Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Mol Immunol ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937625

ABSTRACT

CD28 and 4-1BB costimulatory endodomains included in chimeric antigen receptor (CAR) molecules play a critical role in promoting sustained antitumor activity of CAR-T cells. However, the molecular events associated with the ectopic and constitutive display of either CD28 or 4-1BB in CAR-T cells have been only partially explored. In the current study, we demonstrated that 4-1BB incorporated within the CAR leads to cell cluster formation and cell death in the forms of both apoptosis and necroptosis in the absence of CAR tonic signaling. Mechanistic studies illustrate that 4-1BB sequesters A20 to the cell membrane in a TRAF-dependent manner causing A20 functional deficiency that in turn leads to NF-κB hyperactivity, cell aggregation via ICAM-1 overexpression, and cell death including necroptosis via RIPK1/RIPK3/MLKL pathway. Genetic modulations obtained by either overexpressing A20 or releasing A20 from 4-1BB by deleting the TRAF-binding motifs of 4-1BB rescue cell cluster formation and cell death and enhance the antitumor ability of 4-1BB-costimulated CAR-T cells.

2.
bioRxiv ; 2024 May 28.
Article in English | MEDLINE | ID: mdl-38853827

ABSTRACT

The ubiquitin-like protein ISG15 (interferon-stimulated gene 15) regulates the host response to bacterial and viral infections through its conjugation to proteins (ISGylation) following interferon production. ISGylation is antagonized by the highly specific cysteine protease USP18, which is the major deISGylating enzyme. However, mechanisms underlying USP18's extraordinary specificity towards ISG15 remains elusive. Here, we show that USP18 interacts with its paralog USP41, whose catalytic domain shares 97% identity with USP18. However, USP41 does not act as a deISGylase, which led us to perform a comparative analysis to decipher the basis for this difference, revealing molecular determinants of USP18's specificity towards ISG15. We found that USP18 C-terminus, as well as a conserved Leucine at position 198, are essential for its enzymatic activity and likely act as functional surfaces based on AlphaFold predictions. Finally, we propose that USP41 antagonizes conjugation of the understudied ubiquitin-like protein FAT10 (HLA-F adjacent transcript 10) from substrates in a catalytic-independent manner. Altogether, our results offer new insights into USP18's specificity towards ISG15, while identifying USP41 as a negative regulator of FAT10 conjugation.

4.
bioRxiv ; 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37873169

ABSTRACT

Targeted protein degradation by the ubiquitin-proteasome system is an essential mechanism regulating cellular division. The kinase PLK1 coordinates protein degradation at the G2/M phase of the cell cycle by promoting the binding of substrates to the E3 ubiquitin ligase SCFßTrCP. However, the magnitude to which PLK1 shapes the mitotic proteome has not been characterized. Combining deep, quantitative proteomics with pharmacologic PLK1 inhibition (PLK1i), we identified more than 200 proteins whose abundances were increased by PLK1i at G2/M. We validate many new PLK1-regulated proteins, including several substrates of the cell cycle E3 SCFCyclin F, demonstrating that PLK1 promotes proteolysis through at least two distinct SCF-family E3 ligases. Further, we found that the protein kinase A anchoring protein AKAP2 is cell cycle regulated and that its mitotic degradation is dependent on the PLK1/ßTrCP-signaling axis. Interactome analysis revealed that the strongest interactors of AKAP2 function in signaling networks regulating proliferation, including MAPK, AKT, and Hippo. Altogether, our data demonstrate that PLK1 coordinates a widespread program of protein breakdown at G2/M. We propose that dynamic proteolytic changes mediated by PLK1 integrate proliferative signals with the core cell cycle machinery during cell division. This has potential implications in malignancies where PLK1 is aberrantly regulated.

