Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Biophys Res Commun ; 558: 224-230, 2021 06 18.
Article in English | MEDLINE | ID: mdl-32933748

ABSTRACT

The NF-κB transcription factor is involved in inflammation and cell proliferation, survival, and transformation. It is a heterodimer made of p50 or p52 and a member of the Rel family of proteins. p50 and p52 are derived from limited ubiquitin- and proteasome-mediated proteolytic processing of the larger precursors p105 and p100, respectively. Both precursors can be either processed or completely degraded by the ubiquitin-proteasome system. Previous work in our laboratory identified KPC1 as a ubiquitin ligase that mediates processing of p105 to the p50 subunit. Overexpression of the ligase leads to increased level of p50 with a resultant marked tumor-suppressive effect. In the present study, we identify FBXO7, a known ubiquitin ligase that binds to p105 and ubiquitinates it, but surprisingly, leads to its accumulation and to that of p65 - the Rel partner of p50 - and to increased cell proliferation. Importantly, a ΔF-Box mutant of FBXO7 which is inactive has similar effects on accumulation of p105 and cell proliferation, strongly suggesting that p105 is a pseudo substrate of FBXO7.


Subject(s)
F-Box Proteins/metabolism , NF-kappa B p50 Subunit/metabolism , Amino Acid Sequence , Cell Line , Cell Proliferation/physiology , Enzyme Stability , F-Box Proteins/antagonists & inhibitors , F-Box Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Mutant Proteins/genetics , Mutant Proteins/metabolism , NF-kappa B p50 Subunit/antagonists & inhibitors , NF-kappa B p50 Subunit/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Precursors/metabolism , Protein Processing, Post-Translational , Proteolysis , RNA, Small Interfering/genetics , Substrate Specificity , Transcription Factor RelA/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitination
2.
Nat Chem ; 11(7): 644-652, 2019 07.
Article in English | MEDLINE | ID: mdl-31182821

ABSTRACT

A promising approach in cancer therapy is to find ligands that directly bind ubiquitin (Ub) chains. However, finding molecules capable of tightly and specifically binding Ub chains is challenging given the range of Ub polymer lengths and linkages and their subtle structural differences. Here, we use total chemical synthesis of proteins to generate highly homogeneous Ub chains for screening against trillion-member macrocyclic peptide libraries (RaPID system). De novo cyclic peptides were found that can bind tightly and specifically to K48-linked Ub chains, confirmed by NMR studies. These cyclic peptides protected K48-linked Ub chains from deubiquitinating enzymes and prevented proteasomal degradation of Ub-tagged proteins. The cyclic peptides could enter cells, inhibit growth and induce programmed cell death, opening new opportunities for therapeutic intervention. This highly synthetic approach, with both protein target generation and cyclic peptide discovery performed in vitro, will make other elaborate post-translationally modified targets accessible for drug discovery.


Subject(s)
Lysine/chemistry , Peptides, Cyclic/metabolism , Small Molecule Libraries/metabolism , Ubiquitins/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Deubiquitinating Enzymes , HeLa Cells , Humans , Molecular Structure , Peptides, Cyclic/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/metabolism , Proteasome Inhibitors/pharmacology , Protein Binding , Small Molecule Libraries/pharmacology , Ubiquitins/chemical synthesis , Ubiquitins/chemistry
3.
Proc Natl Acad Sci U S A ; 116(16): 7805-7812, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30867293

ABSTRACT

One of the enigmas in the ubiquitin (Ub) field is the requirement for a poly-Ub chain as a proteasomal targeting signal. The canonical chain appears to be longer than the distance between the two Ub-binding proteasomal receptors. Furthermore, genetic manipulation has shown that one receptor subunit is sufficient, which suggests that a single Ub can serve as a degradation signal. To shed light on this mystery, we chemically synthesized tetra-Ub, di-Ub (K48-based), and mono-Ub adducts of HA-α-globin, where the distal or proximal Ub moieties were tagged differentially with either Myc or Flag. When incubated in a crude cell extract, the distal Ub moiety in the tetra-Ub adduct was mostly removed by deubiquitinating enzymes (DUBs) and reconjugated to other substrates in the extract. In contrast, the proximal moiety was most likely degraded with the substrate. The efficacy of degradation was proportionate to the chain length; while tetra-Ub globin was an efficient substrate, with mono-Ub globin, we observed rapid removal of the Ub moiety with almost no degradation of the free globin. Taken together, these findings suggest that the proximal moieties are necessary for securing the association of the substrate with the proteasome along the proteolytic process, whereas the distal moieties are important in protecting the proximal moieties from premature deubiquitination. Interestingly, when the same experiment was carried out using purified 26S proteasome, mono- and tetra-Ub globin were similarly degraded, highlighting the roles of the entire repertoire of cellular DUBs in regulating the degradation of proteasomal substrates.

4.
Proc Natl Acad Sci U S A ; 113(32): E4639-47, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27385826

ABSTRACT

The "canonical" proteasomal degradation signal is a substrate-anchored polyubiquitin chain. However, a handful of proteins were shown to be targeted following monoubiquitination. In this study, we established-in both human and yeast cells-a systematic approach for the identification of monoubiquitination-dependent proteasomal substrates. The cellular wild-type polymerizable ubiquitin was replaced with ubiquitin that cannot form chains. Using proteomic analysis, we screened for substrates that are nevertheless degraded under these conditions compared with those that are stabilized, and therefore require polyubiquitination for their degradation. For randomly sampled representative substrates, we confirmed that their cellular stability is in agreement with our screening prediction. Importantly, the two groups display unique features: monoubiquitinated substrates are smaller than the polyubiquitinated ones, are enriched in specific pathways, and, in humans, are structurally less disordered. We suggest that monoubiquitination-dependent degradation is more widespread than assumed previously, and plays key roles in various cellular processes.


Subject(s)
Proteasome Endopeptidase Complex/physiology , Proteins/metabolism , Ubiquitination , Humans , MCF-7 Cells , Proteasome Endopeptidase Complex/chemistry , Proteomics
5.
Biochem Biophys Res Commun ; 428(1): 11-6, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23036196

ABSTRACT

FAT10 is a ubiquitin-like protein made of two tandem, head-to-tail, ubiquitin domains. It is known to covalently modify proteins in a mechanism similar, though not identical, to that of other ubiquitin-like proteins. The lack of known physiological substrates covalently conjugated by the protein made it difficult to unravel its biological functions. Here we identify two proteins that are covalently modified by FAT10, the inflammatory mediator LRRFIP2 and the endoplasmic reticulum membrane protein LULL1. LRRFIP2 is involved in NF-κB activation following stimulation of TLR4. It is recruited along with MYD88 to the cytosolic tail of the receptor, and by that mediates activation of the downstream signaling cascade. We show that FATylation of LRRFIP2 occurs on two distinct sites, each being modified by a single FAT10 moiety. Furthermore, the second modification is regulated by the first one. Importantly, FATylation of LRRFIP2 interferes with its recruitment to the membrane by translocating it to the cellular insoluble fraction, thus inhibiting NF-κB activation.


Subject(s)
Carrier Proteins/metabolism , Inflammation/immunology , Lipopolysaccharides/immunology , Ubiquitination , Ubiquitins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Cytosol/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Inflammation/metabolism , Membrane Proteins , Molecular Sequence Data , Mutation , NF-kappa B/metabolism , Substrate Specificity , Toll-Like Receptor 4/immunology , Ubiquitins/genetics
6.
Mol Biol Cell ; 23(1): 225-32, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22072791

