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











Publication year range
1.
Cancer Discov ; 13(12): 2548-2565, 2023 12 12.
Article in English | MEDLINE | ID: mdl-37655965

ABSTRACT

PML nuclear bodies (NB) are disrupted in PML-RARA-driven acute promyelocytic leukemia (APL). Arsenic trioxide (ATO) cures 70% of patients with APL, driving PML-RARA degradation and NB reformation. In non-APL cells, arsenic binding onto PML also amplifies NB formation. Yet, the actual molecular mechanism(s) involved remain(s) elusive. Here, we establish that PML NBs display some features of liquid-liquid phase separation and that ATO induces a gel-like transition. PML B-box-2 structure reveals an alpha helix driving B2 trimerization and positioning a cysteine trio to form an ideal arsenic-binding pocket. Altering either of the latter impedes ATO-driven NB assembly, PML sumoylation, and PML-RARA degradation, mechanistically explaining clinical ATO resistance. This B2 trimer and the C213 trio create an oxidation-sensitive rheostat that controls PML NB assembly dynamics and downstream signaling in both basal state and during stress response. These findings identify the structural basis for arsenic targeting of PML that could pave the way to novel cancer drugs. SIGNIFICANCE: Arsenic curative effects in APL rely on PML targeting. We report a PML B-box-2 structure that drives trimer assembly, positioning a cysteine trio to form an arsenic-binding pocket, which is disrupted in resistant patients. Identification of this ROS-sensitive triad controlling PML dynamics and functions could yield novel drugs. See related commentary by Salomoni, p. 2505. This article is featured in Selected Articles from This Issue, p. 2489.


Subject(s)
Arsenic , Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Arsenic/pharmacology , Promyelocytic Leukemia Nuclear Bodies , Cysteine , Arsenicals/pharmacology , Oxides/pharmacology , Arsenic Trioxide/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Oncogene Proteins , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism
3.
Nat Commun ; 13(1): 5726, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36175410

ABSTRACT

Membrane-less organelles are condensates formed by phase separation whose functions often remain enigmatic. Upon oxidative stress, PML scaffolds Nuclear Bodies (NBs) to regulate senescence or metabolic adaptation. PML NBs recruit many partner proteins, but the actual biochemical mechanism underlying their pleiotropic functions remains elusive. Similarly, PML role in embryonic stem cell (ESC) and retro-element biology is unsettled. Here we demonstrate that PML is essential for oxidative stress-driven partner SUMO2/3 conjugation in mouse ESCs (mESCs) or leukemia, a process often followed by their poly-ubiquitination and degradation. Functionally, PML is required for stress responses in mESCs. Differential proteomics unravel the KAP1 complex as a PML NB-dependent SUMO2-target in arsenic-treated APL mice or mESCs. PML-driven KAP1 sumoylation enables activation of this key epigenetic repressor implicated in retro-element silencing. Accordingly, Pml-/- mESCs re-express transposable elements and display 2-Cell-Like features, the latter enforced by PML-controlled SUMO2-conjugation of DPPA2. Thus, PML orchestrates mESC state by coordinating SUMO2-conjugation of different transcriptional regulators, raising new hypotheses about PML roles in cancer.


Subject(s)
Arsenic , Sumoylation , Animals , DNA Transposable Elements , Embryonic Stem Cells , Mice , Nuclear Bodies , Transcription Factors
4.
J Exp Med ; 214(11): 3197-3206, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-28931625

ABSTRACT

Promyelocytic leukemia (PML) nuclear bodies (NBs) recruit partner proteins, including p53 and its regulators, thereby controlling their abundance or function. Investigating arsenic sensitivity of acute promyelocytic leukemia, we proposed that PML oxidation promotes NB biogenesis. However, physiological links between PML and oxidative stress response in vivo remain unexplored. Here, we identify PML as a reactive oxygen species (ROS) sensor. Pml-/- cells accumulate ROS, whereas PML expression decreases ROS levels. Unexpectedly, Pml-/- embryos survive acute glutathione depletion. Moreover, Pml-/- animals are resistant to acetaminophen hepatotoxicity or fasting-induced steatosis. Molecularly, Pml-/- animals fail to properly activate oxidative stress-responsive p53 targets, whereas the NRF2 response is amplified and accelerated. Finally, in an oxidative stress-prone background, Pml-/- animals display a longevity phenotype, likely reflecting decreased basal p53 activation. Thus, similar to p53, PML exerts basal antioxidant properties but also drives oxidative stress-induced changes in cell survival/proliferation or metabolism in vivo. Through NB biogenesis, PML therefore couples ROS sensing to p53 responses, shedding a new light on the role of PML in senescence or stem cell biology.


