92 results on '"Chelbi-Alix MK"'
Search Results
2. Retinoic acid and interferon signaling cross talk in normal and RA-resistant APL cells
- Author
-
Chelbi-Alix, MK and Pelicano, L
- Published
- 1999
- Full Text
- View/download PDF
3. The PML growth suppressor has an altered expression in human oncogenesis
- Author
-
Koken, MHM, Linares-Cruz, G, Quignon, F, Viron, A, Chelbi-Alix, MK, Sobczak-Thepot, J, Juhlin, L, Degos, L, The, H, de, Koken, MHM, Linares-Cruz, G, Quignon, F, Viron, A, Chelbi-Alix, MK, Sobczak-Thepot, J, Juhlin, L, Degos, L, and The, H, de
- Published
- 1995
4. Crosstalk Between SUMO and Ubiquitin-Like Proteins: Implication for Antiviral Defense.
- Author
-
Chelbi-Alix MK and Thibault P
- Abstract
Interferon (IFN) is a crucial first line of defense against viral infection. This cytokine induces the expression of several IFN-Stimulated Genes (ISGs), some of which act as restriction factors. Upon IFN stimulation, cells also express ISG15 and SUMO, two key ubiquitin-like (Ubl) modifiers that play important roles in the antiviral response. IFN itself increases the global cellular SUMOylation in a PML-dependent manner. Mass spectrometry-based proteomics enables the large-scale identification of Ubl protein conjugates to determine the sites of modification and the quantitative changes in protein abundance. Importantly, a key difference amongst SUMO paralogs is the ability of SUMO2/3 to form poly-SUMO chains that recruit SUMO ubiquitin ligases such RING finger protein RNF4 and RNF111, thus resulting in the proteasomal degradation of conjugated substrates. Crosstalk between poly-SUMOylation and ISG15 has been reported recently, where increased poly-SUMOylation in response to IFN enhances IFN-induced ISGylation, stabilizes several ISG products in a TRIM25-dependent fashion, and results in enhanced IFN-induced antiviral activities. This contribution will highlight the relevance of the global SUMO proteome and the crosstalk between SUMO, ubiquitin and ISG15 in controlling both the stability and function of specific restriction factors that mediate IFN antiviral defense., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Chelbi-Alix and Thibault.)
- Published
- 2021
- Full Text
- View/download PDF
5. Interferon, restriction factors and SUMO pathways.
- Author
-
El-Asmi F, McManus FP, Thibault P, and Chelbi-Alix MK
- Subjects
- Antiviral Agents, Humans, Promyelocytic Leukemia Protein metabolism, SUMO-1 Protein, Interferons, Sumoylation
- 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., (Copyright © 2020 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
6. Cross-talk between SUMOylation and ISGylation in response to interferon.
- Author
-
El-Asmi F, McManus FP, Brantis-de-Carvalho CE, Valle-Casuso JC, Thibault P, and Chelbi-Alix MK
- Subjects
- 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, Interferon-alpha pharmacology, Sumoylation drug effects
- 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., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
7. [PML isoforms and TGF-β response].
- Author
-
El-Asmi F and Chelbi-Alix MK
- Subjects
- 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, Promyelocytic Leukemia Protein physiology, Transforming Growth Factor beta metabolism
- 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-β-induced apoptosis. Cytoplasmic PML is an essential activator of TGF-β signaling by increasing the phosphorylation of transcription factors SMAD2/3 while nuclear PML plays a role in TGF-β-induced caspase 8 activation and apoptosis. TGF-β 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-β, 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-β response., (© 2020 médecine/sciences – Inserm.)
- Published
- 2020
- Full Text
- View/download PDF
8. TGF-β induces PML SUMOylation, degradation and PML nuclear body disruption.
- Author
-
El-Asmi F, El-Mchichi B, Maroui MA, Dianoux L, and Chelbi-Alix MK
- Subjects
- 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, Small Ubiquitin-Related Modifier Proteins metabolism, Cell Nucleus metabolism, Promyelocytic Leukemia Protein metabolism, Proteolysis drug effects, Sumoylation drug effects, Transforming Growth Factor beta pharmacology
- 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-β-induced apoptosis and that cytoplasmic PML is an essential activator of TGF-β signaling. The role and the fate of interferon (IFN)-enhanced PML NBs in response to TGF-β have not been investigated. Here we show that IFNα potentiated TGF-β-mediated apoptosis in human cells. IFNα or ectopic expression of PMLIV, but not of PMLIII, enhanced TGF-β-induced caspase 8 activation. In response to TGF-β, 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-β-induced apoptosis and caspase 8 activation., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
9. Rhabdoviruses, Antiviral Defense, and SUMO Pathway.
- Author
-
El Asmi F, Brantis-de-Carvalho CE, Blondel D, and Chelbi-Alix MK
- Subjects
- 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, Rhabdoviridae immunology, Rhabdoviridae Infections immunology, Signal Transduction, Small Ubiquitin-Related Modifier Proteins metabolism
- 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., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2018
- Full Text
- View/download PDF
10. Promyelocytic Leukemia Protein (PML) Requirement for Interferon-induced Global Cellular SUMOylation.
- Author
-
Maroui MA, Maarifi G, McManus FP, Lamoliatte F, Thibault P, and Chelbi-Alix MK
- Subjects
- HEK293 Cells, Humans, Interferon-alpha pharmacology, Promyelocytic Leukemia Protein metabolism, Sumoylation drug effects
- Abstract
We report that interferon (IFN) α treatment at short and long periods increases the global cellular SUMOylation and requires the presence of the SUMO E3 ligase promyelocytic leukemia protein (PML), the organizer of PML nuclear bodies (NBs). Several PML isoforms (PMLI-PMLVII) derived from a single PML gene by alternative splicing, share the same N-terminal region but differ in their C-terminal sequences. Introducing each of the human PML isoform in PML-negative cells revealed that enhanced SUMOylation in response to IFN is orchestrated by PMLIII and PMLIV. Large-scale proteomics experiments enabled the identification of 558 SUMO sites on 389 proteins, of which 172 sites showed differential regulation upon IFNα stimulation, including K49 from UBC9, the sole SUMO E2 protein. Furthermore, IFNα induces PML-dependent UBC9 transfer to the nuclear matrix where it colocalizes with PML within the NBs and enhances cellular SUMOylation levels. Our results demonstrate that SUMOylated UBC9 and PML are key players for IFN-increased cellular SUMOylation., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
11. [Modulation by SUMO of PKR activation and stability].
- Author
-
El Asmi F, Maarifi G, Ali Maroui M, Dianoux L, and Chelbi-Alix MK
- Subjects
- Gene Expression Regulation physiology, Humans, Signal Transduction physiology, Small Ubiquitin-Related Modifier Proteins metabolism, eIF-2 Kinase metabolism
- Published
- 2018
- Full Text
- View/download PDF
12. Differential effects of SUMO1 and SUMO3 on PKR activation and stability.
- Author
-
Maarifi G, El Asmi F, Maroui MA, Dianoux L, and Chelbi-Alix MK
- Subjects
- Active Transport, Cell Nucleus, Cell Nucleus metabolism, Enzyme Stability, HeLa Cells, Humans, Phosphorylation, SUMO-1 Protein genetics, Sumoylation, Ubiquitins genetics, SUMO-1 Protein metabolism, Ubiquitins metabolism, eIF-2 Kinase metabolism
- 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.
