Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
PLoS Genet ; 18(1): e1010015, 2022 01.
Article in English | MEDLINE | ID: mdl-35025870

ABSTRACT

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset disorder characterized by progressive weakness and degeneration of specific muscles. OPMD is due to extension of a polyalanine tract in poly(A) binding protein nuclear 1 (PABPN1). Aggregation of the mutant protein in muscle nuclei is a hallmark of the disease. Previous transcriptomic analyses revealed the consistent deregulation of the ubiquitin-proteasome system (UPS) in OPMD animal models and patients, suggesting a role of this deregulation in OPMD pathogenesis. Subsequent studies proposed that UPS contribution to OPMD involved PABPN1 aggregation. Here, we use a Drosophila model of OPMD to address the functional importance of UPS deregulation in OPMD. Through genome-wide and targeted genetic screens we identify a large number of UPS components that are involved in OPMD. Half dosage of UPS genes reduces OPMD muscle defects suggesting a pathological increase of UPS activity in the disease. Quantification of proteasome activity confirms stronger activity in OPMD muscles, associated with degradation of myofibrillar proteins. Importantly, improvement of muscle structure and function in the presence of UPS mutants does not correlate with the levels of PABPN1 aggregation, but is linked to decreased degradation of muscle proteins. Oral treatment with the proteasome inhibitor MG132 is beneficial to the OPMD Drosophila model, improving muscle function although PABPN1 aggregation is enhanced. This functional study reveals the importance of increased UPS activity that underlies muscle atrophy in OPMD. It also provides a proof-of-concept that inhibitors of proteasome activity might be an attractive pharmacological approach for OPMD.


Subject(s)
Muscular Atrophy/pathology , Muscular Dystrophy, Oculopharyngeal/pathology , Poly(A)-Binding Protein I/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Disease Models, Animal , Drosophila melanogaster , Gene Expression Regulation , Genetic Testing , Humans , Leupeptins/pharmacology , Leupeptins/therapeutic use , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscular Dystrophy, Oculopharyngeal/drug therapy , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/metabolism , Mutation , Poly(A)-Binding Protein I/chemistry , Proof of Concept Study , Protein Aggregates/drug effects
2.
Traffic ; 23(5): 287-304, 2022 05.
Article in English | MEDLINE | ID: mdl-35466519

ABSTRACT

Proteasomes are major non-lysosomal proteolytic complexes localized in the cytoplasm and in the nucleus of eukaryotic cells. Strikingly, high levels of extracellular proteasome have also been evidenced in the plasma (p-proteasome) of patients with specific diseases. Here, we examined the process by which proteasomes are secreted, as well as their structural and functional features once in the extracellular space. We demonstrate that assembled 20S core particles are secreted by cells within microvesicles budding from the plasma membrane. Part of the extracellular proteasome pool is also free of membranes in the supernatant of cultured cells, and likely originates from microvesicles leakage. We further demonstrate that this free proteasome released by cells (cc-proteasome for cell culture proteasome) possesses latent proteolytic activity and can degrade various extracellular proteins. Both standard (no immune-subunits) and intermediate (containing some immune-subunits) forms of 20S are observed. Moreover, we show that galectin-3, which displays a highly disordered N-terminal region, is efficiently cleaved by purified cc-proteasome, without SDS activation, likely after its binding to PSMA3 (α7) subunit through its intrinsically disordered region. As a consequence, galectin-3 is unable to induce red blood cells agglutination when preincubated with cc-proteasome. These results highlight potential novel physio- and pathologic functions for the extracellular proteasome.


Subject(s)
Galectin 3 , Proteasome Endopeptidase Complex , Agglutination , Cytoplasm/metabolism , Galectin 3/metabolism , Humans , Proteasome Endopeptidase Complex/metabolism , Proteolysis
3.
Trends Biochem Sci ; 44(5): 383-387, 2019 05.
Article in English | MEDLINE | ID: mdl-30777377

ABSTRACT

Protein homeostasis (proteostasis) is at the core of cellular functions. The European network PROTEOSTASIS was created to steer research and foster collaborations in the interconnected fields of posttranslational modifications by ubiquitin family members and protein turnover by proteasome, autophagy, and lysosomal systems in health and diseases, across the kingdoms of life.