5.
Nat Struct Mol Biol ; 30(11): 1663-1674, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37735619

ABSTRACT

Substrate polyubiquitination drives a myriad of cellular processes, including the cell cycle, apoptosis and immune responses. Polyubiquitination is highly dynamic, and obtaining mechanistic insight has thus far required artificially trapped structures to stabilize specific steps along the enzymatic process. So far, how any ubiquitin ligase builds a proteasomal degradation signal, which is canonically regarded as four or more ubiquitins, remains unclear. Here we present time-resolved cryogenic electron microscopy studies of the 1.2 MDa E3 ubiquitin ligase, known as the anaphase-promoting complex/cyclosome (APC/C), and its E2 co-enzymes (UBE2C/UBCH10 and UBE2S) during substrate polyubiquitination. Using cryoDRGN (Deep Reconstructing Generative Networks), a neural network-based approach, we reconstruct the conformational changes undergone by the human APC/C during polyubiquitination, directly visualize an active E3-E2 pair modifying its substrate, and identify unexpected interactions between multiple ubiquitins with parts of the APC/C machinery, including its coactivator CDH1. Together, we demonstrate how modification of substrates with nascent ubiquitin chains helps to potentiate processive substrate polyubiquitination, allowing us to model how a ubiquitin ligase builds a proteasomal degradation signal.


Subject(s)
Anaphase , Ubiquitin , Humans , Anaphase-Promoting Complex-Cyclosome/chemistry , Cryoelectron Microscopy , Ubiquitination , Ubiquitin/metabolism , Cell Cycle Proteins/metabolism
6.
EMBO J ; 41(3): e108823, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34942047

ABSTRACT

Polyubiquitination by E2 and E3 enzymes is crucial to cell cycle control, epigenetic regulation, and development. The hallmark of the E2 family is the ubiquitin (Ub)-conjugating (UBC) domain that forms a dynamic thioester conjugate with ubiquitin (E2~Ub). Numerous studies have focused on E2 surfaces, such as the N-terminal and crossover helices, that directly interact with an E3 or the conjugated ubiquitin to stabilize the active, "closed" state of the E2~Ub. However, it remains unclear how other E2 surfaces regulate ubiquitin transfer. Here, we demonstrate the helix-turn-helix (HTH) motif of the UBC tunes the intrinsic polyubiquitination activity through distinct functions in different E2s. Interestingly, the E2HTH motif is repurposed in UBE2S and UBE2R2 to interact with the conjugated or acceptor ubiquitin, respectively, modulating ubiquitin transfer. Furthermore, we propose that Anaphase-Promoting Complex/Cyclosome binding to the UBE2SHTH reduces the conformational space of the flexible E2~Ub, demonstrating an atypical E3-dependent activation mechanism. Altogether, we postulate the E2HTH motif evolved to provide new functionalities that can be harnessed by E3s and permits additional regulation to facilitate specific E2-E3-mediated polyubiquitination.


Subject(s)
Ubiquitin-Conjugating Enzymes/chemistry , Amino Acid Motifs , Catalytic Domain , Humans , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
7.
PLoS Biol ; 18(12): e3000975, 2020 12.
Article in English | MEDLINE | ID: mdl-33306668

ABSTRACT

The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase and critical regulator of cell cycle progression. Despite its vital role, it has remained challenging to globally map APC/C substrates. By combining orthogonal features of known substrates, we predicted APC/C substrates in silico. This analysis identified many known substrates and suggested numerous candidates. Unexpectedly, chromatin regulatory proteins are enriched among putative substrates, and we show experimentally that several chromatin proteins bind APC/C, oscillate during the cell cycle, and are degraded following APC/C activation, consistent with being direct APC/C substrates. Additional analysis revealed detailed mechanisms of ubiquitylation for UHRF1, a key chromatin regulator involved in histone ubiquitylation and DNA methylation maintenance. Disrupting UHRF1 degradation at mitotic exit accelerates G1-phase cell cycle progression and perturbs global DNA methylation patterning in the genome. We conclude that APC/C coordinates crosstalk between cell cycle and chromatin regulatory proteins. This has potential consequences in normal cell physiology, where the chromatin environment changes depending on proliferative state, as well as in disease.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Chromatin/metabolism , Ubiquitin-Protein Ligases/metabolism , Anaphase-Promoting Complex-Cyclosome/physiology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/physiology , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cell Line , Chromatin/genetics , Computer Simulation , HEK293 Cells , HeLa Cells , Humans , Protein Processing, Post-Translational , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitination
8.
Nat Struct Mol Biol ; 27(6): 550-560, 2020 06.
Article in English | MEDLINE | ID: mdl-32393902