- Published
- 2018
- Full Text
- View/download PDF
13. Uncovering the SUMOylation and ubiquitylation crosstalk in human cells using sequential peptide immunopurification.
- Author
-
Lamoliatte F, McManus FP, Maarifi G, Chelbi-Alix MK, and Thibault P
- Subjects
- Amino Acid Motifs, HEK293 Cells, Humans, Promyelocytic Leukemia Protein chemistry, Promyelocytic Leukemia Protein genetics, Promyelocytic Leukemia Protein metabolism, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism, Protein Interaction Maps, Proteins chemistry, Proteins genetics, Proteolysis, Sumoylation, Ubiquitin chemistry, Ubiquitination, Chromatography, Affinity methods, Peptides chemistry, Proteins metabolism
- Abstract
Crosstalk between the SUMO and ubiquitin pathways has recently been reported. However, no approach currently exists to determine the interrelationship between these modifications. Here, we report an optimized immunoaffinity method that permits the study of both protein ubiquitylation and SUMOylation from a single sample. This method enables the unprecedented identification of 10,388 SUMO sites in HEK293 cells. The sequential use of SUMO and ubiquitin remnant immunoaffinity purification facilitates the dynamic profiling of SUMOylated and ubiquitylated proteins in HEK293 cells treated with the proteasome inhibitor MG132. Quantitative proteomic analyses reveals crosstalk between substrates that control protein degradation, and highlights co-regulation of SUMOylation and ubiquitylation levels on deubiquitinase enzymes and the SUMOylation of proteasome subunits. The SUMOylation of the proteasome affects its recruitment to promyelocytic leukemia protein (PML) nuclear bodies, and PML lacking the SUMO interacting motif fails to colocalize with SUMOylated proteasome further demonstrating that this motif is required for PML catabolism.
- Published
- 2017
- Full Text
- View/download PDF
14. MxA Mediates SUMO-Induced Resistance to Vesicular Stomatitis Virus.
- Author
-
Maarifi G, Hannoun Z, Geoffroy MC, El Asmi F, Zarrouk K, Nisole S, Blondel D, and Chelbi-Alix MK
- Subjects
- 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, Interferon-alpha pharmacology, Myxovirus Resistance Proteins pharmacology, Small Ubiquitin-Related Modifier Proteins pharmacology, Vesicular Stomatitis prevention & control, Vesicular stomatitis Indiana virus physiology
- 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., (Copyright © 2016, American Society for Microbiology. All Rights Reserved.)
- Published
- 2016
- Full Text
- View/download PDF
15. The implication of SUMO in intrinsic and innate immunity.
- Author
-
Hannoun Z, Maarifi G, and Chelbi-Alix MK
- Subjects
- Animals, Humans, Janus Kinases immunology, STAT Transcription Factors immunology, Immunity, Innate, SUMO-1 Protein immunology, Signal Transduction immunology, Virus Diseases immunology
- Abstract
Since its discovery, SUMOylation has emerged as a key post-translational modification involved in the regulation of host-virus interactions. SUMOylation has been associated with the replication of a large number of viruses, either through the direct modification of viral proteins or through the modulation of cellular proteins implicated in antiviral defense. SUMO can affect protein function via covalent or non-covalent binding. There is growing evidence that SUMO regulates several host proteins involved in intrinsic and innate immunity, thereby contributing to the process governing interferon production during viral infection; as well as the interferon-activated Jak/STAT pathway. Unlike the interferon-mediated innate immune response, intrinsic antiviral resistance is mediated by constitutively expressed antiviral proteins (defined as restriction factors), which confer direct viral resistance through a variety of mechanisms. The aim of this review is to evaluate the role of SUMO in intrinsic and innate immunity; highlighting the involvement of the TRIM family proteins, with a specific focus on the mechanism through which SUMO affects i- interferon production upon viral infection, ii-interferon Jak/STAT signaling and biological responses, iii-the relationship between restriction factors and RNA viruses., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
16. [SUMO paralogs and interferon response].
- Author
-
Maarifi G, Dianoux L, Nisole S, and Chelbi-Alix MK
- Subjects
- Antiviral Agents pharmacology, Gene Expression Regulation drug effects, Humans, Phosphorylation drug effects, Promyelocytic Leukemia Protein metabolism, Protein Isoforms physiology, Protein Kinases metabolism, Proteolysis, STAT1 Transcription Factor metabolism, Interferons pharmacology, Small Ubiquitin-Related Modifier Proteins physiology
- Published
- 2016
- Full Text
- View/download PDF
17. PML/TRIM19-Dependent Inhibition of Retroviral Reverse-Transcription by Daxx.
- Author
-
Dutrieux J, Maarifi G, Portilho DM, Arhel NJ, Chelbi-Alix MK, and Nisole S
- Subjects
- Cell Nucleus metabolism, Co-Repressor Proteins, HIV-1 metabolism, Humans, Molecular Chaperones, Promyelocytic Leukemia Protein, Protein Binding physiology, Transcription, Genetic, Adaptor Proteins, Signal Transducing metabolism, Nuclear Proteins metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
PML (Promyelocytic Leukemia protein), also known as TRIM19, belongs to the family of tripartite motif (TRIM) proteins. PML is mainly expressed in the nucleus, where it forms dynamic structures known as PML nuclear bodies that recruit many other proteins, such as Sp100 and Daxx. While the role of PML/TRIM19 in antiviral defense is well documented, its effect on HIV-1 infection remains unclear. Here we show that infection by HIV-1 and other retroviruses triggers the formation of PML cytoplasmic bodies, as early as 30 minutes post-infection. Quantification of the number and size of PML cytoplasmic bodies revealed that they last approximately 8 h, with a peak at 2 h post-infection. PML re-localization is blocked by reverse-transcription inhibitors and is not observed following infection with unrelated viruses, suggesting it is specifically triggered by retroviral reverse-transcription. Furthermore, we show that PML interferes with an early step of retroviral infection since PML knockdown dramatically increases reverse-transcription efficiency. We demonstrate that PML does not inhibit directly retroviral infection but acts through the stabilization of one of its well-characterized partners, Daxx. In the presence of PML, cytoplasmic Daxx is found in the vicinity of incoming HIV-1 capsids and inhibits reverse-transcription. Interestingly, Daxx not only interferes with exogenous retroviral infections but can also inhibit retrotransposition of endogenous retroviruses, thus identifying Daxx as a broad cellular inhibitor of reverse-transcription. Altogether, these findings unravel a novel antiviral function for PML and PML nuclear body-associated protein Daxx.
- Published
- 2015
- Full Text
- View/download PDF
18. Small Ubiquitin-like Modifier Alters IFN Response.
- Author
-
Maarifi G, Maroui MA, Dutrieux J, Dianoux L, Nisole S, and Chelbi-Alix MK
- Subjects
- 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, Interferons pharmacology, SUMO-1 Protein metabolism, Small Ubiquitin-Related Modifier Proteins metabolism, Ubiquitins metabolism
- 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., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
19. Resistance to Rhabdoviridae Infection and Subversion of Antiviral Responses.
- Author
-
Blondel D, Maarifi G, Nisole S, and Chelbi-Alix MK
- Subjects
- Animals, Humans, Interferons genetics, Interferons immunology, Rhabdoviridae genetics, Rhabdoviridae Infections genetics, Rhabdoviridae Infections virology, Rhabdoviridae physiology, Rhabdoviridae Infections immunology
- Abstract
Interferon (IFN) treatment induces the expression of hundreds of IFN-stimulated genes (ISGs). However, only a selection of their products have been demonstrated to be responsible for the inhibition of rhabdovirus replication in cultured cells; and only a few have been shown to play a role in mediating the antiviral response in vivo using gene knockout mouse models. IFNs inhibit rhabdovirus replication at different stages via the induction of a variety of ISGs. This review will discuss how individual ISG products confer resistance to rhabdoviruses by blocking viral entry, degrading single stranded viral RNA, inhibiting viral translation or preventing release of virions from the cell. Furthermore, this review will highlight how these viruses counteract the host IFN system.
