Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 9: 671067, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33968942

RESUMO

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.

2.
Cytokine Growth Factor Rev ; 55: 37-47, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32591223

RESUMO

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.


Assuntos
Interferons , Sumoilação , Antivirais , Humanos , Proteína da Leucemia Promielocítica/metabolismo , Proteína SUMO-1
3.
Cytokine ; 129: 155025, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32044670

RESUMO

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.


Assuntos
Interferon-alfa/farmacologia , Sumoilação/efeitos dos fármacos , Antivirais/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinas/metabolismo
4.
Med Sci (Paris) ; 36(1): 50-56, 2020 Jan.
Artigo em Francês | MEDLINE | ID: mdl-32014098

RESUMO

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


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


Assuntos
Proteína da Leucemia Promielocítica/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Humanos , Camundongos , Proteína da Leucemia Promielocítica/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/farmacologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
5.
Cytokine ; 120: 264-272, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31153006

RESUMO

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


Assuntos
Núcleo Celular/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Proteólise , Sumoilação , Fator de Crescimento Transformador beta/farmacologia , Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Células HEK293 , Humanos , Interferon-alfa/farmacologia , Matriz Nuclear/metabolismo , Proteína da Leucemia Promielocítica/química , Ligação Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação/efeitos dos fármacos
6.
Viruses ; 10(12)2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30513968

RESUMO

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.


Assuntos
Infecções por Rhabdoviridae/imunologia , Rhabdoviridae/imunologia , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Humanos , Imunidade Inata , Interferons/imunologia , Camundongos , Proteínas de Resistência a Myxovirus/genética , Ligação Proteica , Vírus da Raiva/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/imunologia , Sumoilação , Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Vírus da Estomatite Vesicular Indiana/imunologia , eIF-2 Quinase/genética
8.
Mol Cell Proteomics ; 17(6): 1196-1208, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29535160

RESUMO

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.


Assuntos
Interferon-alfa/farmacologia , Proteína da Leucemia Promielocítica/metabolismo , Sumoilação/efeitos dos fármacos , Células HEK293 , Humanos
9.
Sci Rep ; 8(1): 1277, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29352251

RESUMO

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.


Assuntos
Proteína SUMO-1/metabolismo , Ubiquitinas/metabolismo , eIF-2 Quinase/metabolismo , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Estabilidade Enzimática , Células HeLa , Humanos , Fosforilação , Proteína SUMO-1/genética , Sumoilação , Ubiquitinas/genética
10.
Nat Commun ; 8: 14109, 2017 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-28098164

RESUMO

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.


Assuntos
Cromatografia de Afinidade/métodos , Peptídeos/química , Proteínas/metabolismo , Motivos de Aminoácidos , Células HEK293 , Humanos , Proteína da Leucemia Promielocítica/química , Proteína da Leucemia Promielocítica/genética , Proteína da Leucemia Promielocítica/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Mapas de Interação de Proteínas , Proteínas/química , Proteínas/genética , Proteólise , Sumoilação , Ubiquitina/química , Ubiquitinação
11.
J Virol ; 90(14): 6598-6610, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27170750

RESUMO

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.


Assuntos
Interferon-alfa/farmacologia , Proteínas de Resistência a Myxovirus/farmacologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/farmacologia , Estomatite Vesicular/prevenção & controle , Vírus da Estomatite Vesicular Indiana/fisiologia , Antivirais/farmacologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/virologia , Células HeLa , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Processamento de Proteína Pós-Traducional , Raiva/metabolismo , Raiva/prevenção & controle , Raiva/virologia , Vírus da Raiva/fisiologia , Células Tumorais Cultivadas , Estomatite Vesicular/metabolismo , Estomatite Vesicular/virologia
12.
Cytokine Growth Factor Rev ; 29: 3-16, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27157810

RESUMO

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.


Assuntos
Imunidade Inata , Proteína SUMO-1/imunologia , Transdução de Sinais/imunologia , Viroses/imunologia , Animais , Humanos , Janus Quinases/imunologia , Fatores de Transcrição STAT/imunologia
14.
PLoS Pathog ; 11(11): e1005280, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26566030

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Núcleo Celular/metabolismo , Proteínas Correpressoras , HIV-1/metabolismo , Humanos , Chaperonas Moleculares , Proteína da Leucemia Promielocítica , Ligação Proteica/fisiologia , Transcrição Gênica
15.
J Immunol ; 195(5): 2312-24, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26223657

RESUMO

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.


Assuntos
Interferons/farmacologia , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Western Blotting , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Expressão Gênica/efeitos dos fármacos , Células HeLa , Células Hep G2 , Humanos , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Microscopia Confocal , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilação/efeitos dos fármacos , Proteína da Leucemia Promielocítica , Ligação Proteica/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Proteína SUMO-1/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Sumoilação/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinas/genética
16.
Viruses ; 7(7): 3675-702, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26198243

RESUMO

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.


Assuntos
Infecções por Rhabdoviridae/imunologia , Rhabdoviridae/fisiologia , Animais , Humanos , Interferons/genética , Interferons/imunologia , Rhabdoviridae/genética , Infecções por Rhabdoviridae/genética , Infecções por Rhabdoviridae/virologia
17.
Exp Cell Res ; 330(1): 151-63, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25447205

RESUMO

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.


Assuntos
Proteínas de Resistência a Myxovirus/metabolismo , Sumoilação , Motivos de Aminoácidos , Animais , Sítios de Ligação , Células HeLa , Humanos , Camundongos , Proteínas de Resistência a Myxovirus/química , Células NIH 3T3 , Ligação Proteica , Proteína SUMO-1/química , Proteína SUMO-1/metabolismo , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/metabolismo
18.
Nat Commun ; 5: 5409, 2014 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-25391492

RESUMO

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.


Assuntos
Lisina/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Animais , Anticorpos Monoclonais/imunologia , Cromatografia de Afinidade , Cromatografia Gasosa-Espectrometria de Massas , Células HEK293 , Humanos , Hibridomas/metabolismo , Peptídeos/imunologia , Peptídeos/metabolismo , Proteoma/metabolismo , Coelhos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/imunologia , Proteínas Supressoras de Tumor/imunologia , Proteínas Supressoras de Tumor/metabolismo
19.
Biochimie ; 107 Pt B: 247-56, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25241256

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Arsenitos/farmacologia , Herpesvirus Humano 4/efeitos dos fármacos , Compostos de Sódio/farmacologia , Antígenos Nucleares/metabolismo , Trióxido de Arsênio , Arsenicais/farmacologia , Autoantígenos/metabolismo , Autofagia/efeitos dos fármacos , Linfoma de Burkitt/patologia , Linfoma de Burkitt/virologia , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Herpesvirus Humano 4/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Óxidos/farmacologia , Proteína da Leucemia Promielocítica , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas da Matriz Viral/metabolismo
20.
Med Sci (Paris) ; 30(8-9): 765-71, 2014.
Artigo em Francês | MEDLINE | ID: mdl-25174753

RESUMO

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


Assuntos
Imunidade Adaptativa , Núcleo Celular/metabolismo , Imunidade Inata , Corpos de Inclusão/fisiologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Imunidade Adaptativa/genética , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunidade Inata/genética , Corpos de Inclusão/metabolismo , Interferons/farmacologia , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Viroses/genética , Viroses/imunologia , Viroses/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...