Subject(s)
Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteostasis , Ubiquitin/metabolism , Autophagy , Europe , Homeostasis , Humans , Protein Processing, Post-Translational
4.
J Cell Sci ; 134(3)2021 02 01.
Article in English | MEDLINE | ID: mdl-33526472

ABSTRACT

PA28γ (also known as PSME3), a nuclear activator of the 20S proteasome, is involved in the degradation of several proteins regulating cell growth and proliferation and in the dynamics of various nuclear bodies, but its precise cellular functions remain unclear. Here, using a quantitative FLIM-FRET based microscopy assay monitoring close proximity between nucleosomes in living human cells, we show that PA28γ controls chromatin compaction. We find that its depletion induces a decompaction of pericentromeric heterochromatin, which is similar to what is observed upon the knockdown of HP1ß (also known as CBX1), a key factor of the heterochromatin structure. We show that PA28γ is present at HP1ß-containing repetitive DNA sequences abundant in heterochromatin and, importantly, that HP1ß on its own is unable to drive chromatin compaction without the presence of PA28γ. At the molecular level, we show that this novel function of PA28γ is independent of its stable interaction with the 20S proteasome, and most likely depends on its ability to maintain appropriate levels of H3K9me3 and H4K20me3, histone modifications that are involved in heterochromatin formation. Overall, our results implicate PA28γ as a key factor involved in the regulation of the higher order structure of chromatin.


Subject(s)
Chromatin , Proteasome Endopeptidase Complex , Autoantigens , Chromatin/genetics , Chromobox Protein Homolog 5 , Heterochromatin/genetics , Humans , Proteasome Endopeptidase Complex/genetics
5.
Bioorg Chem ; 119: 105539, 2022 02.
Article in English | MEDLINE | ID: mdl-34894575

ABSTRACT

It is urgent to develop less toxic and more efficient treatments for leishmaniases and trypanosomiases. We explore the possibility to target the parasite mitochondrial HslVU protease, which is essential for growth and has no analogue in the human host. For this, we develop compounds potentially inhibiting the complex assembly by mimicking the C-terminal (C-ter) segment of the ATPase HslU. We previously showed that a dodecapeptide derived from Leishmania major HslU C-ter segment (LmC12-U2, Cpd 1) was able to bind to and activate the digestion of a fluorogenic substrate by LmHslV. Here, we present the study of its structure-activity relationships. By replacing each essential residue with related non-proteinogenic residues, we obtained more potent analogues. In particular, a cyclohexylglycine residue at position 11 (cpd 24) allowed a more than three-fold gain in potency while reducing the size of compound 24 from twelve to six residues (cpd 50) without significant loss of potency, opening the way toward short HslU C-ter peptidomimetics as potential inhibitors of HslV proteolytic function. Finally, conjugates constituted of LmC6-U2 analogues and a mitochondrial penetrating peptide were found to penetrate into the promastigote form of L. infantum and to inhibit the parasite growth without showing toxicity toward human THP-1 cells at the same concentration (i.e. 30 µM).


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Adenosine Triphosphatases/metabolism , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Leishmania major/enzymology , Molecular Structure , Structure-Activity Relationship , THP-1 Cells
6.
Cell Mol Life Sci ; 79(1): 45, 2021 Dec 16.
Article in English | MEDLINE | ID: mdl-34913092

ABSTRACT

PA28γ is a nuclear activator of the 20S proteasome that, unlike the 19S regulatory particle, stimulates hydrolysis of several substrates in an ATP- and ubiquitin-independent manner and whose exact biological functions and molecular mechanism of action still remain elusive. In an effort to shed light on these important issues, we investigated the stimulatory effect of PA28γ on the hydrolysis of different fluorogenic peptides and folded or denatured full-length proteins by the 20S proteasome. Importantly, PA28γ was found to dramatically enhance breakdown rates by 20S proteasomes of several naturally or artificially unstructured proteins, but not of their native, folded counterparts. Furthermore, these data were corroborated by experiments in cell lines with a nucleus-tagged myelin basic protein. Finally, mass spectrometry analysis of the products generated during proteasomal degradation of two proteins demonstrated that PA28γ does not increase, but rather decreases, the variability of peptides that are potentially suitable for MHC class I antigen presentation. These unexpected findings indicate that global stimulation of the degradation of unfolded proteins may represent a more general feature of PA28γ and suggests that this proteasomal activator might play a broader role in the pathway of protein degradation than previously believed.