- Published
- 2015
- Full Text
- View/download PDF
20. MxA interacts with and is modified by the SUMOylation machinery.
- Author
-
Brantis-de-Carvalho CE, Maarifi G, Gonçalves Boldrin PE, Zanelli CF, Nisole S, Chelbi-Alix MK, and Valentini SR
- Subjects
- Amino Acid Motifs, Animals, Binding Sites, HeLa Cells, Humans, Mice, Myxovirus Resistance Proteins chemistry, NIH 3T3 Cells, Protein Binding, SUMO-1 Protein chemistry, SUMO-1 Protein metabolism, Ubiquitin-Conjugating Enzymes chemistry, Ubiquitin-Conjugating Enzymes metabolism, Myxovirus Resistance Proteins metabolism, Sumoylation
- Abstract
Mx proteins are evolutionarily conserved dynamin-like large GTPases involved in viral resistance triggered by types I and III interferons. The human MxA is a cytoplasmic protein that confers resistance to a large number of viruses. The MxA protein is also known to self-assembly into high molecular weight homo-oligomers. Using a yeast two-hybrid screen, we identified 27 MxA binding partners, some of which are related to the SUMOylation machinery. The interaction of MxA with Small-Ubiquitin MOdifier 1 (SUMO1) and Ubiquitin conjugating enzyme 9 (Ubc9) was confirmed by co-immunoprecipitation and co-localization by confocal microscopy. We identified one SUMO conjugation site at lysine 48 and two putative SUMO interacting motifs (SIMa and SIMb). We showed that MxA interacts with the EIL loop of SUMO1 in a SIM-independent manner via its CID-GED domain. The yeast two-hybrid mapping also revealed that Ubc9 binds to the MxA GTPase domain. Mutation in the putative SIMa and SIMb, which are located in the GTPase binding domain, reduced MxA antiviral activity. In addition, we showed that MxA can be conjugated to SUMO2 or SUMO3 at lysine 48 and that the SUMOylation-deficient mutant of MxA (MxAK48R) retained its capacity to oligomerize and to inhibit Vesicular Stomatitis Virus (VSV) and Influenza A Virus replication, suggesting that MxA SUMOylation is not essential for its antiviral activity., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
21. Sodium arsenite induces apoptosis and Epstein-Barr virus reactivation in lymphoblastoid cells.
- Author
-
Zebboudj A, Maroui MA, Dutrieux J, Touil-Boukoffa C, Bourouba M, Chelbi-Alix MK, and Nisole S
- Subjects
- Antigens, Nuclear metabolism, Arsenic Trioxide, Arsenicals pharmacology, Autoantigens metabolism, Autophagy drug effects, Burkitt Lymphoma pathology, Burkitt Lymphoma virology, Cell Line, Tumor drug effects, Cell Proliferation drug effects, Herpesvirus 4, Human metabolism, Humans, Nuclear Proteins metabolism, Oxides pharmacology, Promyelocytic Leukemia Protein, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism, Viral Matrix Proteins metabolism, Apoptosis drug effects, Arsenites pharmacology, Herpesvirus 4, Human drug effects, Sodium Compounds pharmacology
- Abstract
Epstein-Barr virus (EBV) is associated with several malignancies, including carcinomas, such as nasopharyngeal carcinoma, and lymphomas, such as Burkitt's lymphoma and Hodgkin's lymphoma. The Latent Membrane Protein 1 (LMP1) is the major oncogene protein of EBV as its expression is responsible for the induction of cell transformation, immortalization and proliferation. Arsenic trioxide was shown to induce a cytotoxic effect on nasopharyngeal cancer cells associated with LMP1 down-regulation. However, the effect of arsenic on EBV-associated lymphoproliferative malignancies has been less studied. We investigated the effect of two different arsenical compounds, arsenic trioxide (As2O3) and sodium arsenite (NaAsO2) on the induction of cell death in P3HR1 cells, an Epstein-Barr virus-positive Burkitt lymphoma derived cell line. Both compounds inhibited cell growth and induced cell death. By flow-cytometry and Western blot analysis, we provide evidence that NaAsO2 induced caspase-dependent apoptosis whereas As2O3 triggered autophagic cell death. Furthermore, we show that NaAsO2 treatment led to a dramatic decrease of the expression level of LMP1 and the cellular protein PML. Importantly, this down-regulation was associated with a reactivation of EBV lytic cycle through the induction of immediate-early proteins Zta and Rta. These results are in agreement with a model in which LMP1 maintains EBV in a latent state by stabilizing PML expression. Altogether, our results suggest that NaAsO2 would represent a better therapeutic candidate than As2O3 in EBV-induced B lymphoma for its capacity to promote viral reactivation., (Copyright © 2014 Elsevier B.V. and Société française de biochimie et biologie Moléculaire (SFBBM). All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
22. Large-scale analysis of lysine SUMOylation by SUMO remnant immunoaffinity profiling.
- Author
-
Lamoliatte F, Caron D, Durette C, Mahrouche L, Maroui MA, Caron-Lizotte O, Bonneil E, Chelbi-Alix MK, and Thibault P
- Subjects
- Animals, Antibodies, Monoclonal immunology, Chromatography, Affinity, Gas Chromatography-Mass Spectrometry, HEK293 Cells, Humans, Hybridomas metabolism, Peptides immunology, Peptides metabolism, Proteome metabolism, Rabbits, Small Ubiquitin-Related Modifier Proteins immunology, Tumor Suppressor Proteins immunology, Tumor Suppressor Proteins metabolism, Lysine metabolism, Small Ubiquitin-Related Modifier Proteins metabolism, Sumoylation
- Abstract
Small ubiquitin-related modifiers (SUMO) are evolutionarily conserved ubiquitin-like proteins that regulate several cellular processes including cell cycle progression, intracellular trafficking, protein degradation and apoptosis. Despite the importance of protein SUMOylation in different biological pathways, the global identification of acceptor sites in complex cell extracts remains a challenge. Here we generate a monoclonal antibody that enriches for peptides containing SUMO remnant chains following tryptic digestion. We identify 954 SUMO3-modified lysine residues on 538 proteins and profile by quantitative proteomics the dynamic changes of protein SUMOylation following proteasome inhibition. More than 86% of these SUMOylation sites have not been reported previously, including 5 sites on the tumour suppressor parafibromin (CDC73). The modification of CDC73 at K136 affects its nuclear retention within PML nuclear bodies on proteasome inhibition. In contrast, a CDC73 K136R mutant translocates to the cytoplasm under the same conditions, further demonstrating the effectiveness of our method to characterize the dynamics of lysine SUMOylation.
- Published
- 2014
- Full Text
- View/download PDF
23. PML control of cytokine signaling.
- Author
-
Maarifi G, Chelbi-Alix MK, and Nisole S
- Subjects
- Animals, Humans, Inflammasomes immunology, Inflammasomes metabolism, Nuclear Proteins genetics, Promyelocytic Leukemia Protein, Protein Isoforms genetics, Protein Structure, Tertiary, Transcription Factors genetics, Tumor Suppressor Proteins genetics, Cytokines immunology, Nuclear Proteins physiology, Signal Transduction, Transcription Factors physiology, Tumor Suppressor Proteins physiology
- Abstract
The promyelocytic leukemia (PML) protein is a tumor suppressor acting as the organizer of nuclear matrix-associated structures named nuclear bodies (NBs). The involvement of PML in various cell processes, including cell death, senescence or antiviral defense underlines the multiple functions of PML due to its ability to interact with various partners either in the cytoplasm or in the nucleus. The importance of paracrine signaling in the regulation of PML expression is well established. More recently, a growing body of evidence also supports PML as a key regulator of cytokine signaling. These findings shed light on unsuspected biological functions of PML such as immune response, inflammation and cytokine-induced apoptosis. Here we review the current understanding of the pleiotropic activities of PML on cytokine-induced signaling., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
24. [Implication of PML nuclear bodies in intrinsic and innate immunity].
- Author
-
Maroui MA, El Asmi F, Dutrieux J, Chelbi-Alix MK, and Nisole S
- Subjects
- Animals, Gene Expression Regulation drug effects, Humans, 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, Adaptive Immunity genetics, Cell Nucleus metabolism, Immunity, Innate genetics, Inclusion Bodies physiology, Nuclear Proteins metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- 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-β 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., (© 2014 médecine/sciences – Inserm.)
- Published
- 2014
- Full Text
- View/download PDF
25. Implication of PMLIV in both intrinsic and innate immunity.
- Author
-
El Asmi F, Maroui MA, Dutrieux J, Blondel D, Nisole S, and Chelbi-Alix MK
- Subjects
- 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, Immunity, Innate immunology, Nuclear Proteins immunology, Rhabdoviridae Infections immunology, Signal Transduction immunology, Transcription Factors immunology, Tumor Suppressor Proteins immunology
- 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-β 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-β 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-β 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-β. Whereas the intrinsic antiviral activity of PMLIV is specific to VSV, its effect on IFN-β 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-β synthesis in response to different inducers, thus adding PML/TRIM19 to the growing list of TRIM proteins implicated in both intrinsic and innate immunity.
