Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Biochem J ; 474(9): 1467-1479, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28264989

ABSTRACT

ADAM9 (A Disintegrin And Metalloprotease 9) is a membrane-anchored metalloproteinase that has been implicated in pathological retinal neovascularization and in tumor progression. ADAM9 has constitutive catalytic activity in both biochemical and cell-based assays and can cleave several membrane proteins, including epidermal growth factor and Ephrin receptor B4; yet little is currently known about the catalytic properties of ADAM9 and its post-translational regulation and inhibitor profile in cell-based assays. To address this question, we monitored processing of the membrane-anchored Ephrin receptor B4 (EphB4) by co-expressing ADAM9, with the catalytically inactive ADAM9 E > A mutant serving as a negative control. We found that ADAM9-dependent shedding of EphB4 was not stimulated by three commonly employed activators of ADAM-dependent ectodomain shedding: phorbol esters, pervanadate or calcium ionophores. With respect to the inhibitor profile, we found that ADAM9 was inhibited by the hydroxamate-based metalloprotease inhibitors marimastat, TAPI-2, BB94, GM6001 and GW280264X, and by 10 nM of the tissue inhibitor of metalloproteinases (TIMP)-3, but not by up to 20 nM of TIMP-1 or -2. Additionally, we screened a non-hydroxamate small-molecule library for novel ADAM9 inhibitors and identified four compounds that selectively inhibited ADAM9-dependent proteolysis over ADAM10- or ADAM17-dependent processing. Taken together, the present study provides new information about the molecular fingerprint of ADAM9 in cell-based assays by showing that it is not stimulated by strong activators of ectodomain shedding and by defining a characteristic inhibitor profile. The identification of novel non-hydroxamate inhibitors of ADAM9 could provide the basis for designing more selective compounds that block the contribution of ADAM9 to pathological neovascularization and cancer.


Subject(s)
ADAM Proteins/antagonists & inhibitors , ADAM Proteins/metabolism , Cell Membrane/enzymology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Animals , COS Cells , Catalysis , Cell Membrane/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Mice
2.
Blood ; 125(3): 474-82, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25395419

ABSTRACT

The human T-cell lymphotropic virus type I (HTLV-1) Tax transactivator initiates transformation in adult T-cell leukemia/lymphoma (ATL), a highly aggressive chemotherapy-resistant malignancy. The arsenic/interferon combination, which triggers degradation of the Tax oncoprotein, selectively induces apoptosis of ATL cell lines and has significant clinical activity in Tax-driven murine ATL or human patients. However, the role of Tax loss in ATL response is disputed, and the molecular mechanisms driving degradation remain elusive. Here we demonstrate that ATL-derived or HTLV-1-transformed cells are dependent on continuous Tax expression, suggesting that Tax degradation underlies clinical responses to the arsenic/interferon combination. The latter enforces promyelocytic leukemia protein (PML) nuclear body (NB) formation and partner protein recruitment. In arsenic/interferon-treated HTLV-1 transformed or ATL cells, Tax is recruited onto NBs and undergoes PML-dependent hyper-sumoylation by small ubiquitin-like modifier (SUMO)2/3 but not SUMO1, ubiquitination by RNF4, and proteasome-dependent degradation. Thus, the arsenic/interferon combination clears ATL through degradation of its Tax driver, and this regimen could have broader therapeutic value by promoting degradation of other pathogenic sumoylated proteins.


Subject(s)
Arsenicals/pharmacology , Gene Products, tax/metabolism , Interferons/pharmacology , Leukemia-Lymphoma, Adult T-Cell/virology , Nuclear Proteins/metabolism , Proteolysis/drug effects , SUMO-1 Protein/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Antiviral Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Cell Transformation, Viral/drug effects , Drug Therapy, Combination , Flow Cytometry , Fluorescent Antibody Technique , HeLa Cells , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/pathogenicity , Humans , Immunoprecipitation , Leukemia-Lymphoma, Adult T-Cell/genetics , Promyelocytic Leukemia Protein , Sulfhydryl Reagents/pharmacology , Sumoylation/drug effects , Ubiquitination/drug effects
3.
Blood ; 125(22): 3447-54, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25800051