ABSTRACT

The interplay between E2 and E3 enzymes regulates the polyubiquitination of substrates in eukaryotes. Among the several RING-domain E3 ligases in humans, many utilize two distinct E2s for polyubiquitination. For example, the cell cycle regulatory E3, human anaphase-promoting complex/cyclosome (APC/C), relies on UBE2C to prime substrates with ubiquitin (Ub) and on UBE2S to extend polyubiquitin chains. However, the potential coordination between these steps in ubiquitin chain formation remains undefined. While numerous studies have unveiled how RING E3s stimulate individual E2s for Ub transfer, here we change perspective to describe a case where the chain-elongating E2 UBE2S feeds back and directly stimulates the E3 APC/C to promote substrate priming and subsequent multiubiquitination by UBE2C. Our work reveals an unexpected model for the mechanisms of RING E3-dependent ubiquitination and for the diverse and complex interrelationship between components of the ubiquitination cascade.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Anaphase-Promoting Complex-Cyclosome/chemistry , Anaphase-Promoting Complex-Cyclosome/genetics , Apc4 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc4 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc4 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Cytidine Triphosphate/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , HeLa Cells , Humans , Polyubiquitin/metabolism , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
9.
Semin Cancer Biol ; 67(Pt 2): 145-158, 2020 12.
Article in English | MEDLINE | ID: mdl-32201366

ABSTRACT

Since its discovery forty years ago, protein ubiquitination has been an ever-expanding field. Virtually all biological processes are controlled by the post-translational conjugation of ubiquitin onto target proteins. In addition, since ubiquitin controls substrate degradation through the action of hundreds of enzymes, many of which represent attractive therapeutic candidates, harnessing the ubiquitin system to reshape proteomes holds great promise for improving disease outcomes. Among the numerous physiological functions controlled by ubiquitin, the cell cycle is among the most critical. Indeed, the discovery that the key drivers of cell cycle progression are regulated by the ubiquitin-proteasome system (UPS) epitomizes the connection between ubiquitin signaling and proliferation. Since cancer is a disease of uncontrolled cell cycle progression and proliferation, targeting the UPS to stop cancer cells from cycling and proliferating holds enormous therapeutic potential. Ubiquitination is reversible, and ubiquitin is removed from substrates by catalytic proteases termed deubiquitinases or DUBs. While ubiquitination is tightly linked to proliferation and cancer, the role of DUBs represents a layer of complexity in this landscape that remains poorly captured. Due to their ability to remodel the proteome by altering protein degradation dynamics, DUBs play an important and underappreciated role in the cell cycle and proliferation of both normal and cancer cells. Moreover, due to their enzymatic protease activity and an open ubiquitin binding pocket, DUBs are likely to be important in the future of cancer treatment, since they are among the most druggable enzymes in the UPS. In this review we summarize new and important findings linking DUBs to cell cycle and proliferation, as well as to the etiology and treatment of cancer. We also highlight new advances in developing pharmacological approaches to attack DUBs for therapeutic benefit.


Subject(s)
Deubiquitinating Enzymes/metabolism , Neoplasms/pathology , Cell Cycle , Deubiquitinating Enzyme CYLD/genetics , Deubiquitinating Enzyme CYLD/metabolism , Deubiquitinating Enzymes/genetics , Gene Expression Regulation, Neoplastic , Humans , Mitosis , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitin-Specific Peptidase 7/metabolism
10.
Cell Rep ; 26(11): 3076-3086.e6, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30865895

ABSTRACT

The transcription factor FOXM1 contributes to cell cycle progression and is significantly upregulated in basal-like breast cancer (BLBC). Despite its importance in normal and cancer cell cycles, we lack a complete understanding of mechanisms that regulate FOXM1. We identified USP21 in an RNAi-based screen for deubiquitinases that control FOXM1 abundance. USP21 increases the stability of FOXM1, and USP21 binds and deubiquitinates FOXM1 in vivo and in vitro, indicating a direct enzyme-substrate relationship. Depleting USP21 downregulates the FOXM1 transcriptional network and causes a significant delay in cell cycle progression. Significantly, USP21 depletion sensitized BLBC cell lines and mouse xenograft tumors to paclitaxel, an anti-mitotic, frontline therapy in BLBC treatment. USP21 is the most frequently amplified deubiquitinase in BLBC patient tumors, and its amplification co-occurs with the upregulation of FOXM1 protein. Altogether, these data suggest a role for USP21 in the proliferation and potentially treatment of FOXM1-high, USP21-high BLBC.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Cell Cycle , Drug Resistance, Neoplasm , Forkhead Box Protein M1/metabolism , Mammary Neoplasms, Experimental/metabolism , Paclitaxel/therapeutic use , Ubiquitination , Animals , Female , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Ubiquitin Thiolesterase/metabolism
11.
Cell Cycle ; 18(6-7): 652-660, 2019.
Article in English | MEDLINE | ID: mdl-30874463

