephalomyocarditis virus (EMCV) enhanced PKR conjugation to SUMO1 and SUMO3 but only SUMO3 expression promoted caspase-dependent EMCV-induced PKR degradation. Furthermore, the higher EMCV-induced PKR activation by SUMO1 was correlated with an inhibition of EMCV. Importantly SUMO1, by inducing PKR activation in the absence of viral infection, and SUMO3, by counteracting both PKR activation and stability upon viral infection, shed a new light on the differential effects of SUMO-modified PKR.


Subject(s)
SUMO-1 Protein/metabolism , Ubiquitins/metabolism , eIF-2 Kinase/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Enzyme Stability , HeLa Cells , Humans , Phosphorylation , SUMO-1 Protein/genetics , Sumoylation , Ubiquitins/genetics
5.
J Immunol ; 195(5): 2312-24, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26223657

ABSTRACT

IFNs orchestrate immune defense through induction of hundreds of genes. Small ubiquitin-like modifier (SUMO) is involved in various cellular functions, but little is known about its role in IFN responses. Prior work identified STAT1 SUMOylation as an important mode of regulation of IFN-γ signaling. In this study, we investigated the roles of SUMO in IFN signaling, gene expression, protein stability, and IFN-induced biological responses. We first show that SUMO overexpression leads to STAT1 SUMOylation and to a decrease in IFN-induced STAT1 phosphorylation. Interestingly, IFNs exert a negative retrocontrol on their own signaling by enhancing STAT1 SUMOylation. Furthermore, we show that expression of each SUMO paralog inhibits IFN-γ-induced transcription without affecting that of IFN-α. Further, we focused on IFN-induced gene products associated to promyelocytic leukemia (PML) nuclear bodies, and we show that neither IFN-α nor IFN-γ could increase PML and Sp100 protein expression because they enhanced their SUMO3 conjugation and subsequent proteasomal degradation. Because it is known that SUMO3 is important for the recruitment of RING finger protein 4, a poly-SUMO-dependent E3 ubiquitin ligase, and that PML acts as a positive regulator of IFN-induced STAT1 phosphorylation, we went on to show that RING finger protein 4 depletion stabilizes PML and is correlated with a positive regulation of IFN signaling. Importantly, inhibition of IFN signaling by SUMO is associated with a reduction of IFN-induced apoptosis, cell growth inhibition, antiviral defense, and chemotaxis. Conversely, inhibition of SUMOylation results in higher IFN-γ-induced STAT1 phosphorylation and biological responses. Altogether, our results uncover a new role for SUMO in the modulation of IFN response.


Subject(s)
Interferons/pharmacology , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitins/metabolism , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Blotting, Western , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression/drug effects , HeLa Cells , Hep G2 Cells , Humans , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Microscopy, Confocal , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Promyelocytic Leukemia Protein , Protein Binding/drug effects , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , SUMO-1 Protein/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitins/genetics
6.
PLoS One ; 7(9): e44949, 2012.
Article in English | MEDLINE | ID: mdl-23028697

ABSTRACT

PML, the organizer of nuclear bodies (NBs), is expressed in several isoforms designated PMLI to VII which differ in their C-terminal region due to alternative splicing of a single gene. This variability is important for the function of the different PML isoforms. PML NB formation requires the covalent linkage of SUMO to PML. Arsenic trioxide (As2O3) enhances PML SUMOylation leading to an increase in PML NB size and promotes its interaction with RNF4, a poly-SUMO-dependent ubiquitin E3 ligase responsible for proteasome-mediated PML degradation. Furthermore, the presence of a bona fide SUMO Interacting Motif (SIM) within the C-terminal region of PML seems to be required for recruitment of other SUMOylated proteins within PML NBs. This motif is present in all PML isoforms, except in the nuclear PMLVI and in the cytoplasmic PMLVII. Using a bioluminescence resonance energy transfer (BRET) assay in living cells, we found that As2O3 enhanced the SUMOylation and interaction with RNF4 of nuclear PML isoforms (I to VI). In addition, among the nuclear PML isoforms, only the one lacking the SIM sequence, PMLVI, was resistant to As2O3-induced PML degradation. Similarly, mutation of the SIM in PMLIII abrogated its sensitivity to As2O3-induced degradation. PMLVI and PMLIII-SIM mutant still interacted with RNF4. However, their resistance to the degradation process was due to their inability to be polyubiquitinated and to recruit efficiently the 20S core and the ß regulatory subunit of the 11S complex of the proteasome in PML NBs. Such resistance of PMLVI to As2O3-induced degradation was alleviated by overexpression of RNF4. Our results demonstrate that the SIM of PML is dispensable for PML SUMOylation and interaction with RNF4 but is required for efficient PML ubiquitination, recruitment of proteasome components within NBs and proteasome-dependent degradation of PML in response to As2O3.