Subject(s)
Autoantigens/metabolism , Intrinsically Disordered Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , HeLa Cells , Humans , Proteolysis , Unfolded Protein Response
7.
Traffic ; 20(7): 516-536, 2019 07.
Article in English | MEDLINE | ID: mdl-31042005

ABSTRACT

The aim of our study was to investigate the impact of macroautophagy on exosome secretion. Exosomes are small membrane vesicles released in the extracellular space upon fusion of multivesicular endosomes with the plasma membrane. They were initially discovered as a way to remodel the reticulocyte plasma membrane before entering the blood circulation (Current Opinion in Hematology 2010, 17:177-183) and are now essentially studied as mediators of intercellular communication. Using iTRAQ proteomics, we compared the protein composition of purified exosomes secreted by cells impaired or not for macroautophagy by Atg5 depletion, during serum starvation conditions or complete medium culture. We show that the absence of serum modifies exosomal content, especially inducing secretion of two cytoplasmic protein complexes, namely proteasomal 19S regulatory particle (RP) and components of noncanonical translation preinitiation complex (PIC). This process is enhanced when autophagy is impaired by Atg5 depletion. Moreover, we show that the proteasome 20S core particle (CP) is released in the extracellular space. However, in striking contrast to what seen for its 19S RP regulator, release is independent of the exosomal vesicles, Atg5 expression and cell culture conditions. Exosome secretion can thus be considered as a cell process that participates in and reflects cell homeostasis, and care must be taken when studying potential extracellular function of exosomes due to the possible copurification of proteasome 20S CP.


Subject(s)
Exosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteome/metabolism , Autophagy , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Cell Line, Tumor , Culture Media, Serum-Free/pharmacology , Cytoplasmic Granules/metabolism , Eukaryotic Initiation Factors/metabolism , Exosomes/drug effects , Humans , Protein Transport , Ribosomal Proteins/metabolism , Vesicular Transport Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 115(28): E6477-E6486, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29934401

ABSTRACT

PA28γ is a nuclear activator of the 20S proteasome involved in the regulation of several essential cellular processes, such as cell proliferation, apoptosis, nuclear dynamics, and cellular stress response. Unlike the 19S regulator of the proteasome, which specifically recognizes ubiquitylated proteins, PA28γ promotes the degradation of several substrates by the proteasome in an ATP- and ubiquitin-independent manner. However, its exact mechanisms of action are unclear and likely involve additional partners that remain to be identified. Here we report the identification of a cofactor of PA28γ, PIP30/FAM192A. PIP30 binds directly and specifically via its C-terminal end and in an interaction stabilized by casein kinase 2 phosphorylation to both free and 20S proteasome-associated PA28γ. Its recruitment to proteasome-containing complexes depends on PA28γ and its expression increases the association of PA28γ with the 20S proteasome in cells. Further dissection of its possible roles shows that PIP30 alters PA28γ-dependent activation of peptide degradation by the 20S proteasome in vitro and negatively controls in cells the presence of PA28γ in Cajal bodies by inhibition of its association with the key Cajal body component coilin. Taken together, our data show that PIP30 deeply affects PA28γ interactions with cellular proteins, including the 20S proteasome, demonstrating that it is an important regulator of PA28γ in cells and thus a new player in the control of the multiple functions of the proteasome within the nucleus.