ABSTRACT

Nucleophosmin-1 (NPM1) is the most frequently mutated gene in acute myeloid leukemia (AML). Addition of retinoic acid (RA) to chemotherapy was proposed to improve survival of some of these patients. Here, we found that RA or arsenic trioxide synergistically induce proteasomal degradation of mutant NPM1 in AML cell lines or primary samples, leading to differentiation and apoptosis. NPM1 mutation not only delocalizes NPM1 from the nucleolus, but it also disorganizes promyelocytic leukemia (PML) nuclear bodies. Combined RA/arsenic treatment significantly reduced bone marrow blasts in 3 patients and restored the subnuclear localization of both NPM1 and PML. These findings could explain the proposed benefit of adding RA to chemotherapy in NPM1 mutant AMLs, and warrant a broader clinical evaluation of regimen comprising a RA/arsenic combination.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Oxides/pharmacology , Proteolysis/drug effects , Tretinoin/pharmacology , Aged , Aged, 80 and over , Apoptosis/genetics , Arsenic Trioxide , Cell Differentiation/drug effects , Cell Differentiation/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutant Proteins/drug effects , Mutant Proteins/metabolism , Mutation , Nucleophosmin , Tumor Cells, Cultured
4.
J Pathol ; 234(3): 289-91, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25138686

ABSTRACT

PML nuclear bodies (NBs) were first described by electron microscopy and rediscovered through their treatment-reversible disruption in a rare leukaemia. They recruit multiple partner proteins and now emerge as interferon- and oxidative stress-responsive sumoylation factories. NBs mediate interferon-induced viral restriction, enhance proteolysis, finely tune metabolism and enforce stress-induced senescence. Apart from being markers of cellular stress, PML NBs could be harnessed pharmacologically in a number of conditions, including cancer, viral infection or neurodegenerative diseases.


Subject(s)
Intranuclear Inclusion Bodies , Nuclear Proteins , Transcription Factors , Tumor Suppressor Proteins , Animals , Humans , Promyelocytic Leukemia Protein
5.
Article in English | MEDLINE | ID: mdl-36935613

ABSTRACT

SOD1 is the first identified causative gene for amyotrophic lateral sclerosis. Recently, a novel syndrome, presenting with severe childhood-onset spastic tetraplegia and axial hypotonia caused by the homozygous truncating variants in the SOD1 gene, is described. A 22-month-old boy was admitted with a loss of motor functions that began at the age of 9 months. Neurological was significant for axial hypotonia with spastic tetraplegia and hyperekplexia-like jerky movements. In WES, we found a novel homozygous variant (c.52_56del5ins154) in the SOD1 gene, resulting in a total loss of SOD1 mRNA expression in the real-time PCR analysis. Western blot analyses confirmed the lack of protein production. Erythrocyte superoxide dismutase enzymatic activity was nearly abolished. The heterozygous family members displayed reduced superoxide dismutase 1 protein expression and enzymatic activity (by about 40%), compared with the healthy control. Our study expanded the mutation spectrum of SOD1.


Subject(s)
Amyotrophic Lateral Sclerosis , Humans , Infant , Male , Amyotrophic Lateral Sclerosis/genetics , Muscle Hypotonia/genetics , Mutation , Superoxide Dismutase/genetics , Superoxide Dismutase-1/genetics
6.
Cells ; 11(5)2022 02 26.
Article in English | MEDLINE | ID: mdl-35269436

ABSTRACT

Sumoylation is an essential post-translational modification that has evolved to regulate intricate networks within emerging complexities of eukaryotic cells. Thousands of target substrates are modified by SUMO peptides, leading to changes in protein function, stability or localization, often by modulating interactions. At the cellular level, sumoylation functions as a key regulator of transcription, nuclear integrity, proliferation, senescence, lineage commitment and stemness. A growing number of prokaryotic and viral proteins are also emerging as prime sumoylation targets, highlighting the role of this modification during infection and in immune processes. Sumoylation also oversees epigenetic processes. Accordingly, at the physiological level, it acts as a crucial regulator of development. Yet, perhaps the most prominent function of sumoylation, from mammals to plants, is its role in orchestrating organismal responses to environmental stresses ranging from hypoxia to nutrient stress. Consequently, a growing list of pathological conditions, including cancer and neurodegeneration, have now been unambiguously associated with either aberrant sumoylation of specific proteins and/or dysregulated global cellular sumoylation. Therapeutic enforcement of sumoylation can also accomplish remarkable clinical responses in various diseases, notably acute promyelocytic leukemia (APL). In this review, we will discuss how this modification is emerging as a novel drug target, highlighting from the perspective of translational medicine, its potential and limitations.