ABSTRACT

The Anaphase-Promoting Complex/Cyclosome (APC/C) is an E3 ubiquitin ligase and a key regulator of cell cycle progression. By triggering the degradation of mitotic cyclins, APC/C controls cell cycle-dependent oscillations in cyclin-dependent kinase (CDK) activity. Thus, the dynamic activities of both APC/C and CDK sit at the core of the cell cycle oscillator. The APC/C controls a large number of substrates and is regulated through multiple mechanisms, including cofactor-dependent activation. These cofactors, Cdc20 and Cdh1, recognize substrates, while the specific E2 enzymes UBE2C/UbcH10 and UBE2S cooperate with APC/C to build K11-linked ubiquitin chains on substrates to target them for proteasomal degradation. However, whether deubiquitinating enzymes (DUBs) can antagonize APC/C substrate ubiquitination during mitosis has remained largely unknown. We recently demonstrated that Cezanne/OTUD7B is a cell cycle-regulated DUB that opposes the ubiquitination of APC/C substrates. Cezanne binds APC/C substrates, reverses their ubiquitination and protects them from degradation. Accordingly, Cezanne depletion accelerates APC/C substrate degradation, leading to errors in mitotic progression and formation of micronuclei. Moreover, Cezanne is significantly amplified and overexpressed in breast cancers. This suggests a potential role for APC/C antagonism in the pathogenesis of disease. APC/C contributes to chromosome segregation fidelity in mitosis raising the possibility that copy-number and expression changes in Cezanne observed in cancer contribute to the etiology of disease. Collectively, these observations identify a new player in cell cycle progression, define mechanisms of tempered APC/C substrate destruction and highlight the importance of this regulation in maintaining chromosome stability.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Endopeptidases/metabolism , Mitosis/physiology , Ubiquitin/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Nucleus Division/physiology , Chromosomal Instability/physiology , Chromosome Segregation/physiology , Humans , Ubiquitination/physiology
12.
Biochim Biophys Acta Mol Cell Res ; 1865(12): 1924-1933, 2018 12.
Article in English | MEDLINE | ID: mdl-30290241

ABSTRACT

The cell cycle is principally controlled by Cyclin Dependent Kinases (CDKs), whose oscillating activities are determined by binding to Cyclin coactivators. Cyclins exhibit dynamic changes in abundance as cells pass through the cell cycle. The sequential, timed accumulation and degradation of Cyclins, as well as many other proteins, imposes order on the cell cycle and contributes to genome maintenance. The destruction of many cell cycle regulated proteins, including Cyclins A and B, is controlled by a large, multi-subunit E3 ubiquitin ligase termed the Anaphase Promoting Complex/Cyclosome (APC/C). APC/C activity is tightly regulated during the cell cycle. Its activation state increases dramatically in mid-mitosis and it remains active until the end of G1 phase. Following its mandatory inactivation at the G1/S boundary, APC/C activity remains low until the subsequent mitosis. Due to its role in guarding against the inappropriate or untimely accumulation of Cyclins, the APC/C is a core component of the cell cycle oscillator. In addition to the regulation of Cyclins, APC/C controls the degradation of many other substrates. Therefore, it is vital that the activity of APC/C itself be tightly guarded. The APC/C is most well studied for its role and regulation during mitosis. However, the APC/C also plays a similarly important and conserved role in the maintenance of G1 phase. Here we review the diverse mechanisms counteracting APC/C activity throughout the cell cycle and the importance of their coordinated actions on cell growth, proliferation, and disease.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Animals , Cell Cycle , Cell Proliferation , Gene Expression Regulation , Humans
13.
EMBO J ; 37(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29973362