Subject(s)
Oxidative Stress , Promyelocytic Leukemia Protein/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Blotting, Western , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Intranuclear Inclusion Bodies/metabolism , Male , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Knockout , Microscopy, Confocal , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Promyelocytic Leukemia Protein/genetics , Tumor Suppressor Protein p53/genetics
5.
mBio ; 8(1)2017 01 10.
Article in English | MEDLINE | ID: mdl-28074026

ABSTRACT

The promyelocytic leukemia protein (PML) is the main organizer of stress-responsive subnuclear structures called PML nuclear bodies. These structures recruit multiple interactors and modulate their abundance or their posttranslational modifications, notably by the SUMO ubiquitin-like modifiers. The involvement of PML in antiviral responses is well established. In contrast, the role of PML in bacterial infection remains poorly characterized. Here, we show that PML restricts infection by the pathogenic bacterium Listeria monocytogenes but not by Salmonella enterica serovar Typhimurium. During infection, PML undergoes oxidation-mediated multimerization, associates with the nuclear matrix, and becomes de-SUMOylated due to the pore-forming activity of the Listeria toxin listeriolysin O (LLO). These events trigger an antibacterial response that is not observed during in vitro infection by an LLO-defective Listeria mutant, but which can be phenocopied by specific induction of PML de-SUMOylation. Using transcriptomic and proteomic microarrays, we also characterized a network of immunity genes and cytokines, which are regulated by PML in response to Listeria infection but independently from the listeriolysin O toxin. Our study thus highlights two mechanistically distinct complementary roles of PML in host responses against bacterial infection. IMPORTANCE: The promyelocytic leukemia protein (PML) is a eukaryotic protein that can polymerize in discrete nuclear assemblies known as PML nuclear bodies (NBs) and plays essential roles in many different cellular processes. Key to its function, PML can be posttranslationally modified by SUMO, a ubiquitin-like modifier. Identification of the role of PML in antiviral defenses has been deeply documented. In contrast, the role of PML in antibacterial defenses remains elusive. Here, we identify two mechanistically distinct complementary roles of PML in antibacterial responses against pathogens such as Listeria: (i) we show that PML regulates the expression of immunity genes in response to bacterial infection, and (ii) we unveil the fact that modification of PML SUMOylation by bacterial pore-forming toxins is sensed as a danger signal, leading to a restriction of bacterial intracellular multiplication. Taken together, our data reinforce the concept that intranuclear bodies can dynamically regulate important processes, such as defense against invaders.


Subject(s)
Host-Pathogen Interactions , Listeria monocytogenes/growth & development , Listeria monocytogenes/immunology , Promyelocytic Leukemia Protein/metabolism , Protein Processing, Post-Translational , Animals , Bacterial Toxins/metabolism , Cells, Cultured , Gene Expression Profiling , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Humans , Mice , Microarray Analysis , Protein Multimerization , Proteome/analysis , Salmonella typhimurium/growth & development , Salmonella typhimurium/immunology , Sumoylation
6.
Sci Signal ; 9(440): tc1, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27507651

ABSTRACT

Fasci et al proposed that a SENP1-mediated switch from SUMO2 to SUMO1 conjugation on Lys(65) in promyelocytic leukemia protein (PML) is required for arsenic-induced PML degradation, the basis for the antileukemic activity of arsenic. We found that PML or PML/RARA (retinoic acid receptor α) mutants that cannot be SUMO-conjugated on this specific site nevertheless underwent immediate arsenic-triggered SUMO modification. Moreover, these mutants were efficiently degraded in cells and even in vivo, demonstrating that SUMOylation of Lys(65) was dispensable for arsenic response. The existence and putative role of a SUMO switch on PML should thus be reassessed.


Subject(s)
Arsenic , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Tumor Suppressor Proteins/genetics , Ubiquitination
7.
Nat Commun ; 5: 4187, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24942926

ABSTRACT

Small ubiquitin-related modifier (SUMO) protein conjugation onto target proteins regulates multiple cellular functions, including defence against pathogens, stemness and senescence. SUMO1 peptides are limiting in quantity and are thus mainly conjugated to high-affinity targets. Conjugation of SUMO2/3 paralogues is primarily stress inducible and may initiate target degradation. Here we demonstrate that the expression of SUMO1/2/3 is dramatically enhanced by interferons through an miRNA-based mechanism involving the Lin28/let-7 axis, a master regulator of stemness. Normal haematopoietic progenitors indeed display much higher SUMO contents than their differentiated progeny. Critically, SUMOs contribute to the antiviral effects of interferons against HSV1 or HIV. Promyelocytic leukemia (PML) nuclear bodies are interferon-induced domains, which facilitate sumoylation of a subset of targets. Our findings thus identify an integrated interferon-responsive PML/SUMO pathway that impedes viral replication by enhancing SUMO conjugation and possibly also modifying the repertoire of targets. Interferon-enhanced post-translational modifications may be essential for senescence or stem cell self-renewal, and initiate SUMO-dependent proteolysis.