Subject(s)
Small Ubiquitin-Related Modifier Proteins , Sumoylation , Animals , Mammals/metabolism , Plants/metabolism , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/metabolism , Viral Proteins/metabolism
7.
Life Sci Alliance ; 5(6)2022 06.
Article in English | MEDLINE | ID: mdl-35181598

ABSTRACT

During infection, the human immunodeficiency virus type 1 (HIV-1) manipulates host cell mechanisms to its advantage, thereby controlling its replication or latency, and evading immune responses. Sumoylation is an essential post-translational modification that controls vital cellular activities including proliferation, stemness, or anti-viral immunity. SUMO peptides oppose pathogen replication and mediate interferon-dependent anti-viral activities. In turn, several viruses and bacteria attack sumoylation to disarm host immune responses. Here, we show that HIV-1 impairs cellular sumoylation and targets the host SUMO E1-activating enzyme. HIV-1 expression in cultured HEK293 cells or in CD4+ Jurkat T lymphocytes diminishes sumoylation by both SUMO paralogs, SUMO1 and SUMO2/3. HIV-1 causes a sharp and specific decline in UBA2 protein levels, a subunit of the heterodimeric SUMO E1 enzyme, which likely serves to reduce the efficiency of global protein sumoylation. Furthermore, HIV-1-infected individuals display a significant reduction in total leukocyte sumoylation that is uncoupled from HIV-induced cytopenia. Because sumoylation is vital for immune function, T-cell expansion and activity, loss of sumoylation during HIV disease may contribute to immune system deterioration in patients.


Subject(s)
HIV Infections , HIV-1 , HEK293 Cells , Humans , Protein Processing, Post-Translational , Sumoylation , Ubiquitin-Activating Enzymes
8.
Life Sci Alliance ; 5(4)2022 04.
Article in English | MEDLINE | ID: mdl-35022246

ABSTRACT

CRISPR/Cas9 is a popular genome editing technology. Although widely used, little is known about how this prokaryotic system behaves in humans. An unwanted consequence of eukaryotic Cas9 expression is off-target DNA binding leading to mutagenesis. Safer clinical implementation of CRISPR/Cas9 necessitates a finer understanding of the regulatory mechanisms governing Cas9 behavior in humans. Here, we report our discovery of Cas9 sumoylation and ubiquitylation, the first post-translational modifications to be described on this enzyme. We found that the major SUMO2/3 conjugation site on Cas9 is K848, a key positively charged residue in the HNH nuclease domain that is known to interact with target DNA and contribute to off-target DNA binding. Our results suggest that Cas9 ubiquitylation leads to decreased stability via proteasomal degradation. Preventing Cas9 sumoylation through conversion of K848 into arginine or pharmacologic inhibition of cellular sumoylation enhances the enzyme's turnover and diminishes guide RNA-directed DNA binding efficacy, suggesting that sumoylation at this site regulates Cas9 stability and DNA binding. More research is needed to fully understand the implications of these modifications for Cas9 specificity.


Subject(s)
CRISPR-Associated Protein 9 , DNA/metabolism , Lysine , Sumoylation/genetics , CRISPR-Associated Protein 9/chemistry , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , HEK293 Cells , Humans , Lysine/chemistry , Lysine/genetics , Protein Stability , Small Ubiquitin-Related Modifier Proteins/metabolism
9.
FEBS J ; 287(15): 3110-3140, 2020 08.
Article in English | MEDLINE | ID: mdl-32255256

ABSTRACT

Sumoylation is an essential post-translational modification intimately involved in a diverse range of eukaryotic cellular mechanisms. Small ubiquitin-like modifier (SUMO) protein isoforms can be reversibly linked to lysine residues that reside within specific motifs on thousands of target substrates, leading to modulations in stability, solubility, localization, and interactor profile. Since its initial discovery almost 25 years ago, SUMO has been described as a key regulator of genomic stability, cell proliferation, and infection among other processes. In this review, we trace the exciting developments in the history of this critical modifier, highlighting SUMO's roles in pathogenesis as well as its potential for the development of targeted therapies for numerous diseases.


Subject(s)
Anniversaries and Special Events , Infections/pathology , Neoplasms/pathology , Neurodegenerative Diseases/pathology , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Humans , Infections/metabolism , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism
10.
J Cell Biol ; 164(5): 769-79, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14993236

ABSTRACT

All ligands of the epidermal growth factor receptor (EGFR), which has important roles in development and disease, are released from the membrane by proteases. In several instances, ectodomain release is critical for activation of EGFR ligands, highlighting the importance of identifying EGFR ligand sheddases. Here, we uncovered the sheddases for six EGFR ligands using mouse embryonic cells lacking candidate-releasing enzymes (a disintegrin and metalloprotease [ADAM] 9, 10, 12, 15, 17, and 19). ADAM10 emerged as the main sheddase of EGF and betacellulin, and ADAM17 as the major convertase of epiregulin, transforming growth factor alpha, amphiregulin, and heparin-binding EGF-like growth factor in these cells. Analysis of adam9/12/15/17-/- knockout mice corroborated the essential role of adam17-/- in activating the EGFR in vivo. This comprehensive evaluation of EGFR ligand shedding in a defined experimental system demonstrates that ADAMs have critical roles in releasing all EGFR ligands tested here. Identification of EGFR ligand sheddases is a crucial step toward understanding the mechanism underlying ectodomain release, and has implications for designing novel inhibitors of EGFR-dependent tumors.


Subject(s)
Endopeptidases/metabolism , ErbB Receptors/metabolism , Metalloendopeptidases/metabolism , Phenylalanine/analogs & derivatives , ADAM Proteins , ADAM12 Protein , ADAM17 Protein , Amphiregulin , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Betacellulin , Cells, Cultured , Disintegrins/genetics , Disintegrins/metabolism , EGF Family of Proteins , Embryo, Mammalian/anatomy & histology , Endopeptidases/genetics , Epidermal Growth Factor/metabolism , Epiregulin , Fibroblasts/cytology , Fibroblasts/metabolism , Genotype , Glycoproteins/metabolism , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Phenylalanine/metabolism , Protease Inhibitors/metabolism , Protein Structure, Tertiary , Tetradecanoylphorbol Acetate/metabolism , Thiophenes/metabolism , Transforming Growth Factor alpha/metabolism
11.
Life Sci Alliance ; 2(4)2019 08.
Article in English | MEDLINE | ID: mdl-31431473

ABSTRACT

Aberrant androgen signaling drives prostate cancer and is targeted by drugs that diminish androgen production or impede androgen-androgen receptor (AR) interaction. Clinical resistance arises from AR overexpression or ligand-independent constitutive activation, suggesting that complete AR elimination could be a novel therapeutic strategy in prostate cancers. IRC117539 is a new molecule that targets AR for proteasomal degradation. Exposure to IRC117539 promotes AR sumoylation and ubiquitination, reminiscent of therapy-induced PML/RARA degradation in acute promyelocytic leukemia. Critically, ex vivo, IRC117539-mediated AR degradation induces prostate cancer cell viability loss by inhibiting AR signaling, even in androgen-insensitive cells. This approach may be beneficial for castration-resistant prostate cancer, which remains a clinical issue. In xenograft models, IRC117539 is as potent as enzalutamide in impeding growth, albeit less efficient than expected from ex vivo studies. Unexpectedly, IRC117539 also behaves as a weak proteasome inhibitor, likely explaining its suboptimal efficacy in vivo. Our studies highlight the feasibility of AR targeting for degradation and off-target effects' importance in modulating drug activity in vivo.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Androgen Antagonists/metabolism , Androgen Receptor Antagonists/metabolism , Androgens/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostate/metabolism , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
FEBS Lett ; 581(1): 41-4, 2007 Jan 09.
Article in English | MEDLINE | ID: mdl-17169360