Subject(s)
Autoantigens/metabolism , Cell Nucleus/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteins/metabolism , Autoantigens/genetics , Cell Nucleus/genetics , HeLa Cells , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/genetics , Protein Binding , Protein Domains , Proteins/genetics
9.
Adv Exp Med Biol ; 1233: 55-100, 2020.
Article in English | MEDLINE | ID: mdl-32274753

ABSTRACT

The proteasome is involved in the regulation of all cellular pathways and consequently plays a central role in the control of cellular homeostasis. Together with its regulators, it is at the frontline, both as an actor and as a target, in human health and when homeostasis is disturbed in disease. In this review, we aim to provide an overview of the many levels at which the functions of the proteasome and its regulators can be regulated to cope with cellular needs or are altered in pathological conditions.


Subject(s)
Disease , Health , Proteasome Endopeptidase Complex , Humans , Proteasome Endopeptidase Complex/metabolism
10.
Int J Mol Sci ; 20(5)2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30813632

ABSTRACT

HslVU is an ATP-dependent proteolytic complex present in certain bacteria and in the mitochondrion of some primordial eukaryotes, including deadly parasites such as Leishmania. It is formed by the dodecameric protease HslV and the hexameric ATPase HslU, which binds via the C-terminal end of its subunits to HslV and activates it by a yet unclear allosteric mechanism. We undertook the characterization of HslV from Leishmania major (LmHslV), a trypanosomatid that expresses two isoforms for HslU, LmHslU1 and LmHslU2. Using a novel and sensitive peptide substrate, we found that LmHslV can be activated by peptides derived from the C-termini of both LmHslU1 and LmHslU2. Truncations, Ala- and D-scans of the C-terminal dodecapeptide of LmHslU2 (LmC12-U2) showed that five out of the six C-terminal residues of LmHslU2 are essential for binding to and activating HslV. Peptide cyclisation with a lactam bridge allowed shortening of the peptide without loss of potency. Finally, we found that dodecapeptides derived from HslU of other parasites and bacteria are able to activate LmHslV with similar or even higher efficiency. Importantly, using electron microscopy approaches, we observed that the activation of LmHslV was accompanied by a large conformational remodeling, which represents a yet unidentified layer of control of HslV activation.


Subject(s)
Leishmania major/enzymology , Peptides/pharmacology , Serine Endopeptidases/metabolism , Amino Acid Sequence , Enzyme Activation/drug effects , Peptides/chemistry , Protein Structure, Secondary , Recombinant Proteins/isolation & purification , Serine Endopeptidases/chemistry , Substrate Specificity
11.
J Biol Chem ; 291(25): 13147-59, 2016 Jun 17.
Article in English | MEDLINE | ID: mdl-27129254

ABSTRACT

The proteasome is an intracellular protease complex consisting of the 20S catalytic core and its associated regulators, including the 19S complex, PA28αß, PA28γ, PA200, and PI31. Inhibition of the proteasome induces autoregulatory de novo formation of 20S and 26S proteasome complexes. Formation of alternative proteasome complexes, however, has not been investigated so far. We here show that catalytic proteasome inhibition results in fast recruitment of PA28γ and PA200 to 20S and 26S proteasomes within 2-6 h. Rapid formation of alternative proteasome complexes did not involve transcriptional activation of PA28γ and PA200 but rather recruitment of preexisting activators to 20S and 26S proteasome complexes. Recruitment of proteasomal activators depended on the extent of active site inhibition of the proteasome with inhibition of ß5 active sites being sufficient for inducing recruitment. Moreover, specific inhibition of 26S proteasome activity via siRNA-mediated knockdown of the 19S subunit RPN6 induced recruitment of only PA200 to 20S proteasomes, whereas PA28γ was not mobilized. Here, formation of alternative PA200 complexes involved transcriptional activation of the activator. Alternative proteasome complexes persisted when cells had regained proteasome activity after pulse exposure to proteasome inhibitors. Knockdown of PA28γ sensitized cells to proteasome inhibitor-mediated growth arrest. Thus, formation of alternative proteasome complexes appears to be a formerly unrecognized but integral part of the cellular response to impaired proteasome function and altered proteostasis.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Autoantigens/metabolism , Bortezomib/pharmacology , Cells, Cultured , Gene Knockdown Techniques , Humans , Nuclear Proteins/metabolism , Oligopeptides/pharmacology , Proteasome Inhibitors/pharmacology , Protein Multimerization , Transcription, Genetic
12.
J Cell Sci ; 127(Pt 10): 2145-50, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24634511