Subject(s)
HIV-1/physiology , Herpesvirus 1, Human/physiology , MicroRNAs/immunology , RNA-Binding Proteins/immunology , SUMO-1 Protein/immunology , Small Ubiquitin-Related Modifier Proteins/immunology , Ubiquitins/immunology , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Humans , Interferons/immunology , MicroRNAs/genetics , RNA-Binding Proteins/genetics , SUMO-1 Protein/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Ubiquitins/genetics , Virus Replication
8.
J Cell Biol ; 204(6): 931-45, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24637324

ABSTRACT

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.


Subject(s)
Nuclear Proteins/metabolism , Oxidative Stress , Sumoylation , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , CHO Cells , COS Cells , Cell Nucleus/metabolism , Cellular Senescence , Chlorocebus aethiops , Cricetinae , Cricetulus , HeLa Cells , Humans , Mice , Promyelocytic Leukemia Protein , Protein Transport , Reactive Oxygen Species/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases
9.
Cancer Cell ; 18(1): 88-98, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20609355

ABSTRACT

As(2)O(3) cures acute promyelocytic leukemia (APL) by initiating PML/RARA oncoprotein degradation, through sumoylation of its PML moiety. However, how As(2)O(3) initiates PML sumoylation has remained largely unexplained. As(2)O(3) binds vicinal cysteines and increases reactive oxygen species (ROS) production. We demonstrate that upon As(2)O(3) exposure, PML undergoes ROS-initiated intermolecular disulfide formation and binds arsenic directly. Disulfide-linked PML or PML/RARA multimers form nuclear matrix-associated nuclear bodies (NBs), become sumoylated and are degraded. Hematopoietic progenitors transformed by an As(2)O(3)-binding PML/RARA mutant exhibit defective As(2)O(3) response. Conversely, nonarsenical oxidants elicit PML/RARA multimerization, NB-association, degradation, and leukemia response in vivo, but do not affect PLZF/RARA-driven APLs. Thus, PML oxidation regulates NB-biogenesis, while oxidation-enforced PML/RARA multimerization and direct arsenic-binding cooperate to enforce APL's exquisite As(2)O(3) sensitivity.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/metabolism , Oxides/pharmacology , Animals , Arsenic Trioxide , Blotting, Western , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Disulfides/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Intranuclear Inclusion Bodies/metabolism , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Knockout , Mutation/genetics , Nuclear Proteins/physiology , Oncogene Proteins, Fusion/genetics , Promyelocytic Leukemia Protein , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Processing, Post-Translational , Reactive Oxygen Species/metabolism , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology
10.
Nat Med ; 14(12): 1333-42, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19029980

ABSTRACT

Retinoic acid and arsenic trioxide target the protein stability and transcriptional repression activity of the fusion oncoprotein PML-RARA, resulting in regression of acute promyelocytic leukemia (APL). Phenotypically, retinoic acid induces differentiation of APL cells. Here we show that retinoic acid also triggers growth arrest of leukemia-initiating cells (LICs) ex vivo and their clearance in PML-RARA mouse APL in vivo. Retinoic acid treatment of mouse APLs expressing the fusion protein PLZF-RARA triggers full differentiation, but not LIC loss or disease remission, establishing that differentiation and LIC loss can be uncoupled. Although retinoic acid and arsenic synergize to clear LICs through cooperative PML-RARA degradation, this combination does not enhance differentiation. A cyclic AMP (cAMP)-dependent phosphorylation site in PML-RARA is crucial for retinoic acid-induced PML-RARA degradation and LIC clearance. Moreover, activation of cAMP signaling enhances LIC loss by retinoic acid, identifying cAMP as another potential APL therapy. Thus, whereas transcriptional activation of PML-RARA is likely to control differentiation, its catabolism triggers LIC eradication and long-term remission of mouse APL. Therapy-triggered degradation of oncoproteins could be a general strategy to eradicate cancer stem cells.


Subject(s)
Leukemia, Promyelocytic, Acute/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Nude , Oncogene Proteins, Fusion/genetics , Phosphorylation , Serine/genetics , Serine/metabolism , Signal Transduction , Tretinoin/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL