Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
2.
Cell Rep ; 42(8): 112925, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37552601

ABSTRACT

The neddylation inhibitor MLN4924/Pevonedistat is in clinical trials for multiple cancers. Efficacy is generally attributed to cullin RING ligase (CRL) inhibition, but the contribution of non-CRL targets is unknown. Here, CRISPR screens map MLN4924-monotherapy sensitivity in retinoblastoma to a classic DNA damage-induced p53/E2F3/BAX-dependent death effector network, which synergizes with Nutlin3a or Navitoclax. In monotherapy-resistant cells, MLN4924 plus standard-of-care topotecan overcomes resistance, but reduces DNA damage, instead harnessing ribosomal protein nucleolar-expulsion to engage an RPL11/p21/MYCN/E2F3/p53/BAX synergy network that exhibits extensive cross-regulation. Strikingly, unneddylatable RPL11 substitutes for MLN4924 to perturb nucleolar function and enhance topotecan efficacy. Orthotopic tumors exhibit complete responses while preserving visual function. Moreover, MLN4924 plus melphalan deploy this DNA damage-independent strategy to synergistically kill multiple myeloma cells. Thus, MLN4924 synergizes with standard-of-care drugs to unlock a nucleolar death effector network across cancer types implying broad therapeutic relevance.


Subject(s)
Topotecan , Tumor Suppressor Protein p53 , bcl-2-Associated X Protein , Cell Line, Tumor , Cyclopentanes/pharmacology , Ribosomal Proteins , Apoptosis , NEDD8 Protein
3.
Sci Adv ; 9(13): eabq7585, 2023 03 31.
Article in English | MEDLINE | ID: mdl-37000881

ABSTRACT

The elimination of aberrant inclusions is regarded as a therapeutic approach in neurodegeneration. In amyotrophic lateral sclerosis (ALS), mutations in proteins found within cytoplasmic condensates called stress granules (SGs) are linked to the formation of pathological SGs, aberrant protein inclusions, and neuronal toxicity. We found that inhibition of NEDP1, the enzyme that processes/deconjugates the ubiquitin-like molecule NEDD8, promotes the disassembly of physiological and pathological SGs. Reduction in poly(ADP-ribose) polymerase1 activity through hyper-NEDDylation is a key mechanism for the observed phenotype. These effects are related to improved cell survival in human cells, and in C. elegans, nedp1 deletion ameliorates ALS phenotypes related to animal motility. Our studies reveal NEDP1 as potential therapeutic target for ALS, correlated to the disassembly of pathological SGs.


Subject(s)
Amyotrophic Lateral Sclerosis , Animals , Humans , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Caenorhabditis elegans/genetics , Stress Granules , Ubiquitin , Phenotype
4.
Methods Mol Biol ; 2602: 137-149, 2023.
Article in English | MEDLINE | ID: mdl-36446972

ABSTRACT

The identification of modification sites for ubiquitin and ubiquitin-like modifiers is an essential step in the elucidation of controlled processes. The ubiquitin-like modifier NEDD8 is an important regulator of plethora of biological processes both under homeostatic and proteotoxic stress conditions. Here, we describe a detailed protocol for proteome-wide identification of NEDDylation sites. The approach is based on the use of cell lines stably expressing the NEDD8R74K mutant. Digestion of samples with Lysyl endopeptidase generates peptides with a di-glycine remnant only from proteins modified with NEDD8R74K but not with ubiquitin or ISG15. The isolation of these peptides with anti-di-glycine antibodies (K-ε-GG) allows the identification of NEDDylation sites by liquid chromatography tandem mass spectrometry (LC-MS/MS).


Subject(s)
Fabaceae , Ubiquitin , Chromatography, Liquid , Tandem Mass Spectrometry , Proteome , Glycine
5.
Semin Cell Dev Biol ; 132: 27-37, 2022 12.
Article in English | MEDLINE | ID: mdl-35078718

ABSTRACT

Post-translational modification of proteins with the Ubiquitin-like molecule NEDD8 is a critical regulatory mechanism for several biological processes and a potential target for therapeutic intervention. The role of NEDD8 has been mainly characterised through its modification as single moiety on the cullin family of proteins and control of Cullin-Ring-Ligases, but also on non-cullin substrates. In addition to monoNEDDylation, recent studies have now revealed that NEDD8 can also generate diverse polymers. This is either through modification of the 9 available lysines in NEDD8 and the formation of polyNEDD8 chains, or NEDDylation of Ubiquitin and SUMO-2 for the generation of hybrid NEDD8 chains. Here, we review recent findings that characterise the formation of NEDD8 polymers under distinct modes of protein NEDDylation (canonical/atypical) and their potential role as regulatory signals of the proteotoxic stress response and the Protein Quality Control system.


Subject(s)
Polymers , Ubiquitins , Ubiquitins/metabolism , Ubiquitin/metabolism , Cullin Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
6.
Biochem Soc Trans ; 49(3): 1075-1083, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34156462

ABSTRACT

Molecular chaperones are essential components of the protein quality control system and maintenance of homeostasis. Heat Shock Protein 70 (HSP70), a highly evolutionarily conserved family of chaperones is a key regulator of protein folding, oligomerisation and prevents the aggregation of misfolded proteins. HSP70 chaperone function depends on the so-called 'HSP70-cycle', where HSP70 interacts with and is released from substrates via ATP hydrolysis and the assistance of HSP70 co-factors/co-chaperones, which also provide substrate specificity. The identification of regulatory modules for HSP70 allows the elucidation of HSP70 specificity and target selectivity. Here, we discuss how the HSP70 cycle is functionally linked with the cycle of the Ubiquitin-like molecule NEDD8. Using as an example the DNA damage response, we present a model where HSP70 acts as a sensor of the NEDD8 cycle. The NEDD8 cycle acts as a regulatory module of HSP70 activity, where conversion of poly-NEDD8 chains into mono-NEDD8 upon DNA damage activates HSP70, facilitating the formation of the apoptosome and apoptosis execution.


Subject(s)
DNA Damage , DNA Repair , HSP70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , NEDD8 Protein/metabolism , Adenosine Triphosphate/metabolism , Animals , DNA/genetics , DNA/metabolism , Humans , Hydrolysis
7.
Mol Metab ; 53: 101275, 2021 11.
Article in English | MEDLINE | ID: mdl-34153521

ABSTRACT

OBJECTIVE: Neddylation is a druggable and reversible ubiquitin-like post-translational modification upregulated in many diseases, including liver fibrosis, hepatocellular carcinoma, and more recently, non-alcoholic fatty liver disease (NAFLD). Herein, we propose to address the effects of neddylation inhibition and the underlying mechanisms in pre-clinical models of NAFLD. METHODS: Hepatic neddylation measured by immunohistochemical analysis and NEDD8 serum levels measured by ELISA assay were evaluated in NAFLD clinical and pre-clinical samples. The effects of neddylation inhibition by using a pharmacological small inhibitor, MLN4924, or molecular approaches were assessed in isolated mouse hepatocytes and pre-clinical mouse models of diet-induced NAFLD, male adult C57BL/6 mice, and the AlfpCre transgenic mice infected with AAV-DIO-shNedd8. RESULTS: Neddylation inhibition reduced lipid accumulation in oleic acid-stimulated mouse primary hepatocytes and ameliorated liver steatosis, preventing lipid peroxidation and inflammation in the mouse models of diet-induced NAFLD. Under these conditions, increased Deptor levels and the concomitant repression of mTOR signaling were associated with augmented fatty acid oxidation and reduced lipid content. Moreover, Deptor silencing in isolated mouse hepatocytes abolished the anti-steatotic effects mediated by neddylation inhibition. Finally, serum NEDD8 levels correlated with hepatic neddylation during the disease progression in the clinical and pre-clinical models CONCLUSIONS: Overall, the upregulation of Deptor, driven by neddylation inhibition, is proposed as a novel effective target and therapeutic approach to tackle NAFLD.


Subject(s)
Fatty Acids/metabolism , Hepatocytes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , TOR Serine-Threonine Kinases/metabolism , Adolescent , Adult , Aged , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Non-alcoholic Fatty Liver Disease/chemically induced , Signal Transduction , Young Adult
8.
Cell Rep ; 34(3): 108635, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33472076

ABSTRACT

The ubiquitin-like molecule NEDD8 controls several biological processes and is a promising target for therapeutic intervention. NEDDylation occurs through specific NEDD8 enzymes (canonical) or enzymes of the ubiquitin system (atypical). Identification of NEDD8 sites on substrates is critical for delineating the processes controlled by NEDDylation. By combining the use of the NEDD8 R74K mutant with anti-di-glycine (anti-diGly) antibodies, we identified 1,101 unique NEDDylation sites in 620 proteins. Bioinformatics analysis reveals that canonical and atypical NEDDylation have distinct proteomes; the spliceosome/mRNA surveillance/DNA replication and ribosome/proteasome, respectively. The data also reveal the formation of poly-NEDD8, hybrid NEDD8-ubiquitin, and NEDD8-SUMO-2 chains as potential molecular signals. In particular, NEDD8-SUMO-2 chains are induced upon proteotoxic stress (atypical) through NEDDylation of K11 in SUMO-2, and conjugates accumulate in previously described nucleolus-related inclusions. The study uncovers a diverse proteome for NEDDylation and is consistent with the concept of extensive cross-talk between ubiquitin and Ubls under proteotoxic stress conditions.


Subject(s)
NEDD8 Protein/metabolism , Proteome/metabolism , Catalytic Domain , Cell Nucleolus/metabolism , Endopeptidases/metabolism , HCT116 Cells , Humans , NEDD8 Protein/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism
9.
Mol Cell ; 79(1): 155-166.e9, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32454028

ABSTRACT

To understand gene function, the encoding DNA or mRNA transcript can be manipulated and the consequences observed. However, these approaches do not have a direct effect on the protein product of the gene, which is either permanently abrogated or depleted at a rate defined by the half-life of the protein. We therefore developed a single-component system that could induce the rapid degradation of the specific endogenous protein itself. A construct combining the RING domain of ubiquitin E3 ligase RNF4 with a protein-specific camelid nanobody mediates target destruction by the ubiquitin proteasome system, a process we describe as antibody RING-mediated destruction (ARMeD). The technique is highly specific because we observed no off-target protein destruction. Furthermore, bacterially produced nanobody-RING fusion proteins electroporated into cells induce degradation of target within minutes. With increasing availability of protein-specific nanobodies, this method will allow rapid and specific degradation of a wide range of endogenous proteins.


Subject(s)
Endopeptidases/metabolism , NEDD8 Protein/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Single-Domain Antibodies/metabolism , Transcription Factors/metabolism , Ubiquitin/metabolism , Endopeptidases/immunology , HeLa Cells , Humans , NEDD8 Protein/immunology , Nuclear Proteins/immunology , Proteasome Endopeptidase Complex/immunology , Proteolysis , Single-Domain Antibodies/immunology , Transcription Factors/immunology , Ubiquitination
10.
Cell Rep ; 29(1): 212-224.e8, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31577950

ABSTRACT

Ubiquitin and ubiquitin-like chains are finely balanced by conjugating and de-conjugating enzymes. Alterations in this balance trigger the response to stress conditions and are often observed in pathologies. How such changes are detected is not well understood. We identify the HSP70 chaperone as a sensor of changes in the balance between mono- and poly-NEDDylation. Upon DNA damage, the induction of the de-NEDDylating enzyme NEDP1 restricts the formation of NEDD8 chains, mainly through lysines K11/K48. This promotes APAF1 oligomerization and apoptosis induction, a step that requires the HSP70 ATPase activity. HSP70 binds to NEDD8, and, in vitro, the conversion of NEDD8 chains into mono-NEDD8 stimulates HSP70 ATPase activity. This effect is independent of NEDD8 conjugation onto substrates. The study indicates that the NEDD8 cycle is a regulatory module of HSP70 function. These findings may be important in tumorigenesis, as we find decreased NEDP1 levels in hepatocellular carcinoma with concomitant accumulation of NEDD8 conjugates.


Subject(s)
Adenosine Triphosphatases/genetics , DNA Damage/genetics , Endopeptidases/genetics , HSP70 Heat-Shock Proteins/genetics , NEDD8 Protein/genetics , Amino Acid Sequence , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Female , Humans , Lysine/genetics , MCF-7 Cells , Mice , Ubiquitin/genetics
11.
FASEB J ; 33(1): 643-651, 2019 01.
Article in English | MEDLINE | ID: mdl-30024791

ABSTRACT

The ribosomal protein L11 (RPL11) integrates different types of stress into a p53-mediated response. Here, we analyzed the impact of the ubiquitin-like protein SUMO on the RPL11-mouse double-minute 2 homolog-p53 signaling. We show that small ubiquitin-related modifier (SUMO)1 and SUMO2 covalently modify RPL11. We find that SUMO negatively modulates the conjugation of the ubiquitin-like protein neural precursor cell-expressed developmentally downregulated 8 (NEDD8) to RPL11 and promotes the translocation of the RP outside of the nucleoli. Moreover, the SUMO-conjugating enzyme, Ubc9, is required for RPL11-mediated activation of p53. SUMOylation of RPL11 is triggered by ribosomal stress, as well as by alternate reading frame protein upregulation. Collectively, our data identify SUMO protein conjugation to RPL11 as a new regulator of the p53-mediated cellular response to different types of stress and reveal a previously unknown SUMO-NEDD8 interplay.-El Motiam, A., Vidal, S., de la Cruz-Herrera, C. F., Da Silva-Álvarez, S., Baz-Martínez, M., Seoane, R., Vidal, A., Rodríguez, M. S., Xirodimas, D. P., Carvalho, A. S., Beck, H. C., Matthiesen, R., Collado, M., Rivas, C. Interplay between SUMOylation and NEDDylation regulates RPL11 localization and function.


Subject(s)
NEDD8 Protein/metabolism , Neoplasms/pathology , Protein Processing, Post-Translational , Ribosomal Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Ubiquitins/metabolism , HEK293 Cells , Humans , Neoplasms/metabolism , Tumor Cells, Cultured
12.
Nat Commun ; 9(1): 4376, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30349034

ABSTRACT

Spatial management of stress-induced protein aggregation is an integral part of the proteostasis network. Protein modification by the ubiquitin-like molecule NEDD8 increases upon proteotoxic stress and it is characterised by the formation of hybrid NEDD8/ubiquitin conjugates. However, the biological significance of this response is unclear. Combination of quantitative proteomics with biological analysis shows that, during proteotoxic stress, NEDDylation promotes nuclear protein aggregation, including ribosomal proteins as a major group. This correlates with protection of the nuclear Ubiquitin Proteasome System from stress-induced dysfunction. Correspondingly, we show that NEDD8 compromises ubiquitination and prevents targeting and processing of substrates by the proteasome. Moreover, we identify HUWE1 as a key E3-ligase that is specifically required for NEDDylation during proteotoxic stress. The study reveals a specific role for NEDD8 in nuclear protein aggregation upon stress and is consistent with the concept that transient aggregate formation is part of a defence mechanism against proteotoxicity.


Subject(s)
NEDD8 Protein/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitins/metabolism , Blotting, Western , Cell Line, Tumor , HEK293 Cells , Humans , Microscopy, Fluorescence , NEDD8 Protein/genetics , Nuclear Proteins/genetics , Proteomics , Real-Time Polymerase Chain Reaction , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/genetics , Ubiquitination/physiology , Ubiquitins/genetics
13.
Cell Rep ; 18(7): 1791-1803, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28199849

