Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cytokine Growth Factor Rev ; 55: 37-47, 2020 10.
Article in English | MEDLINE | ID: mdl-32591223

ABSTRACT

SUMOylation is a reversible post-translational modification that regulates several cellular processes including protein stability, subcellular localization, protein-protein interactions and plays a key role in the interferon (IFN) pathway and antiviral defense. In human, three ubiquitously expressed SUMO paralogs (SUMO1, 2 and 3) have been described for their implication in both intrinsic and innate immunity. Differential effects between SUMO paralogs are emerging such as their distinctive regulations of IFN synthesis, of IFN signaling and of the expression and function of IFN-stimulated gene (ISG) products. Several restriction factors are conjugated to SUMO and their modifications are further enhanced in response to IFN. Also, IFN itself was shown to increase global cellular SUMOylation and requires the presence of the E3 SUMO ligase PML that coordinates the assembly of PML nuclear bodies. This review focuses on differential effects of SUMO paralogs on IFN signaling and the stabilization/destabilization of ISG products, highlighting the crosstalk between SUMOylation and other post-translational modifications such as ubiquitination and ISGylation.


Subject(s)
Interferons , Sumoylation , Antiviral Agents , Humans , Promyelocytic Leukemia Protein/metabolism , SUMO-1 Protein
2.
Cytokine ; 129: 155025, 2020 05.
Article in English | MEDLINE | ID: mdl-32044670

ABSTRACT

Interferon (IFN) plays a central role in regulating host immune response to viral pathogens through the induction of IFN-Stimulated Genes (ISGs). IFN also enhances cellular SUMOylation and ISGylation, though the functional interplay between these modifications remains unclear. Here, we used a system-level approach to profile global changes in protein abundance in SUMO3-expressing cells stimulated by IFNα. These analyses revealed the stabilization of several ISG factors including SAMHD1, MxB, GBP1, GBP5, Tetherin/BST2 and members of IFITM, IFIT and IFI families. This process was correlated with enhanced IFNα-induced anti-HIV-1 and HSV-1 activities. Also IFNα upregulated protein ISGylation through increased abundance of E2 conjugating enzyme UBE2L6, and E3 ISG15 ligases TRIM25 and HERC5. Remarkably, TRIM25 depletion blocked SUMO3-dependent protein stabilization in response to IFNα. Our data identify a new mechanism by which SUMO3 regulates ISG product stability and reinforces the relevance of the SUMO pathway in controlling both the expression and functions of the restriction factors and IFN antiviral response.


Subject(s)
Interferon-alpha/pharmacology , Sumoylation/drug effects , Antiviral Agents/pharmacology , Cell Line , Cell Line, Tumor , Gene Expression/drug effects , HEK293 Cells , HeLa Cells , Humans , Signal Transduction/drug effects , Transcription Factors/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism
3.
Med Sci (Paris) ; 36(1): 50-56, 2020 Jan.
Article in French | MEDLINE | ID: mdl-32014098

ABSTRACT

PML/TRIM19 is the organizer of PML nuclear bodies (NB), a multiprotein complex associated to the nuclear matrix, which recruit a large number of proteins involved in various cellular processes. Alternative splicing from a single PML gene generates 6 nuclear PML isoforms (PMLI to PMLVI) and one cytoplasmic isoform, PMLVII. Murine PML-null primary cells are resistant to TGF-ß-induced apoptosis. Cytoplasmic PML is an essential activator of TGF-ß signaling by increasing the phosphorylation of transcription factors SMAD2/3 while nuclear PML plays a role in TGF-ß-induced caspase 8 activation and apoptosis. TGF-ß targets nuclear PML by inducing its conjugation to SUMO. In the nucleus, PML is mainly expressed in the nucleoplasm with a small fraction in the nuclear matrix. In response to TGF-ß, PML and caspase 8 shift to the nuclear matrix, where both PML and caspase 8 colocalise within PML NBs. Here, we review the implication of cytoplasmic and nuclear PML isoforms in TGF-ß response.


TITLE: Les isoformes de PML et la réponse au TGF-ß. ABSTRACT: PML (promyelocytic leukemia) est la protéine organisatrice des corps nucléaires, une structure multiprotéique associée à la matrice nucléaire, impliquée dans différents processus cellulaires. Sept isoformes principales de PML, dont six nucléaires (PMLI à VI) et une cytoplasmique (PMLVII), sont générées par épissage alternatif d'un gène unique. D'une part, PML dans le cytoplasme régule positivement le signal de transduction donné par le TGF-ß, en augmentant la phosphorylation des facteurs de transcription SMAD2/3 et, d'autre part, PML augmente dans le noyau l'activation de la caspase 8 et l'apoptose en réponse au TGF-ß. L'absence de PML rend les cellules résistantes à l'apoptose induite par le TGF-ß. Dans le noyau, PML est localisée majoritairement dans le nucléoplasme, une petite fraction étant cependant retrouvée dans la matrice nucléaire. Le TGF-ß cible PML dans le noyau en induisant sa conjugaison à SUMO (small ubiquitin modifier), son transfert et celui de la caspase 8 vers la matrice nucléaire où les deux protéines se localisent au sein des corps nucléaires PML. Cette revue rend compte des implications de PML dans le cytoplasme et le noyau dans la réponse au TGF-ß.


Subject(s)
Promyelocytic Leukemia Protein/physiology , Transforming Growth Factor beta/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Humans , Mice , Promyelocytic Leukemia Protein/genetics , Protein Isoforms/genetics , Protein Isoforms/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
4.
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
5.
Viruses ; 10(12)2018 12 03.
Article in English | MEDLINE | ID: mdl-30513968

ABSTRACT

Small Ubiquitin-like MOdifier (SUMO) conjugation to proteins has essential roles in several processes including localization, stability, and function of several players implicated in intrinsic and innate immunity. In human, five paralogs of SUMO are known of which three are ubiquitously expressed (SUMO1, 2, and 3). Infection by rhabdoviruses triggers cellular responses through the activation of pattern recognition receptors, which leads to the production and secretion of interferon. This review will focus on the effects of the stable expression of the different SUMO paralogs or Ubc9 depletion on rhabdoviruses-induced interferon production and interferon signaling pathways as well as on the expression and functions of restriction factors conferring the resistance to rhabdoviruses.


Subject(s)
Rhabdoviridae Infections/immunology , Rhabdoviridae/immunology , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Humans , Immunity, Innate , Interferons/immunology , Mice , Myxovirus Resistance Proteins/genetics , Protein Binding , Rabies virus/immunology , Receptors, Pattern Recognition/immunology , Small Ubiquitin-Related Modifier Proteins/immunology , Sumoylation , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Vesicular stomatitis Indiana virus/immunology , eIF-2 Kinase/genetics
7.
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
8.
J Virol ; 90(14): 6598-6610, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27170750

ABSTRACT

UNLABELLED: Multiple cellular pathways are regulated by small ubiquitin-like modifier (SUMO) modification, including ubiquitin-mediated proteolysis, signal transduction, innate immunity, and antiviral defense. In the study described in this report, we investigated the effects of SUMO on the replication of two members of the Rhabdoviridae family, vesicular stomatitis virus (VSV) and rabies virus (RABV). We show that stable expression of SUMO in human cells confers resistance to VSV infection in an interferon-independent manner. We demonstrate that SUMO expression did not alter VSV entry but blocked primary mRNA synthesis, leading to a reduction of viral protein synthesis and viral production, thus protecting cells from VSV-induced cell lysis. MxA is known to inhibit VSV primary transcription. Interestingly, we found that the MxA protein was highly stabilized in SUMO-expressing cells. Furthermore, extracts from cells stably expressing SUMO exhibited an increase in MxA oligomers, suggesting that SUMO plays a role in protecting MxA from degradation, thus providing a stable intracellular pool of MxA available to combat invading viruses. Importantly, MxA depletion in SUMO-expressing cells abrogated the anti-VSV effect of SUMO. Furthermore, SUMO expression resulted in interferon-regulatory factor 3 (IRF3) SUMOylation, subsequently decreasing RABV-induced IRF3 phosphorylation and interferon synthesis. As expected, this rendered SUMO-expressing cells more sensitive to RABV infection, even though MxA was stabilized in SUMO-expressing cells, since its expression did not confer resistance to RABV. Our findings demonstrate opposing effects of SUMO expression on two viruses of the same family, intrinsically inhibiting VSV infection through MxA stabilization while enhancing RABV infection by decreasing IFN induction. IMPORTANCE: We report that SUMO expression reduces interferon synthesis upon RABV or VSV infection. Therefore, SUMO renders cells more sensitive to RABV but unexpectedly renders cells resistant to VSV by blocking primary mRNA synthesis. Unlike the interferon-mediated innate immune response, intrinsic antiviral resistance is mediated by constitutively expressed restriction factors. Among the various anti-VSV restriction factors, only MxA is known to inhibit VSV primary transcription, and we show here that its expression does not alter RABV infection. Interestingly, MxA depletion abolished the inhibition of VSV by SUMO, demonstrating that MxA mediates SUMO-induced intrinsic VSV resistance. Furthermore, MxA oligomerization is known to be critical for its protein stability, and we show that higher levels of oligomers were formed in cells expressing SUMO than in wild-type cells, suggesting that SUMO may play a role in protecting MxA from degradation, providing a stable intracellular pool of MxA able to protect cells from viral infection.


Subject(s)
Interferon-alpha/pharmacology , Myxovirus Resistance Proteins/pharmacology , Small Ubiquitin-Related Modifier Proteins/pharmacology , Vesicular Stomatitis/prevention & control , Vesicular stomatitis Indiana virus/physiology , Antiviral Agents/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/virology , HeLa Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Protein Processing, Post-Translational , Rabies/metabolism , Rabies/prevention & control , Rabies/virology , Rabies virus/physiology , Tumor Cells, Cultured , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/virology
9.
Med Sci (Paris) ; 30(8-9): 765-71, 2014.
Article in French | MEDLINE | ID: mdl-25174753

ABSTRACT

PML/TRIM19 is the organizer of PML nuclear bodies (NB), large multiprotein structures associated to the nuclear matrix, which recruit a great number of proteins and which are implicated in various cellular processes including antiviral defense. The conjugation of PML to SUMO is required for the formation and function of PML NB. Alternative splicing from a single PML gene generates several PML isoforms (PMLI to PMLVIIb), each harboring a specific carboxy-terminal region. This variability allows each isoform to recruit different partners and thus confers them specific functions. PML gene is directly induced by interferon and certain PML isoforms are implicated in its antiviral properties, as they display intrinsic antiviral activities against RNA or DNA viruses. One isoform, PMLIV, is also implicated in innate immunity by enhancing IFN-ß production during a viral infection. Here we review recent findings on PML/TRIM19 implication in interferon response and antiviral defense, at the interface between intrinsic and innate immunity.


Subject(s)
Adaptive Immunity , Cell Nucleus/metabolism , Immunity, Innate , Inclusion Bodies/physiology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adaptive Immunity/genetics , Animals , Gene Expression Regulation/drug effects , Humans , Immunity, Innate/genetics , Inclusion Bodies/metabolism , Interferons/pharmacology , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism
10.
PLoS Pathog ; 10(2): e1003975, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586174

ABSTRACT

PML/TRIM19, the organizer of nuclear bodies (NBs), has been implicated in the antiviral response to diverse RNA and DNA viruses. Several PML isoforms generated from a single PML gene by alternative splicing, share the same N-terminal region containing the RBCC/tripartite motif but differ in their C-terminal sequences. Recent studies of all the PML isoforms reveal the specific functions of each. The knockout of PML renders mice more sensitive to vesicular stomatitis virus (VSV). Here we report that among PML isoforms (PMLI to PMLVIIb), only PMLIII and PMLIV confer resistance to VSV. Unlike PMLIII, whose anti-VSV activity is IFN-independent, PMLIV can act at two stages: it confers viral resistance directly in an IFN-independent manner and also specifically enhances IFN-ß production via a higher activation of IRF3, thus protecting yet uninfected cells from oncoming infection. PMLIV SUMOylation is required for both activities. This demonstrates for the first time that PMLIV is implicated in innate immune response through enhanced IFN-ß synthesis. Depletion of IRF3 further demonstrates the dual activity of PMLIV, since it abrogated PMLIV-induced IFN synthesis but not PMLIV-induced inhibition of viral proteins. Mechanistically, PMLIV enhances IFN-ß synthesis by regulating the cellular distribution of Pin1 (peptidyl-prolyl cis/trans isomerase), inducing its recruitment to PML NBs where both proteins colocalize. The interaction of SUMOylated PMLIV with endogenous Pin1 and its recruitment within PML NBs prevents the degradation of activated IRF3, and thus potentiates IRF3-dependent production of IFN-ß. Whereas the intrinsic antiviral activity of PMLIV is specific to VSV, its effect on IFN-ß synthesis is much broader, since it affects a key actor of innate immune pathways. Our results show that, in addition to its intrinsic anti-VSV activity, PMLIV positively regulates IFN-ß synthesis in response to different inducers, thus adding PML/TRIM19 to the growing list of TRIM proteins implicated in both intrinsic and innate immunity.


Subject(s)
Immunity, Innate/immunology , Nuclear Proteins/immunology , Rhabdoviridae Infections/immunology , Signal Transduction/immunology , Transcription Factors/immunology , Tumor Suppressor Proteins/immunology , Animals , Cell Line , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , Interferon-beta/biosynthesis , Interferon-beta/immunology , Mice , Mice, Knockout , Promyelocytic Leukemia Protein , Protein Isoforms , Real-Time Polymerase Chain Reaction , Transfection , Vesiculovirus
11.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...