ABSTRACT

FAT10 is a ubiquitin-like protein modifier that is induced in vertebrates following certain inflammatory stimuli. Its functions and the repertoire of its target substrates have remained elusive. In contrast to ubiquitin, its cellular abundance is tightly controlled by both transcriptional and posttranslational regulation, and it was reported to be rapidly degraded by the proteasome. Here we provide data to indicate that the degradation of FAT10 requires ubiquitination: degradation was inhibited in cells expressing a ubiquitin mutant that cannot be polymerized and in a mutant cell harboring a thermolabile ubiquitin-activating enzyme, E1. Of importance, FAT10 can serve as a degradation signal for otherwise stable proteins, and in this case, too, the targeting to the proteasome requires ubiquitination. Degradation of FAT10 is accelerated after induction of apoptosis, suggesting that it plays a role in prosurvival pathways.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitins/metabolism , Animals , Apoptosis , CHO Cells , Cell Survival , Cell-Free System , Cricetinae , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Leupeptins/pharmacology , Proteasome Inhibitors , Protein Binding , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Ubiquitination , Ubiquitins/chemistry
7.
Int J Cancer ; 128(4): 817-25, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-20473904

ABSTRACT

We report that MDM2, a negative regulator of p53, can bind to EBNA-5. Using GST pull-down assay, immunoprecipitation, surface plasmon resonance and immunostaining of lymphoblastoid cells, we found that trimolecular complexes are formed between EBNA-5, MDM2 and p53, where MDM2 serves as a bridge. The EBNA-5 binding to MDM2 counteracted destabilizing effect of the latter on the p53. In ubiquitination and degradation assays in vitro, EBNA-5 inhibited p53 polyubiquitination (but not monoubiquitination) in a concentration-dependent manner. This resembles the effect of p14ARF on p53. Moreover, EBNA-5 was found to inhibit the degradation of p53 in vitro. High levels of p53 expression were maintained in LCLs. The binding of EBNA-5 to MDM2 also could impair the functional activity of p53. The p53-dependent genes P21 and VDR were not induced in EBV-infected, in contrast to mitogen-activated cells. This may explain the tolerance of established LCLs to high levels of p53 without undergoing apoptosis.


Subject(s)
B-Lymphocytes/metabolism , Breast Neoplasms/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Trans-Activators , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , B-Lymphocytes/pathology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cells, Cultured , Chromatin Immunoprecipitation , Epstein-Barr Virus Nuclear Antigens/genetics , Female , Humans , Protein Binding , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Surface Plasmon Resonance , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Ubiquitination
8.
Proc Natl Acad Sci U S A ; 105(41): 15690-5, 2008 Oct 14.
Article in English | MEDLINE | ID: mdl-18836078

ABSTRACT

A growing number of proteins, including the myogenic transcription factor MyoD, are targeted for proteasomal degradation after N-terminal ubiquitination (NTU) where the first ubiquitin moiety is conjugated to the N-terminal residue rather than to an internal lysine. NTU might be essential in targeting both lysine-containing and naturally occurring lysine-less proteins such as p16(INK4a) and p14(ARF); however, the mechanisms that underlie this process are largely unknown. Specifically, the recognition motif(s) in the target substrates and the ubiquitin ligase(s) that catalyze NTU are still obscure. Here we show that the N-terminal domain of MyoD is critical for its degradation and that its destabilizing effect depends on nuclear localization of the protein. Deletion of the first 15 aa of MyoD blocked completely its lysine-independent degradation. Importantly, transfer of the first 30 N-terminal residues of MyoD to GFP destabilized this otherwise stable protein, and, here too, targeting for degradation depended on localization of the protein to the nucleus. Deletion of the N-terminal domain of lysine-less MyoD did not abolish completely ubiquitination of the protein, suggesting that this domain may be required for targeting the protein also in a postubiquitination step. Interestingly, NTU is evolutionarily conserved: in the yeast Saccharomyces cerevisiae lysine-less (LL) MyoD is degraded in a ubiquitin-, N-terminal domain-, and nuclear localization-dependent manner. Taken together, our data suggest that a short N-terminal segment of MyoD is necessary and sufficient to render MyoD susceptible for ubiquitin- and nuclear-dependent degradation.


Subject(s)
Cell Nucleus/metabolism , MyoD Protein/metabolism , Ubiquitin/metabolism , Animals , Conserved Sequence , Lysine , Mice , MyoD Protein/chemistry , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...