- Published
- 2014
- Full Text
- View/download PDF
26. Differential Roles of PML Isoforms.
- Author
-
Nisole S, Maroui MA, Mascle XH, Aubry M, and Chelbi-Alix MK
- Abstract
The tumor suppressor promyelocytic leukemia (PML) protein is fused to the retinoic acid receptor alpha in patients suffering from acute promyelocytic leukemia (APL). Treatment of APL patients with arsenic trioxide (As2O3) reverses the disease phenotype by a process involving the degradation of the fusion protein via its PML moiety. Several PML isoforms are generated from a single PML gene by alternative splicing. They 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. Here, we review the nomenclature and structural organization of the PML isoforms in order to clarify the various designations and classifications found in different databases. The functions of the PML isoforms and their differential roles in antiviral defense also are reviewed. Finally, the key players involved in the degradation of the PML isoforms in response to As2O3 or other inducers are discussed.
- Published
- 2013
- Full Text
- View/download PDF
27. Requirement of PML SUMO interacting motif for RNF4- or arsenic trioxide-induced degradation of nuclear PML isoforms.
- Author
-
Maroui MA, Kheddache-Atmane S, El Asmi F, Dianoux L, Aubry M, and Chelbi-Alix MK
- Subjects
- 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, 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
- 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 β 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.
- Published
- 2012
- Full Text
- View/download PDF
28. Promyelocytic leukemia isoform IV confers resistance to encephalomyocarditis virus via the sequestration of 3D polymerase in nuclear bodies.
- Author
-
Maroui MA, Pampin M, and Chelbi-Alix MK
- Subjects
- Animals, Cell Line, Cricetinae, Humans, Mice, Promyelocytic Leukemia Protein, Protein Binding, Protein Interaction Mapping, Protein Isoforms metabolism, DNA-Directed RNA Polymerases antagonists & inhibitors, DNA-Directed RNA Polymerases metabolism, Encephalomyocarditis virus enzymology, Encephalomyocarditis virus immunology, Nuclear Proteins metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Promyelocytic leukemia (PML) protein is the organizer of nuclear matrix-associated nuclear bodies (NBs), and its conjugation to the small ubiquitin-like modifier (SUMO) is required for the formation of these structures. Several alternatively spliced PML transcripts from a single PML gene lead to the production of seven PML isoforms (PML isoform I [PMLI] to VII [PMLVII]), which all share a N-terminal region that includes the RBCC (RING, B boxes, and a α-helical coiled-coil) motif but differ in the C-terminal region. This diversity of PML isoforms determines the specific functions of each isoform. There is increasing evidence implicating PML in host antiviral defense and suggesting various strategies involving PML to counteract viral production. We reported that mouse embryonic fibroblasts derived from PML knockout mice are more sensitive than wild-type cells to infection with encephalomyocarditis virus (EMCV). Here, we show that stable expression of PMLIV or PMLIVa inhibited viral replication and protein synthesis, leading to a substantial reduction of EMCV multiplication. This protective effect required PMLIV SUMOylation and was not observed with other nuclear PML isoforms (I, II, III, V, and VI) or with the cytoplasmic PMLVII. We demonstrated that only PMLIV interacted with EMCV 3D polymerase (3Dpol) and sequestered it within PML NBs. The C-terminal region specific to PMLIV was required for both interaction with 3Dpol and the antiviral properties. Also, depletion of PMLIV by RNA interference significantly boosted EMCV production in interferon-treated cells. These findings indicate the mechanism by which PML confers resistance to EMCV. They also reveal a new pathway mediating the antiviral activity of interferon against EMCV.
- Published
- 2011
- Full Text
- View/download PDF
29. Arsenic trioxide reduces 2,4,6-trinitrobenzene sulfonic acid-induced murine colitis via nuclear factor-κB down-regulation and caspase-3 activation.
- Author
-
Singer M, Trugnan G, and Chelbi-Alix MK
- Subjects
- Animals, Anti-Inflammatory Agents adverse effects, Apoptosis drug effects, Arsenic Trioxide, Arsenicals adverse effects, Caspase 3 metabolism, Colitis chemically induced, Colitis immunology, Colon metabolism, Colon pathology, Crohn Disease immunology, Cytokines genetics, Cytokines metabolism, Disease Models, Animal, Humans, Male, Mice, Mice, Inbred BALB C, NF-kappa B genetics, NF-kappa B metabolism, Nitric Oxide Synthase Type II genetics, Nitric Oxide Synthase Type II metabolism, Oxides adverse effects, Peroxidase genetics, Peroxidase metabolism, Trinitrobenzenesulfonic Acid administration & dosage, Anti-Inflammatory Agents administration & dosage, Arsenicals administration & dosage, Colitis drug therapy, Colon drug effects, Crohn Disease drug therapy, Oxides administration & dosage
- Abstract
Arsenic trioxide, As(2)O(3), already used in human anti-cancer therapy, is also an efficient agent against the autoimmune and inflammatory diseases developed in MRL/lpr mice. Inflammatory bowel diseases (IBDs), notably Crohn's disease, which remain without efficient treatment, display autoimmune and inflammatory components. We, therefore, hypothesized that As(2)O( 3) may be active on IBDs. Using the 2,4,6-trinitrobenzene sulfonic acid-induced murine model of colitis, we demonstrate that As(2)O(3) used either in a preventive or a curative mode markedly reduced the induced colitis as assessed by macroscopic and microscopic scores, leading to prolonged mice survival. In addition, As(2)O(3) was able to inhibit NF-κB expression and DNA-binding in colon extracts leading to decreased cytokine gene expression (i.e. tumor necrosis factor-α, interleukin(IL)-1β, IL-12, IL-17, IL-18, and IL-23). Interestingly, As(2)O(3) also reduced keratinocyte-derived chemokine (KC), inducible nitric oxide synthase (iNOS) mRNA levels, and myeloperoxidase (MPO) protein expression suggesting an impairment of neutrophils. This was associated with a marked increase of procaspase-3 and induced caspase-3 activation. This caspase-3 co-localized with MPO in the remaining neutrophils suggesting that As(2)O( 3) might have eliminated inflamed cells probably by inducing their apoptosis. These results assessed the potent anti-inflammatory effect of As(2)O( 3), that targets both NF-κB and caspase-3 pathways, and suggests a therapeutic potential for Crohn's disease and other severe IBDs.
- Published
- 2011
- Full Text
- View/download PDF
30. PML positively regulates interferon gamma signaling.
- Author
-
El Bougrini J, Dianoux L, and Chelbi-Alix MK
- Subjects
- 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, 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, Interferon-gamma metabolism, Nuclear Proteins metabolism, Signal Transduction genetics, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- 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., (Copyright © 2010 Elsevier Masson SAS. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
31. A novel proteomics approach to identify SUMOylated proteins and their modification sites in human cells.
- Author
-
Galisson F, Mahrouche L, Courcelles M, Bonneil E, Meloche S, Chelbi-Alix MK, and Thibault P
- Subjects
- Amino Acid Sequence, Chromatin chemistry, Computational Biology, DNA Repair, HEK293 Cells, Humans, Leukemia, Promyelocytic, Acute drug therapy, Leukemia, Promyelocytic, Acute metabolism, Mass Spectrometry methods, Microscopy, Confocal methods, Molecular Sequence Data, Mutation, Proteome, Subcellular Fractions metabolism, Proteomics methods, Small Ubiquitin-Related Modifier Proteins chemistry
- Abstract
The small ubiquitin-related modifier (SUMO) is a small group of proteins that are reversibly attached to protein substrates to modify their functions. The large scale identification of protein SUMOylation and their modification sites in mammalian cells represents a significant challenge because of the relatively small number of in vivo substrates and the dynamic nature of this modification. We report here a novel proteomics approach to selectively enrich and identify SUMO conjugates from human cells. We stably expressed different SUMO paralogs in HEK293 cells, each containing a His(6) tag and a strategically located tryptic cleavage site at the C terminus to facilitate the recovery and identification of SUMOylated peptides by affinity enrichment and mass spectrometry. Tryptic peptides with short SUMO remnants offer significant advantages in large scale SUMOylome experiments including the generation of paralog-specific fragment ions following CID and ETD activation, and the identification of modified peptides using conventional database search engines such as Mascot. We identified 205 unique protein substrates together with 17 precise SUMOylation sites present in 12 SUMO protein conjugates including three new sites (Lys-380, Lys-400, and Lys-497) on the protein promyelocytic leukemia. Label-free quantitative proteomics analyses on purified nuclear extracts from untreated and arsenic trioxide-treated cells revealed that all identified SUMOylated sites of promyelocytic leukemia were differentially SUMOylated upon stimulation.