ABSTRACT

Cyclin A2 is a key player in the regulation of the cell cycle. Its degradation in mid-mitosis relies on the ubiquitin-proteasome system (UPS). Using high-resolution microscopic imaging, we find that cyclin A2 persists beyond metaphase. Indeed, we identify a novel cyclin-A2-containing compartment that forms dynamic foci. Förster (or fluorescence) resonance energy transfer (FRET) and fluorescence lifetime imaging microscopy (FLIM) analyses show that cyclin A2 ubiquitylation takes place predominantly in these foci before spreading throughout the cell. Moreover, inhibition of autophagy in proliferating cells induces the stabilisation of a subset of cyclin A2, whereas induction of autophagy accelerates the degradation of cyclin A2, thus showing that autophagy is a novel regulator of cyclin A2 degradation.


Subject(s)
Autophagy/physiology , Cyclin A2/metabolism , Fluorescence Resonance Energy Transfer/methods , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Cell Communication , Humans , MCF-7 Cells , Microscopy, Fluorescence/methods
13.
Nat Cell Biol ; 9(3): 331-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17293853

ABSTRACT

The p300-CBP-associated factor (PCAF) is a histone acetyltransferase (HAT) involved in the reversible acetylation of various transcriptional regulators, including the tumour suppressor p53. It is implicated in many cellular processes, such as transcription, differentiation, proliferation and apoptosis. We observed that knockdown of PCAF expression in HeLa or U2OS cell lines induces stabilization of the oncoprotein Hdm2, a RING finger E3 ligase primarily known for its role in controlling p53 stability. To investigate the molecular basis of this effect, we examined whether PCAF is involved in Hdm2 ubiquitination. Here, we show that PCAF, in addition to its acetyltransferase activity, possesses an intrinsic ubiquitination activity that is critical for controlling Hdm2 expression levels, and thus p53 functions. Our data highlight a regulatory crosstalk between PCAF and Hdm2 activities, which is likely to have a central role in the subtle control of p53 activity after DNA damage.


Subject(s)
Cell Cycle Proteins/metabolism , Histone Acetyltransferases/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Binding Sites/genetics , Catalytic Domain/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage/drug effects , DNA Damage/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , HeLa Cells , Histone Acetyltransferases/genetics , Humans , Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Antisense/genetics , RNA, Small Interfering/genetics , Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ultraviolet Rays , Zinostatin/pharmacology , p300-CBP Transcription Factors
14.
Biochimie ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38901793

ABSTRACT

Proteins are essential molecular actors in every cellular process. From their synthesis to their degradation, they are subject to continuous quality control mechanisms to ensure that they fulfil cellular needs in proper and timely fashion. Proteostasis is a key process allowing cells or organisms to maintain an appropriate but dynamic equilibrium of their proteome (the ensemble of all their proteins). It relies on multiple mechanisms that together control the level, fate and function of individual proteins, and ensure elimination of abnormal ones. The proteostasis network is essential for development and adaptation to environmental changes or challenges. Its dysfunctions can lead to accumulation of deleterious proteins or, conversely, to excessive degradation of beneficial ones, and are implicated in many diseases such as cancers, neurodegeneration, or developmental and aging disorders. Manipulating this network to control abundance of selected target proteins is therefore a strategy with enormous therapeutic or biotechnological potential. The ProteoCure COST Action gathers more than 350 researchers and their teams (31 countries represented) from the academic, clinical, and industrial sectors, who share the conviction that our understanding of proteostasis is mature enough to develop novel and highly specific therapies based on selective tuning of protein levels. Towards this objective, the Action organizes community-building activities to foster synergies among its participants and reinforce training of the next generation of European researchers. Its ambition is to function as a knowledge-based network and a creative exchange hub on normal and pathologic proteostasis, focusing on developing innovative tools modulating the level of specific protein(s).