Subject(s)
Arsenicals/pharmacology , Cell Nucleus/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Oxides/pharmacology , Proteolysis/drug effects , SUMO-1 Protein/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Amino Acid Motifs , Animals , Arsenic Trioxide , Cell Line, Tumor , Cell Nucleus/drug effects , Cytoplasm/drug effects , Cytoplasm/metabolism , Humans , Mice , Mutation , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sumoylation/drug effects , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
7.
Biochimie ; 93(3): 389-98, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21115099

ABSTRACT

PML, also known as TRIM19, belongs to the family encoding a characteristic RBCC/TRIM motif comprising several cysteine-rich zinc-binding domains (RING and B-boxes) and a coiled-coil domain. The RBCC domain and the covalent modification of PML by the small ubiquitin-like modifier (SUMO) are required for PML localization within the nuclear bodies (NBs). Analysis of PML(-/-) mice provided evidence for a physiological role of PML in apoptosis. Cells derived from these mice are defective in the induction of apoptosis by interferon (IFN). PML is expressed as a family of cytoplasmic and nuclear isoforms (PML I-VII) as a result of alternative splicing. Herein, we show that overexpression of all nuclear PML isoforms (I-VI) in human cells increased IFNγ-induced STAT1 phosphorylation, resulting in higher binding of STAT1 to DNA, higher activation of IFN-stimulated genes (ISGs), and an increase in the expression of their products. These effects, observed with IFNγ and not IFNα, required PML localization in the nucleus as they were not observed with the cytoplasmic isoform PMLVIIb or the cytoplasmic variants of PMLIV. They also necessitated PML SUMOylation and its RING finger domain. Conversely, downregulation of PML by RNA interference was accompanied by decrease in IFNγ-induced STAT1 phosphorylation, STAT1 DNA binding, transcription of ISGs and in the expression of their products. In addition, IFNγ-mediated STAT1 DNA-binding activity was decreased in PML(-/-) MEFs compared with wild-type MEFs. Taken together these results demonstrate that PML functions as a positive regulator of IFNγ signaling.


Subject(s)
Interferon-gamma/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , DNA/metabolism , Gene Knockdown Techniques , Humans , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/genetics , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phosphorylation/genetics , Promyelocytic Leukemia Protein , Protein Isoforms/chemistry , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , RING Finger Domains , RNA Interference , STAT1 Transcription Factor/chemistry , STAT1 Transcription Factor/metabolism , Signal Transduction/genetics , Sumoylation/genetics , Transcription Factors/chemistry , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tyrosine
8.
J Virol ; 84(22): 11634-45, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20826694

ABSTRACT

The promyelocytic leukemia (PML) protein is expressed in the diffuse nuclear fraction of the nucleoplasm and in matrix-associated structures, known as nuclear bodies (NBs). PML NB formation requires the covalent modification of PML to SUMO. The noncovalent interactions of SUMO with PML based on the identification of a SUMO-interacting motif within PML seem to be required for further recruitment within PML NBs of SUMOylated proteins. RNA viruses whose replication takes place in the cytoplasm and is inhibited by PML have developed various strategies to counteract the antiviral defense mediated by PML NBs. We show here that primary fibroblasts derived from PML knockout mice are more sensitive to infection with encephalomyocarditis virus (EMCV), suggesting that the absence of PML results in an increase in EMCV replication. Also, we found that EMCV induces a decrease in PML protein levels both in interferon-treated cells and in PMLIII-expressing cells. Reduction of PML was carried out by the EMCV 3C protease. Indeed, at early times postinfection, EMCV induced PML transfer from the nucleoplasm to the nuclear matrix and PML conjugation to SUMO-1, SUMO-2, and SUMO-3, leading to an increase in PML body size where the viral protease 3C and the proteasome component were found colocalizing with PML within the NBs. This process was followed by PML degradation occurring in a proteasome- and SUMO-dependent manner and did not involve the SUMO-interacting motif of PML. Together, these findings reveal a new mechanism evolved by EMCV to antagonize the PML pathway in the interferon-induced antiviral defense.


