首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
In acute promyelocytic leukemia (APL), the promyelocytic leukemia (PML) protein is fused to the retinoic acid receptor alpha (RAR). Arsenic is an effective treatment for this disease as it induces SUMO-dependent ubiquitin-mediated proteasomal degradation of the PML-RAR fusion protein. Here we analyze the nuclear trafficking dynamics of PML and its SUMO-dependent ubiquitin E3 ligase, RNF4 in response to arsenic. After administration of arsenic, PML immediately transits into nuclear bodies where it undergoes SUMO modification. This initial recruitment of PML into nuclear bodies is not dependent on RNF4, but RNF4 quickly follows PML into the nuclear bodies where it is responsible for ubiquitylation of SUMO-modified PML and its degradation by the proteasome. While arsenic restricts the mobility of PML, FRAP analysis indicates that RNF4 continues to rapidly shuttle into PML nuclear bodies in a SUMO-dependent manner. Under these conditions FRET studies indicate that RNF4 interacts with SUMO in PML bodies but not directly with PML. These studies indicate that arsenic induces the rapid reorganization of the cell nucleus by SUMO modification of nuclear body-associated PML and uptake of the ubiquitin E3 ligase RNF4 leading to the ubiquitin-mediated degradation of PML.  相似文献   

2.
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.  相似文献   

3.
Components of promyelocytic leukaemia (PML) nuclear bodies (ND10) are recruited to sites associated with herpes simplex virus type 1 (HSV-1) genomes soon after they enter the nucleus. This cellular response is linked to intrinsic antiviral resistance and is counteracted by viral regulatory protein ICP0. We report that the SUMO interaction motifs of PML, Sp100 and hDaxx are required for recruitment of these repressive proteins to HSV-1 induced foci, which also contain SUMO conjugates and PIAS2β, a SUMO E3 ligase. SUMO modification of PML and elements of its tripartite motif (TRIM) are also required for recruitment in cells lacking endogenous PML. Mutants of PML isoform I and hDaxx that are not recruited to virus induced foci are unable to reproduce the repression of ICP0 null mutant HSV-1 infection mediated by their wild type counterparts. We conclude that recruitment of ND10 components to sites associated with HSV-1 genomes reflects a cellular defence against invading pathogen DNA that is regulated through the SUMO modification pathway.  相似文献   

4.
Intrinsic antiviral resistance represents the first line of intracellular defence against virus infection. During herpes simplex virus type-1 (HSV-1) infection this response can lead to the repression of viral gene expression but is counteracted by the viral ubiquitin ligase ICP0. Here we address the mechanisms by which ICP0 overcomes this antiviral response. We report that ICP0 induces the widespread proteasome-dependent degradation of SUMO-conjugated proteins during infection and has properties related to those of cellular SUMO-targeted ubiquitin ligases (STUbLs). Mutation of putative SUMO interaction motifs within ICP0 not only affects its ability to degrade SUMO conjugates, but also its capacity to stimulate HSV-1 lytic infection and reactivation from quiescence. We demonstrate that in the absence of this viral countermeasure the SUMO conjugation pathway plays an important role in mediating intrinsic antiviral resistance and the repression of HSV-1 infection. Using PML as a model substrate, we found that whilst ICP0 preferentially targets SUMO-modified isoforms of PML for degradation, it also induces the degradation of PML isoform I in a SUMO modification-independent manner. PML was degraded by ICP0 more rapidly than the bulk of SUMO-modified proteins in general, implying that the identity of a SUMO-modified protein, as well as the presence of SUMO modification, is involved in ICP0 targeting. We conclude that ICP0 has dual targeting mechanisms involving both SUMO- and substrate-dependent targeting specificities in order to counteract intrinsic antiviral resistance to HSV-1 infection.  相似文献   

5.
The small ubiquitin related modifier SUMO regulates protein functions to maintain cell homeostasis. SUMO attachment is executed by the hierarchical action of E1, E2 and E3 enzymes of which E3 ligases ensure substrate specificity. We recently identified the ZNF451 family as novel class of SUMO2/3 specific E3 ligases and characterized their function in SUMO chain formation. The founding member, ZNF451 isoform1 (ZNF451-1) partially resides in PML bodies, nuclear structures organized by the promyelocytic leukemia gene product PML. As PML and diverse PML components are well known SUMO substrates the question arises whether ZNF451-1 is involved in their sumoylation. Here, we show that ZNF451-1 indeed functions as SUMO2/3 specific E3 ligase for PML and selected PML components in vitro. Mutational analysis indicates that substrate sumoylation employs an identical biochemical mechanism as we described for SUMO chain formation. In vivo, ZNF451-1 RNAi depletion leads to PML stabilization and an increased number of PML bodies. By contrast, PML degradation upon arsenic trioxide treatment is not ZNF451-1 dependent. Our data suggest a regulatory role of ZNF451-1 in fine-tuning physiological PML levels in a RNF4 cooperative manner in the mouse neuroblastoma N2a cell-line.  相似文献   