15.
Biol Cell ; 104(3): 165-87, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22188301

ABSTRACT

The modification of intracellular proteins by ubiquitin (Ub) and ubiquitin-like (UbL) proteins is a central mechanism for regulating and fine-tuning all cellular processes. Indeed, these modifications are widely used to control the stability, activity and localisation of many key proteins and, therefore, they are instrumental in regulating cellular functions as diverse as protein degradation, cell signalling, vesicle trafficking and immune response. It is thus no surprise that pathogens in general, and viruses in particular, have developed multiple strategies to either counteract or exploit the complex mechanisms mediated by the Ub and UbL protein conjugation pathways. The aim of this review is to provide an overview on the intricate and conflicting relationships that intimately link HIV-1 and these sophisticated systems of post-translational modifications.


Subject(s)
HIV Infections/physiopathology , HIV-1/metabolism , Protein Processing, Post-Translational , Ubiquitin/metabolism , Ubiquitins/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Nucleus/virology , Cytoplasm/metabolism , Humans , Ubiquitin/genetics , Ubiquitins/genetics
16.
PLoS Pathog ; 6(3): e1000808, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20333243

ABSTRACT

Viruses use cellular machinery to enter and infect cells. In this study we address the cell entry mechanisms of nonenveloped adenoviruses (Ads). We show that protein VI, an internal capsid protein, is rapidly exposed after cell surface attachment and internalization and remains partially associated with the capsid during intracellular transport. We found that a PPxY motif within protein VI recruits Nedd4 E3 ubiquitin ligases to bind and ubiquitylate protein VI. We further show that this PPxY motif is involved in rapid, microtubule-dependent intracellular movement of protein VI. Ads with a mutated PPxY motif can efficiently escape endosomes but are defective in microtubule-dependent trafficking toward the nucleus. Likewise, depletion of Nedd4 ligases attenuates nuclear accumulation of incoming Ad particles and infection. Our data provide the first evidence that virus-encoded PPxY motifs are required during virus entry, which may be of significance for several other pathogens.


Subject(s)
Adenovirus Infections, Human/virology , Adenoviruses, Human/growth & development , Adenoviruses, Human/genetics , Capsid Proteins/genetics , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cell Line, Tumor , Conserved Sequence , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Endosomes/virology , Epithelial Cells/cytology , Epithelial Cells/virology , Humans , Lung/cytology , Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/virology , Microtubules/metabolism , Microtubules/virology , Nedd4 Ubiquitin Protein Ligases , Osteosarcoma , Protein Structure, Tertiary , Retinal Pigment Epithelium/cytology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology
17.
Cancers (Basel) ; 14(4)2022 Feb 12.
Article in English | MEDLINE | ID: mdl-35205670

ABSTRACT

Protein ubiquitylation coordinates crucial cellular events in physiological and pathological conditions. A comparative analysis of the ubiquitin proteome from bortezomib (BTZ)-sensitive and BTZ-resistant mantle cell lymphoma (MCL) revealed an enrichment of the autophagy-lysosome system (ALS) in BTZ-resistant cells. Pharmacological inhibition of autophagy at the level of lysosome-fusion revealed a constitutive activation of proteaphagy and accumulation of proteasome subunits within autophagosomes in different MCL cell lines with acquired or natural resistance to BTZ. Inhibition of the autophagy receptor p62/SQSTM1 upon verteporfin (VTP) treatment disrupted proteaphagosome assembly, reduced co-localization of proteasome subunits with autophagy markers and negatively impacted proteasome activity. Finally, the silencing or pharmacological inhibition of p62 restored the apoptosis threshold at physiological levels in BTZ-resistant cells both in vitro and in vivo. In total, these results demonstrate for the first time a proteolytic switch from the ubiquitin-proteasome system (UPS) to ALS in B-cell lymphoma refractory to proteasome inhibition, pointing out a crucial role for proteaphagy in this phenomenon and paving the way for the design of alternative therapeutic venues in treatment-resistant tumors.