Subject(s)
Cardiovirus Infections/metabolism , Encephalomyocarditis virus/physiology , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , CHO Cells , Cardiovirus Infections/virology , Cell Line , Cell Nucleus/metabolism , Cricetinae , Cricetulus , Humans , Intranuclear Inclusion Bodies/metabolism , Mice , Mice, Knockout , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Transport , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
9.
J Virol ; 84(20): 10719-26, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20702643

ABSTRACT

Various reports implicate PML and PML nuclear bodies (NBs) in an intrinsic antiviral response targeting diverse cytoplasmic replicating RNA viruses. PML conjugation to the small ubiquitin-like modifier (SUMO) is required for its localization within NBs. PML displays antiviral effects in vivo, as PML deficiency renders mice more susceptible to infection with the rhabdovirus vesicular stomatitis virus (VSV). Cells derived from these mice are also more sensitive to infection with rabies virus, another member of the rhabdovirus family. Alternative splicing from a single gene results in the synthesis of several PML isoforms, and these are classified into seven groups, designated PMLI to -VII. We report here that expression of PMLIV or PMLIVa, which is missing exon 5, inhibited viral mRNA and protein synthesis, leading to a reduction in viral replication. However, the expression of other nuclear isoforms (PMLI to -VI) and cytoplasmic PMLVIIb failed to impair viral production. This antiviral effect required PMLIV SUMOylation, as it was not observed with PMLIV 3KR, in which the lysines involved in SUMO conjugation were mutated. Thus, PMLIV and PMLIVa may exert this isoform-specific function through interaction with specific NB protein partners via their common C-terminal region.


Subject(s)
Nuclear Proteins/immunology , Rabies virus/pathogenicity , Rabies/immunology , Rabies/prevention & control , Transcription Factors/immunology , Tumor Suppressor Proteins/immunology , Animals , Base Sequence , Cells, Cultured , DNA Primers/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Interferon Type I/pharmacology , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Viral/biosynthesis , RNA, Viral/genetics , Rabies/genetics , Rabies virus/genetics , Rabies virus/immunology , Rabies virus/physiology , Recombinant Proteins , Sumoylation , Transcription Factors/deficiency , Transcription Factors/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Virus Replication
10.
J Biol Chem ; 284(24): 16595-16608, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19380586

ABSTRACT

Promyelocytic leukemia protein (PML) is a tumor suppressor acting as the organizer of subnuclear structures called PML nuclear bodies (NBs). Both covalent modification of PML by the small ubiquitin-like modifier (SUMO) and non-covalent binding of SUMO to the PML SUMO binding domain (SBD) are necessary for PML NB formation and maturation. PML sumoylation and proteasome-dependent degradation induced by the E3 ubiquitin ligase, RNF4, are enhanced by the acute promyelocytic leukemia therapeutic agent, arsenic trioxide (As2O3). Here, we established a novel bioluminescence resonance energy transfer (BRET) assay to dissect and monitor PML/SUMO interactions dynamically in living cells upon addition of therapeutic agents. Using this sensitive and quantitative SUMO BRET assay that distinguishes PML sumoylation from SBD-mediated PML/SUMO non-covalent interactions, we probed the respective roles of covalent and non-covalent PML/SUMO interactions in PML degradation and interaction with RNF4. We found that, although dispensable for As2O3-enhanced PML sumoylation and RNF4 interaction, PML SBD core sequence was required for As2O3- and RNF4-induced PML degradation. As confirmed with a phosphomimetic mutant, phosphorylation of a stretch of serine residues, contained within PML SBD was needed for PML interaction with SUMO-modified protein partners and thus for NB maturation. However, mutation of these serine residues did not impair As2O3- and RNF4-induced PML degradation, contrasting with the known role of these phosphoserine residues for casein kinase 2-promoted PML degradation. Altogether, these data suggest a model whereby sumoylation- and SBD-dependent PML oligomerization within NBs is sufficient for RNF4-mediated PML degradation and does not require the phosphorylation-dependent association of PML with other sumoylated partners.


Subject(s)
Leukemia, Promyelocytic, Acute/metabolism , Nuclear Proteins/metabolism , SUMO-1 Protein/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents/pharmacology , Arsenic Trioxide , Arsenicals/pharmacology , Bacterial Proteins/genetics , Cell Line , Fluorescence Resonance Energy Transfer/methods , Humans , In Vitro Techniques , Kidney/cytology , Leukemia, Promyelocytic, Acute/pathology , Luciferases, Renilla/genetics , Luminescent Measurements , Luminescent Proteins/genetics , Mutagenesis, Site-Directed , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Oxides/pharmacology , Promyelocytic Leukemia Protein , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Tertiary , SUMO-1 Protein/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...