ABSTRACT

All ligands of the epidermal growth factor receptor (EGFR), which has important roles in development and disease, are made as transmembrane precursors. Proteolytic processing by ADAMs (a disintegrin and metalloprotease) regulates the bioavailability of several EGFR-ligands, yet little is known about the enzyme responsible for processing the recently identified EGFR ligand, epigen. Here we show that ectodomain shedding of epigen requires ADAM17, which can be stimulated by phorbol esters, phosphatase inhibitors and calcium influx. These results suggest that ADAM17 might be a good target to block the release of bioactive epigen, a highly mitogenic ligand of the EGFR which has been implicated in wound healing and cancer.


Subject(s)
ADAM Proteins/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Fibroblasts/enzymology , Growth Substances/metabolism , Membrane Proteins/metabolism , Protein Precursors/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAM17 Protein , Animals , Cells, Cultured , Epidermal Growth Factor/genetics , Epigen , ErbB Receptors/agonists , ErbB Receptors/genetics , Fibroblasts/cytology , Growth Substances/genetics , Humans , Ligands , Membrane Proteins/genetics , Mice , Neoplasms/genetics , Neoplasms/metabolism , Protease Inhibitors/pharmacology , Protein Precursors/genetics , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Protein Structure, Tertiary/genetics , Wound Healing/drug effects , Wound Healing/genetics
13.
Mol Cell Biol ; 24(1): 96-104, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14673146

ABSTRACT

Congenital heart disease is the most common form of human birth defects, yet much remains to be learned about its underlying causes. Here we report that mice lacking functional ADAM19 (mnemonic for a disintegrin and metalloprotease 19) exhibit severe defects in cardiac morphogenesis, including a ventricular septal defect (VSD), abnormal formation of the aortic and pulmonic valves, leading to valvular stenosis, and abnormalities of the cardiac vasculature. During mouse development, ADAM19 is highly expressed in the conotruncus and the endocardial cushion, structures that give rise to the affected heart valves and the membranous ventricular septum. ADAM19 is also highly expressed in osteoblast-like cells in the bone, yet it does not appear to be essential for bone growth and skeletal development. Most adam19(-/-) animals die perinatally, likely as a result of their cardiac defects. These findings raise the possibility that mutations in ADAM19 may contribute to human congenital heart valve and septal defects.


Subject(s)
Cardiovascular System/embryology , Disintegrins/metabolism , Membrane Proteins/metabolism , Metalloproteases/metabolism , ADAM Proteins , Animals , Blotting, Western , Bone Development/physiology , Brain/metabolism , Lung/growth & development , Mice
14.
Methods Mol Biol ; 327: 99-113, 2006.
Article in English | MEDLINE | ID: mdl-16780215

ABSTRACT

All ligands of the epidermal growth factor receptor (EGFR) are made as membrane anchored precursors that can be proteolytically processed and released from the plasma membrane. This process, which is referred to as protein ectodomain shedding, is emerging as a key regulator of the function of EGFR ligands. In light of the important roles of EGFR signaling in development and disease, it will be important to understand more about the regulation of proteolytic processing of EGFR ligands. This chapter describes a sensitive and semiquantitative method to measure ectodomain shedding of EGFR ligands that was designed to facilitate studies of this process in cells.


Subject(s)
ErbB Receptors/chemistry , ErbB Receptors/metabolism , Alkaline Phosphatase/metabolism , Animals , ErbB Receptors/analysis , Fibroblasts/metabolism , Ligands , Mice , Sensitivity and Specificity
15.
Methods Mol Biol ; 1475: 139-50, 2016.
Article in English | MEDLINE | ID: mdl-27631803

ABSTRACT

Sumoylation is a posttranslational process essential for life and concerns a growing number of crucial proteins. Understanding the influence of this phenomenon on individual proteins or on cellular pathways in which they function has become an intense area of research. A critical step in studying protein sumoylation is to detect sumoylated forms of a particular protein. This has proven to be a challenging task for a number of reasons, especially in the case of endogenous proteins and in vivo studies or when studying rare cells such as stem cells. Proximity ligation assays that allow detection of closely interacting protein partners can be adapted for initial detection of endogenous sumoylation or ubiquitination in a rapid, ultrasensitive, and cheap manner. In addition, modified forms of a given protein can be detected in situ in various cellular compartments. Finally, the flexibility of this technique may allow rapid screening of drugs and stress signals that may modulate protein sumoylation.