- Published
- 2011
- Full Text
- View/download PDF
32. Role of promyelocytic leukemia protein in host antiviral defense.
- Author
-
Geoffroy MC and Chelbi-Alix MK
- Subjects
- Animals, Host-Pathogen Interactions, Humans, Intranuclear Inclusion Bodies physiology, Nuclear Proteins chemistry, Nuclear Proteins genetics, Promyelocytic Leukemia Protein, Protein Interaction Domains and Motifs, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Processing, Post-Translational, Transcription Factors chemistry, Transcription Factors genetics, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins genetics, Virus Diseases immunology, Gene Expression Regulation, Immunity, Innate, Interferons metabolism, Nuclear Proteins metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism, Virus Diseases metabolism
- Abstract
Several pathways have been implicated in the establishment of antiviral state in response to interferon (IFN), one of which implicates the promyelocytic leukemia (PML) protein. The PML gene has been discovered 20 years ago and has led to new insights into oncogenesis, apoptosis, cell senescence, and antiviral defense. PML is induced by IFN, leading to a marked increase of expression of PML isoforms and the number of PML nuclear bodies (NBs). PML is the organizer of the NBs that contains at least 2 permanent NB-associated proteins, the IFN-stimulated gene product Speckled protein of 100 kDa (Sp100) and death-associated dead protein (Daxx), as well as numerous other transient proteins recruited in these structures in response to different stimuli. Accumulating reports have implicated PML in host antiviral defense and revealed various strategies developed by viruses to disrupt PML NBs. This review will focus on the regulation of PML and the implication of PML NBs in conferring resistance to DNA and RNA viruses. The role of PML in mediating an IFN-induced antiviral state will also be discussed.
- Published
- 2011
- Full Text
- View/download PDF
33. SUMOylation promotes PML degradation during encephalomyocarditis virus infection.
- Author
-
El McHichi B, Regad T, Maroui MA, Rodriguez MS, Aminev A, Gerbaud S, Escriou N, Dianoux L, and Chelbi-Alix MK
- Subjects
- 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, 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
- 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.
- Published
- 2010
- Full Text
- View/download PDF
34. Resistance to rabies virus infection conferred by the PMLIV isoform.
- Author
-
Blondel D, Kheddache S, Lahaye X, Dianoux L, and Chelbi-Alix MK
- Subjects
- 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, Nuclear Proteins immunology, Rabies immunology, Rabies prevention & control, Rabies virus pathogenicity, Transcription Factors immunology, Tumor Suppressor Proteins immunology
- 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.
- Published
- 2010
- Full Text
- View/download PDF
35. Role of SUMO in RNF4-mediated promyelocytic leukemia protein (PML) degradation: sumoylation of PML and phospho-switch control of its SUMO binding domain dissected in living cells.
- Author
-
Percherancier Y, Germain-Desprez D, Galisson F, Mascle XH, Dianoux L, Estephan P, Chelbi-Alix MK, and Aubry M
- Subjects
- 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, Leukemia, Promyelocytic, Acute metabolism, Nuclear Proteins metabolism, SUMO-1 Protein metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- 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.
- Published
- 2009
- Full Text
- View/download PDF
36. Novel function of STAT1beta in B cells: induction of cell death by a mechanism different from that of STAT1alpha.
- Author
-
Najjar I, Schischmanoff PO, Baran-Marszak F, Deglesne PA, Youlyouz-Marfak I, Pampin M, Feuillard J, Bornkamm GW, Chelbi-Alix MK, and Fagard R
- Subjects
- Blotting, Western, Cell Nucleus metabolism, Cells, Cultured, Cytoplasm metabolism, Flow Cytometry, Fluorescent Antibody Technique, Humans, Nerve Tissue Proteins metabolism, Protein Isoforms, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Nerve Growth Factor metabolism, Reverse Transcriptase Polymerase Chain Reaction, Transcription, Genetic, Transfection, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Apoptosis physiology, B-Lymphocytes immunology, STAT1 Transcription Factor physiology
- Abstract
Alternate splicing of STAT1 produces two isoforms: alpha, known as the active form, and beta, previously shown to act as a dominant-negative factor. Most studies have dealt with STAT1alpha, showing its involvement in cell growth control and cell death. To examine the specific function of either isoform in cell death, a naturally STAT1-deficient human B cell line was transfected to express STAT1alpha or STAT1beta. STAT1alpha, expressed alone, enhanced cell death, potentiated the fludarabine-induced apoptosis, and enhanced the nuclear location, the phosphorylation, and the transcriptional activity of p53. Unexpectedly, STAT1beta, expressed alone, induced cell death through a mechanism that was independent of the nuclear function of p53. Indeed, in STAT1beta-expressing B cells, p53 was strictly cytoplasmic where it formed clusters, and there was no induction of the transcriptional activity of p53. These data reveal a novel role of STAT1beta in programmed cell death, which is independent of p53.
- Published
- 2008
- Full Text
- View/download PDF
37. [New therapeutic perspectives for arsenic: from acute promyelocytic leukemia to autoimmune diseases].
- Author
-
Bobé P and Chelbi-Alix MK
- Subjects
- Animals, Arsenic Trioxide, Arsenicals therapeutic use, Disease Models, Animal, Growth Inhibitors therapeutic use, Humans, Lupus Erythematosus, Systemic drug therapy, Lymphoproliferative Disorders drug therapy, Mice, Oxides therapeutic use, Antineoplastic Agents therapeutic use, Arsenic therapeutic use, Autoimmune Diseases drug therapy, Leukemia, Promyelocytic, Acute drug therapy
- Abstract
Since 1996, arsenic trioxide (As2O3) is used to treat patients with acute promyelocytic leukemia. We have recently shown that As2O3 is a novel promising therapeutic agent for the autoimmune diseases (human lupus-like syndrome) and the massive lymphoproliferation (human autoimmune lymphoproliferative-like syndrome) developed by MRL/lpr mice. As2O3 is able to achieve an almost complete regression of antibody- and cell-mediated manifestations in MRL/lpr mice. As2O3 eliminated the activated T lymphocytes responsible for lymphoproliferation and skin, lung, and kidney lesions. This treatment also markedly reduced anti-DNA autoantibodies, rheumatoid factor, IL-18, IFN-gamma, nitric oxide metabolites, TNF-alpha, Fas ligand and IL-10 levels, and immune-complex deposits in glomeruli, leading to significantly prolonged survival rates.