6.
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.Promyelocytic leukemia protein (PML)5 is a tumor suppressor (1) whose gene is translocated in cases of acute promyelocytic leukemia (2). PML functions as the organizer of PML NBs, which are dynamic structures harboring numerous transiently and permanently localized proteins (3). The importance of PML NB structural integrity was first revealed in acute promyelocytic leukemia because, in this malignancy, the abnormal fusion protein PML/RARα leads to NB disruption. Patient treatment with As2O3 induces the reversion of the acute promyelocytic leukemia phenotype as well as PML/RARα degradation and PML NB reformation (4).PML is a target for the post-translational modification by SUMO, an ubiquitin-like protein that is covalently coupled to PML lysine residues 65, 160, and 490 via a process called sumoylation (5, 6). Among the four human SUMO paralogs identified, SUMO1, -2, and -3 were found to be conjugated to target proteins. It involves an enzymatic cascade for the transfer of the mature SUMO and the formation of an isopeptide bond between the COOH-terminal glycine of SUMO and a lysine from the target protein. Sumoylation is a reversible process due to the existence of several deconjugating enzymes.PML NB formation requires both the covalent linkage (sumoylation) (reviewed in Ref. 7) and the non-covalent interactions of SUMO with PML through a SUMO binding domain (SBD also named SIM for SUMO interacting motif) (8). Interestingly, PML SBD contains specific serines, acting as substrates for the caseine kinase-2 (CK2), which are implicated in PML ubiquitination and degradation (9) and which phosphorylation status could regulate the function of the SBD.Because sumoylation of proteins is dynamic and reversible, this post-translational modification is difficult to follow in vivo and its detection mainly relies on the identification of sumoylated protein species by Western blot following cell lysis. Recently, we used bioluminescence resonance energy transfer (BRET) to detect covalent linkage of ubiquitin (ubiquitination) in living mammalian cells and in real time (10). In brief, BRET monitors the interaction between a protein fused to a luciferase and a protein fused to yellow or green fluorescent protein (YFP or GFP), upon addition of a luciferase substrate; it is a proximity-based assay that requires that the donor of energy (luciferase fusion) and the acceptor (YFP or GFP fusions) are within 50 to 100 Å for an efficient energy transfer (1113). However, a demonstration that BRET may provide a method of choice to follow the dynamics of protein sumoylation in living cells is lacking. Here, we developed a sensitive and quantitative SUMO BRET assay for the detection of PML interactions with SUMO in living cells. We proved that BRET can be used to detect both SUMO covalent and non-covalent interactions with PML (model, Fig. 1H). For this purpose, we used the PMLIII isoform in which sumoylation is induced by As2O3 and triggers a proteasome-dependent PML degradation (14); the degradation process involves the ubiquitination of poly-SUMO covalently coupled to PML by the poly-SUMO-specific E3 ubiquitin ligase RNF4 (1517). Altogether, our BRET results indicate that, As2O3 and/or RNF4-induced PML degradation are dependent on the integrity of both PML sumoylation target sites and the PML SBD core sequence but not on the CK2 serine phosphorylation sites within the SBD. However, phosphorylation of these serines is required for most PML SBD-dependent non-covalent interactions. This phospho-regulation of PML SBD (“SBD phospho-switch”) establishes another link between the phosphorylation and SUMO, different from the phospho-sumoyl switch (18).Open in a separate windowFIGURE 1.BRET reveals both covalent and non-covalent PML/SUMO1 interactions as well as As2O3-induced PML sumoylation in living cells. A and B, detection of PML/SUMO1 interactions by BRET1 (A) or BRET2 (B) titration assays using HEK293T cells transfected for expression of increasing amounts of YFP-SUMO1 (BRET1) or GFP-SUMO1 (BRET2) and a fixed amount of Luc fusion. Negative controls: BRET pairs including PMLC57,60A-Luc (a non-sumoylatable mutant with Cys57 and Cys60 mutated to Ala) or YFP-SUMO1G (a SUMO1 that cannot be processed) (dotted line) (A) and Luc fused to a NLS (B). C and D, detection of covalent and non-covalent PML/SUMO1 interactions by BRET1 (C) or BRET2 (D) titration assays in the presence (dotted lines, empty symbols) or absence (solid lines and symbols) of As2O3 in HEK293T cells transfected for expression of PMLWT-Luc or its sumoylation deficient mutant PML3K-Luc in pairs with either YFP-SUMO1 (BRET1) or GFP-SUMO1 (BRET2). Negative control: PMLWT-Luc in pairs with YFP-SUMO1G. E, kinetics of As2O3-induced PMLWT-Luc sumoylation revealed by BRET1 (assay on attached cells) and BRET2 (cells in suspension). F, dose-response curve to As2O3 treatment for PMLWT-Luc or PML3KR-Luc/YFP-SUMO1 BRET1 pairs. Negative control: PMLC57,60A-Luc/YFP-SUMO1. G, comparison of As2O3-induced sumoylation of PMLWT, PML3KR, and its single lysine mutants at an identical YFP acceptor/Luc donor expression ratio as derived from titration curves. As2O3 treatment (C–G): 5 μm, 4 h exposure for BRET1 and Western blot or 10 μm, 70-min exposure for BRET2. H, model for the covalent (sumoylation) and non-covalent interactions between a tested protein fused to Luc and SUMO fused to a fluorescent protein (YFP) that generates a BRET signal. The black arrows indicate the bioluminescent transfer of energy (or BRET) that occurs between Luc and GFP fusion upon exposure to the cell-permeable luciferase substrate.  相似文献   