Subject(s)
Antigens, Nuclear/metabolism , Autoantigens/metabolism , Biological Assay , Protein Processing, Post-Translational , SUMO-1 Protein/metabolism , Antibodies/chemistry , Antigens, Nuclear/genetics , Arsenic/pharmacology , Autoantigens/genetics , Fluorescent Antibody Technique , HeLa Cells , Humans , Interferon-alpha/pharmacology , Protein Binding , Protein Interaction Domains and Motifs , SUMO-1 Protein/genetics , Sumoylation
16.
Nucleus ; 5(6): 499-507, 2014.
Article in English | MEDLINE | ID: mdl-25482067

ABSTRACT

PML Nuclear Bodies (NBs) have fascinated cell biologists due to their exquisitely dynamic nature and their involvement in human diseases, notably acute promyelocytic leukemia. NBs, as well as their master organizer--the PML protein--exhibit multiple connections with stress responses. Initially viewed as a tumor suppressor, PML recently re-emerged as a multifaceted protein, capable of controlling numerous aspects of cellular homeostasis. NBs recruit many functionally diverse proteins and function as stress-regulated sumoylation factories. SUMO-initiated partner retention can subsequently facilitate a variety of other post-translational modifications, as well as partner degradation. With this newly elucidated central role of stress-enhanced sumoylation, it should now be possible to build a working model for the different NB-regulated cellular activities. Moreover, pharmacological manipulation of NB formation by interferons or oxidants holds the promise of clearing many undesirable proteins for clinical management of malignant, viral or neurodegenerative diseases.


Subject(s)
Intranuclear Inclusion Bodies/genetics , Nuclear Proteins/genetics , Oxidative Stress/genetics , SUMO-1 Protein/genetics , Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Nucleus/metabolism , Cellular Senescence/genetics , Humans , Protein Processing, Post-Translational , Proteolysis , Repressor Proteins/genetics , Sumoylation/genetics
17.
PLoS One ; 9(7): e102957, 2014.
Article in English | MEDLINE | ID: mdl-25036361

ABSTRACT

Gene silencing by small RNAs has emerged as a powerful post-transcriptional regulator of gene expression, however processes underlying regulation of the small RNA pathway in vivo are still largely elusive. Here, we identified sumoylation as a novel post-translational modification acting on Ago2, the main effector of small RNA-mediated gene silencing. We demonstrate that Ago2 can be modified by SUMO1 and SUMO2/3 and identified Lys402 as the major Ago2 sumoylation site in vivo. Ago2 physically interacts with the SUMO E2 conjugating enzyme Ubc9 and the E3 ligase RanBP2 facilitates Ago2 sumoylation in vitro. Mutation of Lys402 enhances the stability of Ago2 protein and impairment of cellular sumoylation by siRNA- or shRNA-mediated extinction of Ubc9 or in Ubc9 knockout mouse tissues results in increased steady-state levels and enhanced stability of Ago2. Similarly, knockdown of RanBP2 or of the SAE2 E1 enzyme enhances Ago2 protein levels. Lys402 is located in the L2g1 loop linking the PAZ and PIWI domains of Ago2, in the immediate vicinity of Tyr393 which can be phosphorylated, implying that the L2g1 linker represents an easily accessible hot spot for post-translational modifications. Altogether, our results show that sumoylation of Ago2 at Lys402 negatively regulates its stability, thereby establishing a first link between SUMO and the small RNA machinery.


Subject(s)
Argonaute Proteins/metabolism , Lysine/metabolism , Sumoylation/genetics , Animals , Argonaute Proteins/genetics , Cell Line, Tumor , Gene Silencing/physiology , HeLa Cells , Humans , Lysine/genetics , Mice , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Phosphorylation/genetics , Protein Processing, Post-Translational/genetics , RNA, Small Interfering/genetics , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
18.
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
19.
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
SELECTION OF CITATIONS
SEARCH DETAIL