- Published
- 2008
- Full Text
- View/download PDF
38. Implication of TRIM alpha and TRIMCyp in interferon-induced anti-retroviral restriction activities.
- Author
-
Carthagena L, Parise MC, Ringeard M, Chelbi-Alix MK, Hazan U, and Nisole S
- Subjects
- Animals, Antiviral Restriction Factors, Aotus trivirgatus, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Line, Chlorocebus aethiops, Cyclophilin A genetics, Gene Expression Regulation, HIV-1 pathogenicity, HeLa Cells, Humans, Leukemia Virus, Murine pathogenicity, Macaca mulatta, Proteins genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Tripartite Motif Proteins, Ubiquitin-Protein Ligases, Vero Cells, Antiviral Agents pharmacology, Cyclophilin A metabolism, HIV-1 drug effects, Interferons pharmacology, Leukemia Virus, Murine drug effects, Proteins metabolism
- Abstract
Background: TRIM5 alpha is a restriction factor that interferes with retroviral infections in a species-specific manner in primate cells. Although TRIM5 alpha is constitutively expressed, its expression has been shown to be up-regulated by type I interferon (IFN). Among primates, a particular case exists in owl monkey cells, which express a fusion protein between TRIM5 and cyclophilin A, TRIMCyp, specifically interfering with HIV-1 infection. No studies have been conducted so far concerning the possible induction of TRIMCyp by IFN. We investigated the consequences of IFN treatment on retroviral restriction in diverse primate cells and evaluated the implication of TRIM5 alpha or TRIMCyp in IFN-induced anti-retroviral activities., Results: First, we show that human type I IFN can enhance TRIM5 alpha expression in human, African green monkey and macaque cells, as well as TRIMCyp expression in owl monkey cells. In TRIM5 alpha-expressing primate cell lines, type I IFN has little or no effect on HIV-1 infection, whereas it potentiates restriction activity against N-MLV in human and African green monkey cells. In contrast, type I IFN treatment of owl monkey cells induces a great enhancement of HIV-1 restriction, as well as a strain-tropism independent restriction of MLV. We were able to demonstrate that TRIM5 alpha is the main mediator of the IFN-induced activity against N-MLV in human and African green monkey cells, whereas TRIMCyp mediates the IFN-induced HIV-1 restriction enhancement in owl monkey cells. In contrast, the type I IFN-induced anti-MLV restriction in owl monkey cells is independent of TRIMCyp expression., Conclusion: Together, our observations indicate that both TRIM5 alpha and TRIMCyp are implicated in IFN-induced anti-retroviral response in primate cells. Furthermore, we found that type I IFN also induces a TRIMCyp-independent restriction activity specific to MLV in owl monkey cells.
- Published
- 2008
- Full Text
- View/download PDF
39. [Antiviral activities of interferon and PML pathway].
- Author
-
Dianoux L, Bougrini JE, Galisson F, Percherancier Y, and Chelbi-Alix MK
- Abstract
Discovered in 1957 for their antiviral properties, interferons (IFNs) are a growing cytokine family with diverse biological activities including antitumor and immunoregulatory activities. IFN are classified in three types I, II and III. They bind to different specific cell receptors and induce via the Jak/Stat pathway the expression of more than 300 genes, the products of which are believed to mediate their biological effects. Several proteins have been implicated in resistance to viral infection in IFN-treated cells, i.e. the dsRNAdependent protein kinase PKR, the 2'5' oligoadenylate synthetase/RNaseL and Mx proteins. However, it was demonstrated that cells from triple knockout mice lacking PKR, RNase L and Mx are still sensitive to the IFN-induced antiviral state, indicating that other pathways exist. One of these pathways implicates promyelocytic leukemia (PML) protein. This article reviews the potential antiviral activities of the different IFN-induced mediators focusing onPMLpathway and how viruses from different families overcome this defence.
- Published
- 2008
- Full Text
- View/download PDF
40. Interferon, a growing cytokine family: 50 years of interferon research.
- Author
-
Chelbi-Alix MK and Wietzerbin J
- Subjects
- Animals, Antiviral Agents therapeutic use, Humans, Immunity, Innate, Interferons therapeutic use, Models, Biological, Neoplasms metabolism, Neoplasms therapy, RNA, Double-Stranded chemistry, Signal Transduction, Cytokines metabolism, Interferons metabolism
- Abstract
The establishment of an antiviral state in cells is the defining activity of interferons (IFNs) as well as the property that permitted their discovery in 1957 by Isaacs and Lindenmann. In addition, interferons have other cellular functions that have potential clinical applications. Today, interferons are used for the treatment of a variety of malignancies and viral diseases. The publication of this special issue of Biochimie gives us a great opportunity to review the state of the art in knowledge about interferons and to explore possible future directions. This commentary text will introduce the reviews written by colleagues who are experts in different aspects of interferon research, to mark the 50th anniversary of the discovery of interferon.
- Published
- 2007
- Full Text
- View/download PDF
41. PML and PML nuclear bodies: implications in antiviral defence.
- Author
-
Everett RD and Chelbi-Alix MK
- Subjects
- Antigens, Nuclear metabolism, Autoantigens metabolism, DNA chemistry, DNA Repair, Exons, Gene Expression Regulation, Genome, Viral, Interferons metabolism, Models, Biological, Nuclear Proteins metabolism, Protein Denaturation, Protein Folding, RNA chemistry, Tumor Suppressor Protein p53 metabolism, Antiviral Agents pharmacology, Intranuclear Inclusion Bodies metabolism, Leukemia, Promyelocytic, Acute metabolism
- Abstract
The establishment of an intracellular antiviral state is the defining activity of interferons (IFNs) as well as the property that permitted their discovery. Several pathways have been implicated in resistance to viral infection in IFN-treated cells, one of which implicates the ProMyelocytic Leukaemia (PML) protein and PML nuclear bodies (NBs, also known as ND10). PML NBs are dynamic intranuclear structures that require PML for their formation and which harbour numerous other transiently or permanently localised proteins. PML is expressed as a family of isoforms (PML I-VII) as a result of alternative splicing, most of which are found in the nucleus. IFN treatment directly induces transcription of the genes encoding both PML and Sp100, (another major component of PML NBs), resulting in higher levels of expression of these proteins and increases in both the size and number of PML NBs. These and other observations have encouraged the hypothesis that PML, PML NBs and a number of other constituents of these structures are involved in host antiviral defences. For example, exogenous expression of PML III or PML VI can impede infection by a number of RNA and DNA viruses, and certain viral proteins accumulate in PML NBs then cause their disruption by a variety of mechanisms. Although there are many other functions of PML NBs in a wide range of cellular pathways, there is accumulating evidence that they represent preferential targets for viral infections and that PML plays a role in the mechanism of the antiviral action of IFN. This article reviews the potential antiviral activities of PML NB constituent proteins, how RNA and DNA viruses overcome these defences, and the connections between these events and IFN pathways.
- Published
- 2007
- Full Text
- View/download PDF
42. The nucleocytoplasmic rabies virus P protein counteracts interferon signaling by inhibiting both nuclear accumulation and DNA binding of STAT1.
- Author
-
Vidy A, El Bougrini J, Chelbi-Alix MK, and Blondel D
- Subjects
- Cell Nucleus metabolism, Cytoplasm metabolism, DNA metabolism, Electrophoretic Mobility Shift Assay, Immunohistochemistry, Interferon-Stimulated Gene Factor 3, gamma Subunit antagonists & inhibitors, Interferons metabolism, Microscopy, Confocal, Molecular Chaperones, Protein Binding, STAT1 Transcription Factor metabolism, Signal Transduction, Interferons immunology, Phosphoproteins physiology, Rabies virus physiology, Viral Structural Proteins physiology
- Abstract
Rabies virus P protein inhibits alpha interferon (IFN-alpha)- and IFN-gamma-stimulated Jak-STAT signaling by retaining phosphorylated STAT1 in the cytoplasm. Here, we show that P also blocks an intranuclear step that is the STAT1 binding to the DNA promoter of IFN-responsive genes. As P is a nucleocytoplasmic shuttling protein, we first investigated the effect of the cellular distribution of P on the localization of STAT1 and consequently on IFN signaling. We show that the localization of STAT1 is correlated with the localization of P: in cells expressing a nuclear form of P (the short P3 isoform or the complete P in the presence of the export inhibitor leptomycin B), STAT1 is nuclear, whereas in cells expressing a cytoplasmic form of P, STAT1 is cytoplasmic. However, the expression of nuclear forms of P inhibits the signaling of both IFN-gamma and IFN-alpha, demonstrating that the retention of STAT1 in the cytoplasm is not the only mechanism involved in the inhibition of IFN signaling. Electrophoretic mobility shift analysis indicates that P expression in the cell extracts of infected cells or in stable cell lines prevents IFN-induced DNA binding of STAT1. The loss of the DNA binding of STAT1 and ISGF3 was also observed when purified recombinant P or P3 was added to the extracts of IFN-gamma- or IFN-alpha-treated cells, indicating that P directly affects the DNA binding activity of STAT1. Then products of the rabies virus P gene are able to counteract IFN signaling by creating both cytoplasmic and nuclear blocks for STAT1.
- Published
- 2007
- Full Text
- View/download PDF
43. Arsenic trioxide: A promising novel therapeutic agent for lymphoproliferative and autoimmune syndromes in MRL/lpr mice.