7.
8.
9.
Two articles in a recent issue of Molecular Cell (Shen et al., 2006; Lin et al., 2006) demonstrate that noncovalent interactions between the SUMO moieties of SUMO-modified PML, and SUMO binding motifs on PML and other PML nuclear-body-associated proteins, affect the assembly of PML nuclear bodies and the recruitment of proteins in and out of these subnuclear structures.  相似文献   

10.
In acute promyelocytic leukaemia (APL), arsenic trioxide induces degradation of the fusion protein encoded by the PML-RARA oncogene, differentiation of leukaemic cells and produces clinical remissions. SUMOylation of its PML moiety was previously implicated, but the nature of the degradation pathway involved and the role of PML-RARalpha catabolism in the response to therapy have both remained elusive. Here, we demonstrate that arsenic-induced PML SUMOylation triggers its Lys 48-linked polyubiquitination and proteasome-dependent degradation. When exposed to arsenic, SUMOylated PML recruits RNF4, the human orthologue of the yeast SUMO-dependent E3 ubiquitin-ligase, as well as ubiquitin and proteasomes onto PML nuclear bodies. Arsenic-induced differentiation is impaired in cells transformed by a non-degradable PML-RARalpha SUMOylation mutant or in APL cells transduced with a dominant-negative RNF4, directly implicating PML-RARalpha catabolism in the therapeutic response. We thus identify PML as the first protein degraded by SUMO-dependent polyubiquitination. As PML SUMOylation recruits not only RNF4, ubiquitin and proteasomes, but also many SUMOylated proteins onto PML nuclear bodies, these domains could physically integrate the SUMOylation, ubiquitination and degradation pathways.  相似文献   

11.
早幼粒白血病蛋白核体(promyelocytic leukaemia nuclear bodies,PMLNBs)是哺乳动物细胞中普遍存在的一种亚核结构,广泛参与如转录调节、基因组稳定性维持、抗病毒、细胞凋亡、肿瘤抑制等一系列的生物学事件.SUMO(smallubiquitinmodifier)修饰是蛋白质翻译后修饰领域中的研究热点,SUMO修饰对PML核体的形成与降解都发挥着重要作用.近年来研究发现,人的E3泛素连接酶RNF4(RING finger protein4),可促进依赖SUMO-2/3修饰的PML核体的泛素化连接,并且ATO(三氧化二砷)可加速其对PML核体的降解.荧光共振能量转移(fluorescence resonance energy transfer,FRET)技术可完全应用于活细胞内PML核体和SUMO蛋白之间在时间和空间上的精确互作.因此,更深入地研究PML核体形成和降解的机理以及在这个过程中重要蛋白质之间的相互作用具有重要而深远的意义.  相似文献   

12.
The cell nucleus harbors a variety of different bodies that vary in number, composition, and size. Although these bodies coordinate important nuclear processes, little is known about how they are formed. Among the most intensively studied bodies in recent years is the PML body. These bodies have been implicated in gene regulation and other cellular processes and are disrupted in cells from patients suffering from acute promyelocytic leukemia. Using live cell imaging microscopy and immunofluorescence, we show in several cell types that PML bodies are formed at telomeric DNA during interphase. Recent studies revealed that both SUMO modification sites and SUMO interaction motifs in the promyelocytic leukemia (PML) protein are required for PML body formation. We show that SMC5, a component of the SUMO ligase MMS21-containing SMC5/6 complex, localizes temporarily at telomeric DNA during PML body formation, suggesting a possible role for SUMO in the formation of PML bodies at telomeric DNA. Our data identify a novel role of telomeric DNA during PML body formation.  相似文献   

13.
The promyelocytic leukemia (PML) protein organizes PML nuclear bodies (NBs), which are stress-responsive domains where many partner proteins accumulate. Here, we clarify the basis for NB formation and identify stress-induced partner sumoylation as the primary NB function. NB nucleation does not rely primarily on intermolecular interactions between the PML SUMO-interacting motif (SIM) and SUMO, but instead results from oxidation-mediated PML multimerization. Oxidized PML spherical meshes recruit UBC9, which enhances PML sumoylation, allow partner recruitment through SIM interactions, and ultimately enhance partner sumoylation. Intermolecular SUMO–SIM interactions then enforce partner sequestration within the NB inner core. Accordingly, oxidative stress enhances NB formation and global sumoylation in vivo. Some NB-associated sumoylated partners also become polyubiquitinated by RNF4, precipitating their proteasomal degradation. As several partners are protein-modifying enzymes, NBs could act as sensors that facilitate and confer oxidative stress sensitivity not only to sumoylation but also to other post-translational modifications, thereby explaining alterations of stress response upon PML or NB loss.  相似文献   

14.
15.
Herpes simplex virus 1 (HSV-1) immediate-early protein ICP0 localizes to cellular structures known as promyelocytic leukemia protein (PML) nuclear bodies or ND10 and disrupts their integrity by inducing the degradation of PML. There are six PML isoforms with different C-terminal regions in ND10, of which PML isoform I (PML.I) is the most abundant. Depletion of all PML isoforms increases the plaque formation efficiency of ICP0-null mutant HSV-1, and reconstitution of expression of PML.I and PML.II partially reverses this improved replication. ICP0 also induces widespread degradation of SUMO-conjugated proteins during HSV-1 infection, and this activity is linked to its ability to counteract cellular intrinsic antiviral resistance. All PML isoforms are highly SUMO modified, and all such modified forms are sensitive to ICP0-mediated degradation. However, in contrast to the situation with the other isoforms, ICP0 also targets PML.I that is not modified by SUMO, and PML in general is degraded more rapidly than the bulk of other SUMO-modified proteins. We report here that ICP0 interacts with PML.I in both yeast two-hybrid and coimmunoprecipitation assays. This interaction is dependent on PML.I isoform-specific sequences and the N-terminal half of ICP0 and is required for SUMO-modification-independent degradation of PML.I by ICP0. Degradation of the other PML isoforms by ICP0 was less efficient in cells specifically depleted of PML.I. Therefore, ICP0 has two distinct mechanisms of targeting PML: one dependent on SUMO modification and the other via SUMO-independent interaction with PML.I. We conclude that the ICP0-PML.I interaction reflects a countermeasure to PML-related antiviral restriction.  相似文献   

16.
17.
Acute promyelocytic leukemia (APL) is driven by a chromosomal translocation whose product, the PML/retinoic acid (RA) receptor α (RARA) fusion protein, affects both nuclear receptor signaling and PML body assembly. Dissection of APL pathogenesis has led to the rediscovery of PML bodies and revealed their role in cell senescence, disease pathogenesis, and responsiveness to treatment. APL is remarkable because of the fortuitous identification of two clinically effective therapies, RA and arsenic, both of which degrade PML/RARA oncoprotein and, together, cure APL. Analysis of arsenic-induced PML or PML/RARA degradation has implicated oxidative stress in the biogenesis of nuclear bodies and SUMO in their degradation.  相似文献   

18.
The mechanisms of PML-nuclear body formation   总被引:1,自引:0,他引:1  
  相似文献   

19.
20.
The attachment of the SUMO modifier to proteins controls cellular signaling pathways through noncovalent binding to SUMO-interaction motifs (SIMs). Canonical SIMs contain a core of hydrophobic residues that bind to a hydrophobic pocket on SUMO. Negatively charged residues of SIMs frequently contribute to binding by interacting with a basic surface on SUMO. Here we define acetylation within this basic interface as a central mechanism for the control of SUMO-mediated interactions. The acetyl-mediated neutralization of basic charges on SUMO prevents binding to SIMs in PML, Daxx, and PIAS family members but does not affect the interaction between RanBP2 and SUMO. Acetylation is controlled by HDACs and attenuates SUMO- and PIAS-mediated gene silencing. Moreover, it affects the assembly of PML nuclear bodies and restrains the recruitment of the corepressor Daxx to these structures. This acetyl-dependent switch thus expands the regulatory repertoire of SUMO signaling and determines the selectivity and dynamics of SUMO-SIM interactions.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号