ABSTRACT

How metazoan genomes are structured at the nanoscale in living cells and tissues remains unknown. Here, we adapted a quantitative FRET (Förster resonance energy transfer)-based fluorescence lifetime imaging microscopy (FLIM) approach to assay nanoscale chromatin compaction in living organisms. Caenorhabditis elegans was chosen as a model system. By measuring FRET between histone-tagged fluorescent proteins, we visualized distinct chromosomal regions and quantified the different levels of nanoscale compaction in meiotic cells. Using RNAi and repetitive extrachromosomal array approaches, we defined the heterochromatin state and showed that its architecture presents a nanoscale-compacted organization controlled by Heterochromatin Protein-1 (HP1) and SETDB1 H3-lysine-9 methyltransferase homologs in vivo. Next, we functionally explored condensin complexes. We found that condensin I and condensin II are essential for heterochromatin compaction and that condensin I additionally controls lowly compacted regions. Our data show that, in living animals, nanoscale chromatin compaction is controlled not only by histone modifiers and readers but also by condensin complexes.


Subject(s)
Adenosine Triphosphatases/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , Multiprotein Complexes/genetics , Animals , Caenorhabditis elegans/genetics , Cells, Cultured , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomes/genetics , Fluorescence Resonance Energy Transfer/methods , Heterochromatin/metabolism , Histones/genetics , Microscopy, Fluorescence/methods
14.
Sci Rep ; 6: 37775, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27901050

ABSTRACT

Targetting the ubiquitin pathway is an attractive strategy for cancer therapy. The inhibitor of the ubiquitin-like molecule NEDD8 pathway, MLN4924 (Pevonedistat) is in Phase II clinical trials. Protection of healthy cells from the induced toxicity of the treatment while preserving anticancer efficacy is a highly anticipated outcome in chemotherapy. Cyclotherapy was proposed as a promising approach to achieve this goal. We found that cytostatic activation of p53 protects cells against MLN4924-induced toxicity and importantly the effects are reversible. In contrast, cells with mutant or no p53 remain sensitive to NEDD8 inhibition. Using zebrafish embryos, we show that MLN4924-induced apoptosis is reduced upon pre-treatment with actinomycin D in vivo. Our studies show that the cellular effects of NEDD8 inhibition can be manipulated based on the p53 status and that NEDD8 inhibitors can be used in a p53-based cyclotherapy protocol to specifically target cancer cells devoid of wild type p53 function, while healthy cells will be protected from the induced toxicity.


Subject(s)
Cyclopentanes/pharmacology , NEDD8 Protein/antagonists & inhibitors , Pyrimidines/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Line, Tumor , HCT116 Cells , Humans , Ubiquitins/metabolism
15.
Endocr Relat Cancer ; 22(1): T55-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25504797

ABSTRACT

Post-translational modification of proteins with ubiquitin and ubiquitin-like molecules (UBLs) controls a vast if not every biological process in the cell. It is not surprising that deregulation in ubiquitin and UBL signalling has been implicated in the pathogenesis of many diseases and that these pathways are considered as major targets for therapeutic intervention. In this review, we summarise recent advances in our understanding of the role of the UBL neural precursor cell expressed developmentally downregulated-8 (NEDD8) in cancer-related processes and potential strategies for the use of NEDD8 inhibitors as chemotherapeutics.


Subject(s)
Neoplasms/drug therapy , Neoplasms/enzymology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Neoplasms/metabolism , Protein Processing, Post-Translational , Ubiquitin/metabolism
17.
Biochem J ; 449(3): 707-17, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23134341

ABSTRACT

Understanding the determinants for site-specific ubiquitination by E3 ligase components of the ubiquitin machinery is proving to be a challenge. In the present study we investigate the role of an E3 ligase docking site (Mf2 domain) in an intrinsically disordered domain of IRF-1 [IFN (interferon) regulatory factor-1], a short-lived IFNγ-regulated transcription factor, in ubiquitination of the protein. Ubiquitin modification of full-length IRF-1 by E3 ligases such as CHIP [C-terminus of the Hsc (heat-shock cognate) 70-interacting protein] and MDM2 (murine double minute 2), which dock to the Mf2 domain, was specific for lysine residues found predominantly in loop structures that extend from the DNA-binding domain, whereas no modification was detected in the more conformationally flexible C-terminal half of the protein. The E3 docking site was not available when IRF-1 was in its DNA-bound conformation and cognate DNA-binding sequences strongly suppressed ubiquitination, highlighting a strict relationship between ligase binding and site-specific modification at residues in the DNA-binding domain. Hyperubiquitination of a non-DNA-binding mutant supports a mechanism where an active DNA-bound pool of IRF-1 is protected from polyubiquitination and degradation.


Subject(s)
DNA/metabolism , Interferon Regulatory Factor-1/chemistry , Interferon Regulatory Factor-1/metabolism , Amino Acid Sequence , Binding Sites/genetics , Cell Line , Humans , Interferon Regulatory Factor-1/genetics , Lysine/chemistry , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-mdm2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
18.
Bull Math Biol ; 74(7): 1531-79, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22527944

ABSTRACT

There are many intracellular signalling pathways where the spatial distribution of the molecular species cannot be neglected. These pathways often contain negative feedback loops and can exhibit oscillatory dynamics in space and time. Two such pathways are those involving Hes1 and p53-Mdm2, both of which are implicated in cancer. In this paper we further develop the partial differential equation (PDE) models of Sturrock et al. (J. Theor. Biol., 273:15-31, 2011) which were used to study these dynamics. We extend these PDE models by including a nuclear membrane and active transport, assuming that proteins are convected in the cytoplasm towards the nucleus in order to model transport along microtubules. We also account for Mdm2 inhibition of p53 transcriptional activity. Through numerical simulations we find ranges of values for the model parameters such that sustained oscillatory dynamics occur, consistent with available experimental measurements. We also find that our model extensions act to broaden the parameter ranges that yield oscillations. Hence oscillatory behaviour is made more robust by the inclusion of both the nuclear membrane and active transport. In order to bridge the gap between in vivo and in silico experiments, we investigate more realistic cell geometries by using an imported image of a real cell as our computational domain. For the extended p53-Mdm2 model, we consider the effect of microtubule-disrupting drugs and proteasome inhibitor drugs, obtaining results that are in agreement with experimental studies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Models, Biological , Nuclear Envelope/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Animals , Cell Shape , Cells, Cultured , Computer Simulation , Enzyme Inhibitors/pharmacology , Mice , Microtubules/drug effects , Microtubules/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Transcription Factor HES-1
19.
Cell Cycle ; 11(8): 1646-55, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22487680

ABSTRACT

The discovery that the single p53 gene encodes several different p53 protein isoforms has initiated a flurry of research into the function and regulation of these novel p53 proteins. Full-length p53 protein level is primarily regulated by the E3-ligase Mdm2, which promotes p53 ubiquitination and degradation. Here, we report that all of the novel p53 isoforms are ubiquitinated and degraded to varying degrees in an Mdm2-dependent and -independent manner, and that high-risk human papillomavirus can degrade some but not all of the novel isoforms, demonstrating that full-length p53 and the p53 isoforms are differentially regulated. In addition, we provide the first evidence that Mdm2 promotes the NEDDylation of p53ß. Altogether, our data indicates that Mdm2 can distinguish between the p53 isoforms and modify them differently.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , Leupeptins/chemistry , Leupeptins/pharmacology , Protein Isoforms/metabolism , Proteolysis/drug effects , Ubiquitination
20.
Methods Mol Biol ; 832: 133-40, 2012.
Article in English | MEDLINE | ID: mdl-22350881

ABSTRACT

Identification of substrates for post-translational modification with ubiquitin and ubiquitin-like molecules is a key in our understanding of the role of this modification in the regulation of biological processes. The ubiquitin-like molecule NEDD8 plays a vital role in cell growth, viability, and development. In this -chapter, we describe a method for efficient isolation and detection of NEDDylated proteins in human cells.


Subject(s)
Ubiquitin/metabolism , Ubiquitins/metabolism , Blotting, Western/methods , Cell Line , Humans , NEDD8 Protein , Protein Processing, Post-Translational , Ubiquitins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...