- Author
-
Bobé P, Bonardelle D, Benihoud K, Opolon P, and Chelbi-Alix MK
- Subjects
- Animals, Antibodies, Antinuclear immunology, Antigen-Antibody Complex immunology, Antineoplastic Agents therapeutic use, Apoptosis immunology, Arsenic Trioxide, Arsenicals therapeutic use, Caspases immunology, Cytokines immunology, Drug Evaluation, Preclinical methods, Enzyme Activation drug effects, Enzyme Activation immunology, Glutathione immunology, Humans, Lupus Erythematosus, Systemic immunology, Lymphocyte Activation drug effects, Lymphocyte Activation immunology, Lymphoproliferative Disorders immunology, Mice, Mice, Inbred MRL lpr, Nitric Oxide immunology, Oxidation-Reduction drug effects, Oxides therapeutic use, Syndrome, fas Receptor immunology, Antineoplastic Agents pharmacology, Apoptosis drug effects, Arsenicals pharmacology, Lupus Erythematosus, Systemic drug therapy, Lymphoproliferative Disorders drug therapy, Oxides pharmacology
- Abstract
MRL/lpr mice develop a human lupuslike syndrome and, as in autoimmune lymphoproliferative syndrome (ALPS), massive lymphoproliferation due to inactivation of Fas-mediated apoptosis. Presently, no effective therapy exists for ALPS, and long term, therapies for lupus are hazardous. We show herein that arsenic trioxide (As2O3) is able to achieve quasi-total regression of antibody- and cell-mediated manifestations in MRL/lpr mice. As2O3 activated caspases and eliminated the activated T lymphocytes responsible for lymphoproliferation and skin, lung, and kidney lesions, leading to significantly prolonged survival rates. This treatment also markedly reduced anti-DNA autoantibody, rheumatoid factor, IL-18, IFN-gamma, nitric oxide metabolite, TNF-alpha, Fas ligand, and IL-10 levels and immune-complex deposits in glomeruli. As2O3 restored cellular reduced glutathione levels, thereby limiting the toxic effect of nitric oxide, which is overproduced in MRL/lpr mice. Furthermore, As2O3 protected young animals against developing the syndrome and induced almost total disease disappearance in older affected mice, thereby demonstrating that it is a novel promising therapeutic agent for autoimmune diseases.
- Published
- 2006
- Full Text
- View/download PDF
44. Transcriptional profiling reveals a possible role for the timing of the inflammatory response in determining susceptibility to a viral infection.
- Author
-
Ruby T, Whittaker C, Withers DR, Chelbi-Alix MK, Morin V, Oudin A, Young JR, and Zoorob R
- Subjects
- Animals, Apoptosis, Chickens, DNA, Complementary metabolism, Eimeria tenella metabolism, Gene Expression Profiling, Immune System, Nucleic Acid Hybridization, Oligonucleotide Array Sequence Analysis, Tumor Suppressor Protein p53 metabolism, Genetic Predisposition to Disease, Inflammation, Transcription, Genetic, Virus Diseases etiology, Virus Diseases genetics
- Abstract
Using a novel cDNA microarray prepared from sources of actively responding immune system cells, we have investigated the changes in gene expression in the target tissue during the early stages of infection of neonatal chickens with infectious bursal disease virus. Infections of two lines of chickens previously documented as genetically resistant and sensitive to infection were compared in order to ascertain early differences in the response to infection that might provide clues to the mechanism of differential genetic resistance. In addition to major changes that could be explained by previously described changes in infected tissue, some differences in gene expression on infection, and differences between the two chicken lines, were observed that led to a model for resistance in which a more rapid inflammatory response and more-extensive p53-related induction of apoptosis in the target B cells might limit viral replication and consequent pathology. Ironically, the effect in the asymptomatic neonatal infection is that more-severe B-cell depletion is seen in the more genetically resistant chicken. Changes of expression of many chicken genes of unknown function, indicating possible roles in the response to infection, may aid in the functional annotation of these genes.
- Published
- 2006
- Full Text
- View/download PDF
45. Cross talk between PML and p53 during poliovirus infection: implications for antiviral defense.
- Author
-
Pampin M, Simonin Y, Blondel B, Percherancier Y, and Chelbi-Alix MK
- Subjects
- Apoptosis, Cell Line, Tumor, Cell Nucleus Structures metabolism, Humans, Organelles metabolism, Phosphorylation, Promyelocytic Leukemia Protein, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Small Ubiquitin-Related Modifier Proteins metabolism, Tumor Suppressor Protein p53 genetics, Virus Replication, Neoplasm Proteins metabolism, Nuclear Proteins metabolism, Poliovirus pathogenicity, Transcription Factors metabolism, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins metabolism
- Abstract
PML nuclear bodies (NBs) are dynamic intranuclear structures harboring numerous transiently or permanently localized proteins. PML, the NBs' organizer, is directly induced by interferon, and its expression is critical for antiviral host defense. We describe herein the molecular events following poliovirus infection that lead to PML-dependent p53 activation and protection against virus infection. Poliovirus infection induces PML phosphorylation through the extracellular signal-regulated kinase pathway, increases PML SUMOylation, and induces its transfer from the nucleoplasm to the nuclear matrix. These events result in the recruitment of p53 to PML NBs, p53 phosphorylation on Ser15, and activation of p53 target genes leading to the induction of apoptosis. Moreover, the knock-down of p53 by small interfering RNA results in higher poliovirus replication, suggesting that p53 participates in antiviral defense. This effect, which requires the presence of PML, is transient since poliovirus targets p53 by inducing its degradation in a proteasome- and MDM2-dependent manner. Our results provide evidence of how poliovirus counteracts p53 antiviral activity by regulating PML and NBs, thus leading to p53 degradation.
- Published
- 2006
- Full Text
- View/download PDF
46. The exonuclease ISG20 mainly localizes in the nucleolus and the Cajal (Coiled) bodies and is associated with nuclear SMN protein-containing complexes.
- Author
-
Espert L, Eldin P, Gongora C, Bayard B, Harper F, Chelbi-Alix MK, Bertrand E, Degols G, and Mechti N
- Subjects
- Cell Nucleolus drug effects, Cell Nucleolus ultrastructure, Coiled Bodies drug effects, Coiled Bodies ultrastructure, Exoribonucleases, HeLa Cells, Humans, Interferons pharmacology, Microscopy, Immunoelectron, Protein Binding, RNA, Small Nuclear metabolism, SMN Complex Proteins, Cell Nucleolus enzymology, Coiled Bodies enzymology, Cyclic AMP Response Element-Binding Protein metabolism, Exonucleases metabolism, Nerve Tissue Proteins metabolism, RNA-Binding Proteins metabolism
- Abstract
We have previously shown that ISG20, an interferon (IFN)-induced gene, encodes a 3' to 5' exoribonuclease member of the DEDD superfamily of exonucleases. ISG20 specifically degrades single-stranded RNA. In this report, using immunofluorescence analysis, we demonstrate that in addition to a diffuse cytoplasmic and nucleoplasmic localization, the endogenous ISG20 protein was present in the nucleus both in the nucleolus and in the Cajal bodies (CBs). In addition, we show that the ectopic expression of the CBs signature protein, coilin, fused to the red fluorescent protein (coilin-dsRed) increased the number of nuclear dots containing both ISG20 and coilin-dsRed. Using electron microcopy analysis, ISG20 appeared principally concentrated in the dense fibrillar component of the nucleolus, the major site for rRNA processing. We also present evidences that ISG20 was associated with survival of motor neuron (SMN)-containing macromolecular nuclear complexes required for the biogenesis of various small nuclear ribonucleoproteins. Finally, we demonstrate that ISG20 was associated with U1 and U2 snRNAs, and U3 snoRNA. The accumulation of ISG20 in the CBs after IFN treatment strongly suggests its involvement in a new route for IFN-mediated inhibition of protein synthesis by modulating snRNA and rRNA maturation., ((c) 2006 Wiley-Liss, Inc.)