18.
Nat Cell Biol ; 5(8): 754-61, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12883554

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) encodes a potent transactivator, Tat, which functions through binding to a short leader RNA, called transactivation responsive element (TAR). Recent studies suggest that Tat activates the HIV-1 long terminal repeat (LTR), mainly by adapting co-activator complexes, such as p300, PCAF and the positive transcription elongation factor P-TEFb, to the promoter. Here, we show that the proto-oncoprotein Hdm2 interacts with Tat and mediates its ubiquitination in vitro and in vivo. In addition, Hdm2 is a positive regulator of Tat-mediated transactivation, indicating that the transcriptional properties of Tat are stimulated by ubiquitination. Fusion of ubiquitin to Tat bypasses the requirement of Hdm2 for efficient transactivation, supporting the notion that ubiquitin has a non-proteolytic function in Tat-mediated transactivation.


Subject(s)
Gene Products, tat/metabolism , HIV-1/genetics , Nuclear Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Transcriptional Activation , Ubiquitin/metabolism , Cell Line , Gene Products, tat/genetics , HIV Long Terminal Repeat , HIV-1/metabolism , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , RNA, Small Interfering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , tat Gene Products, Human Immunodeficiency Virus
19.
Biochem Soc Trans ; 38(Pt 1): 98-103, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20074043

ABSTRACT

The critical tumour suppressor p53 plays a major role in response to DNA damage and, more generally, to genotoxic stress. The regulation of its expression and functions is under very tight controls, and involves, in particular, an extremely complex set of post-translational modifications, thanks to a variety of 'modifiers', including ubiquitylation E3s and acetyltransferases, that fine-tune the stability and activity of the protein. Work of the last few years has revealed that, in addition to targeting p53, these modifiers also modify each other, forming an intricate network of regulatory molecules and events that must be taken into account to understand p53 regulation. We propose that this network allows a metastable equilibrium that confers both sensitivity and robustness on the p53 pathway, two properties that allow the pathway to respectively answer to a variety of stimuli and return to its initial stage when the stimuli disappear.


Subject(s)
Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , Acetylation , Acetyltransferases/metabolism , Animals , DNA Damage , Humans , Protein Processing, Post-Translational , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
20.
Oncogene ; 39(37): 6009-6023, 2020 09.
Article in English | MEDLINE | ID: mdl-32772043

ABSTRACT

Aurora B kinase plays essential roles in mitosis. Its protein levels increase before the onset of mitosis and sharply decrease during mitosis exit. The latter decrease is due to a balance between the actions of the E3 ubiquitin ligase anaphase-promoting complex or cyclosome (activated by the Cdh1 adapter), and the deubiquitinating enzyme USP35. Aurora B also executes important functions in interphase. Abnormal modulation of Aurora B in interphase leads to cell cycle defects often linked to aberrant chromosomal condensation and segregation. Very little is however known about how Aurora B levels are regulated in interphase. Here we found that USP13-associates with and stabilizes Aurora B in cells, especially before their entry into mitosis. In order for USP13 to exert its stabilizing effect on Aurora B, their association is promoted by the Aurora B-mediated phosphorylation of USP13 at Serine 114. We also present evidence that USP13 instigates Aurora B deubiquitination and/or protect it from degradation in a non-catalytic manner. In addition, we report that genetic or chemical modulation of the cellular levels/activity of USP13 affects unperturbed cell-cycle progression. Overall our study unveils the molecular and cellular connections of the USP13-Aurora B axis, which potentially participates in the rewiring of the cell cycle happening in cancer cells.


Subject(s)
Aurora Kinase B/metabolism , Cell Cycle/genetics , Endopeptidases/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Disease Progression , Endopeptidases/metabolism , Enzyme Stability , Gene Expression , Gene Knockdown Techniques , Humans , Phosphorylation , Protein Binding , Serine/metabolism , Ubiquitin-Specific Proteases
SELECTION OF CITATIONS
SEARCH DETAIL