- Published
- 2006
- Full Text
- View/download PDF
47. Arsenic enhances the apoptosis induced by interferon gamma: key role of IRF-1.
- Author
-
El Bougrini J, Pampin M, and Chelbi-Alix MK
- Subjects
- Amino Acid Motifs, Cell Cycle Proteins metabolism, Cell Proliferation drug effects, GPI-Linked Proteins, Gene Expression drug effects, Growth Inhibitors pharmacology, Humans, Interferon Regulatory Factor-1 metabolism, Membrane Proteins metabolism, Models, Biological, Phosphorylation drug effects, Protein Binding drug effects, STAT1 Transcription Factor metabolism, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols pharmacology, Apoptosis drug effects, Arsenic pharmacology, Interferon Regulatory Factor-1 physiology, Interferon-gamma pharmacology
- Abstract
Interferons (IFNs) and arsenic trioxide (As2O3) are known inhibitors of cell proliferation and have been used in the treatment of certain forms of malignancy. IFNgamma treatment of cells leads to tyrosine phosphorylation of STAT1 followed by dimerization that accumulates in the nucleus. This is followed by DNA binding, activation of target gene transcription, dephosphorylation, and return to the cytoplasm. We have shown earlier that IFNgamma and As2O3 act synergistically in acute promyelocytic leukemia cells to upregulate IRF-1 expression and to induce apoptosis. Here, we show that in the human fibrosarcoma cell line 2fTGH, As2O3 prolongs IFNgamma-induced STAT1 phosphorylation resulting in persistent binding of STAT1 to GAS motif leading to an increase in IRF-1 expression which correlated with both higher anti-proliferative effect and increased apoptosis. These biological responses induced by IFNgamma alone or in combination with As2O3 were abolished when IRF-1 expression was down-regulated by RNA interference, thus demonstrating the key role of IRF-1.
- Published
- 2006
48. Rabies viral mechanisms to escape the IFN system: the viral protein P interferes with IRF-3, Stat1, and PML nuclear bodies.
- Author
-
Chelbi-Alix MK, Vidy A, El Bougrini J, and Blondel D
- Subjects
- Animals, Humans, Interferons immunology, Rabies virology, Rhabdoviridae immunology, Cell Nucleus metabolism, Interferons metabolism, Rabies metabolism, Rhabdoviridae physiology, Transcription Factors metabolism, Viral Proteins metabolism
- Abstract
Interferons (IFNs) are a family of secreted proteins with antiviral, antiproliferative, and immunomodulatory activities. The different biologic actions of IFN are believed to be mediated by the products of specifically IFN-stimulated genes (ISG) in the target cells. The IFN response is the first line of defense against viral infections. Viruses, which require the cellular machinery for their replication, have evolved different ways to counteract the action of IFN by inhibiting IFN production or Jak-Stat signaling or by altering ISG products. This review focuses on the role of viral proteins from the RNA virus family, particularly rabies P protein. P protein mediates inhibition of the IFN system by different pathways: it inhibits IFN production by impairing IFN regulatory factor-3 (IRF-3) phosphorylation and IFN signaling by blocking nuclear transport of Stat1 and alters promyelocytic leukemia (PML) nuclear bodies by retaining PML in the cytoplasm.
- Published
- 2006
- Full Text
- View/download PDF
49. [Negative regulation of the JAK/STAT: pathway implication in tumorigenesis].
- Author
-
Espert L, Dusanter-Fourt I, and Chelbi-Alix MK
- Subjects
- Apoptosis, Cell Differentiation, Cell Proliferation, Cytokines metabolism, Enzyme Activation, JNK Mitogen-Activated Protein Kinases genetics, Phosphorylation, Poly-ADP-Ribose Binding Proteins, Protein Inhibitors of Activated STAT metabolism, Protein Tyrosine Phosphatases metabolism, STAT Transcription Factors genetics, Suppressor of Cytokine Signaling Proteins metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Neoplasms metabolism, STAT Transcription Factors metabolism
- Abstract
Cytokines, hormones or growth factors induce a variety of biological responses including proliferation, differentiation and apoptosis. After binding to their specific cell surface receptors, these stimuli induce the activation of a number of signaling pathways including the activation of JAK (JAnus Kinase) proteins by auto and transphosphorylation. Activated JAK phosphorylate the receptor chains on tyrosines, creating docking sites for cytoplasmic transcription factors named STAT (Signal Transducers and Activators of Transcription). Furthermore, the JAK phosphorylate the STAT which form dimers and migrate to the nucleus where they bind to specific DNA sequences leading to the activation of transcription. The multiplicity of JAK (4 members) and STAT (7 members) and their associations with multiple possible partners allow the formation of various STAT homo and heterodimers and STAT-containing transcriptional complexes. Each of these complexes lead to the specific regulation of gene transcription. Negative regulation of the JAK/STAT signaling pathway is crucial to switch off the cytokine/growth factors' signal. Three families of proteins : the phosphotyrosine phosphatases (SHPs, CD45, PTP1B/TC-PTP), the SOCS proteins (Suppressors Of Cytokine Signaling) and the PIAS (Protein Inhibitor of Activated STAT) are involved in this process. These proteins act at various levels of the JAK/STAT pathway. Thus, tyrosine-phosphatases dephosphorylate activated JAK, STAT or cytokine receptors. PIAS interact with activated STAT and inhibit their DNA binding or their transactivating capacity, probably in relation with their intrinsic SUMO E3-ligase activity. The tyrosine phosphatases and the PIAS are constitutively present in the cell and represent a first level of regulation. The SOCS, which represent a second level of JAK/STAT negative control, are induced by cytokines and exert a negative feed-back loop. Indeed, they interact with activated JAK or with phosphorylated receptors, inhibiting the recruitment of STAT, the activation of the JAK enzymatic activity, or inducing the proteasome-dependant degradation of activated JAK or receptors.
- Published
- 2005
50. Interferons alpha and gamma induce p53-dependent and p53-independent apoptosis, respectively.
- Author
-
Porta C, Hadj-Slimane R, Nejmeddine M, Pampin M, Tovey MG, Espert L, Alvarez S, and Chelbi-Alix MK
- Subjects
- Cell Line, Tumor, Cell Nucleus metabolism, DNA-Binding Proteins metabolism, Humans, Interferon Regulatory Factor-1, Neoplasm Proteins metabolism, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Nuclear Proteins genetics, Nuclear Proteins metabolism, Phosphoproteins metabolism, Promyelocytic Leukemia Protein, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, Proto-Oncogene Proteins c-mdm2, Transcription Factors metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Proteins, Up-Regulation drug effects, fas Receptor genetics, fas Receptor metabolism, Apoptosis drug effects, Interferon-alpha pharmacology, Interferon-gamma pharmacology, Signal Transduction drug effects, Tumor Suppressor Protein p53 metabolism
- Abstract
Type I interferon (IFN) enhances the transcription of the tumor suppressor gene p53. To elucidate the molecular mechanism mediating IFN-induced apoptosis, we analysed programmed cell death in response to type I (IFNalpha) or type II (IFNgamma) treatment in relation to p53 status. In two cell lines (MCF-7, SKNSH), IFNalpha, but not IFNgamma, enhanced apoptosis in a p53-dependent manner. Furthermore, only IFNalpha upregulated p53 as well as p53 target genes (Noxa, Mdm2 and CD95). The apoptotic response to IFNalpha decreased in the presence of ZB4, an anti-CD95 antibody, suggesting that CD95 is involved in this process. When p53 was inactivated by the E6 viral protein or the expression of a p53 mutant, IFNalpha-induced apoptosis and p53 target genes upregulation were abrogated. Altogether these results demonstrate that p53 plays a pivotal role in the IFNalpha-induced apoptotic response. IFNalpha-induced PML was unable to recruit p53 into nuclear bodies and its downregulation by siRNA did not alter CD95 expression. In contrast, IFNgamma-induced apoptosis is p53-independent. CD95 and IFN-regulatory factor 1 (IRF1) are directly upregulated by this cytokine. Apoptotic response to IFNgamma is decreased in the presence of ZB4 and strongly diminished by IRF1 siRNA, implicating both CD95 and IRF1 in IFNgamma-induced apoptotic response. Taken together, these results show that in two different cell lines, IFNalpha and IFNgamma, induce p53-dependent -independent apoptosis, respectively.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.