ABSTRACT

The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase and key regulator of cell cycle progression. Since APC/C promotes the degradation of mitotic cyclins, it controls cell cycle-dependent oscillations in cyclin-dependent kinase (CDK) activity. Both CDKs and APC/C control a large number of substrates and are regulated by analogous mechanisms, including cofactor-dependent activation. However, whereas substrate dephosphorylation is known to counteract CDK, it remains largely unknown whether deubiquitinating enzymes (DUBs) antagonize APC/C substrate ubiquitination during mitosis. Here, we demonstrate that Cezanne/OTUD7B is a cell cycle-regulated DUB that opposes the ubiquitination of APC/C targets. Cezanne is remarkably specific for K11-linked ubiquitin chains, which are formed by APC/C in mitosis. Accordingly, Cezanne binds established APC/C substrates and reverses their APC/C-mediated ubiquitination. Cezanne depletion accelerates APC/C substrate degradation and causes errors in mitotic progression and formation of micronuclei. These data highlight the importance of tempered APC/C substrate destruction in maintaining chromosome stability. Furthermore, Cezanne is recurrently amplified and overexpressed in numerous malignancies, suggesting a potential role in genome maintenance and cancer cell proliferation.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Chromosomal Instability , Deubiquitinating Enzymes/metabolism , Endopeptidases/metabolism , Mitosis , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Proteolysis , Anaphase-Promoting Complex-Cyclosome/genetics , Deubiquitinating Enzymes/genetics , Endopeptidases/genetics , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Micronuclei, Chromosome-Defective , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Ubiquitination
14.
Mol Biol Cell ; 29(2): 96-110, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29167380

ABSTRACT

Spinal muscular atrophy (SMA) is caused by homozygous mutations in human SMN1 Expression of a duplicate gene (SMN2) primarily results in skipping of exon 7 and production of an unstable protein isoform, SMNΔ7. Although SMN2 exon skipping is the principal contributor to SMA severity, mechanisms governing stability of survival motor neuron (SMN) isoforms are poorly understood. We used a Drosophila model system and label-free proteomics to identify the SCFSlmb ubiquitin E3 ligase complex as a novel SMN binding partner. SCFSlmb interacts with a phosphor degron embedded within the human and fruitfly SMN YG-box oligomerization domains. Substitution of a conserved serine (S270A) interferes with SCFSlmb binding and stabilizes SMNΔ7. SMA-causing missense mutations that block multimerization of full-length SMN are also stabilized in the degron mutant background. Overexpression of SMNΔ7S270A, but not wild-type (WT) SMNΔ7, provides a protective effect in SMA model mice and human motor neuron cell culture systems. Our findings support a model wherein the degron is exposed when SMN is monomeric and sequestered when SMN forms higher-order multimers.


Subject(s)
Drosophila Proteins/genetics , Muscular Atrophy, Spinal/genetics , RNA-Binding Proteins/genetics , Survival of Motor Neuron 1 Protein/genetics , Animals , Cells, Cultured , Disease Models, Animal , Drosophila , Homozygote , Humans , Mice , Motor Neurons/metabolism , Mutation, Missense , Nerve Tissue Proteins/metabolism , Polymerization
15.
Cell Rep ; 20(13): 3212-3222, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28954236

ABSTRACT

The oncogenic AKT kinase is a key regulator of apoptosis, cell growth, and cell-cycle progression. Despite its important role in proliferation, it remains largely unknown how AKT is mechanistically linked to the cell cycle. We show here that cyclin F, a substrate receptor F-box protein for the SCF (Skp1/Cul1/F-box) family of E3 ubiquitin ligases, is a bona fide AKT substrate. Cyclin F expression oscillates throughout the cell cycle, a rare feature among the 69 human F-box proteins, and all of its known substrates are involved in proliferation. AKT phosphorylation of cyclin F enhances its stability and promotes assembly into productive E3 ligase complexes. Importantly, expression of mutant versions of cyclin F that cannot be phosphorylated by AKT impair cell-cycle entry. Our data suggest that cyclin F transmits mitogen signaling through AKT to the core cell-cycle machinery. This discovery has potential implications for proliferative control in malignancies where AKT is activated.


Subject(s)
Cell Cycle/physiology , Cyclins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Signal Transduction
16.
PLoS One ; 12(1): e0169988, 2017.
Article in English | MEDLINE | ID: mdl-28099510

ABSTRACT

NUB1 (Nedd8 ultimate buster 1) is an adaptor protein which negatively regulates the ubiquitin-like protein Nedd8 as well as neddylated proteins levels through proteasomal degradation. However, molecular mechanisms underlying this function are not completely understood. Here, we report that the oncogenic E3 ubiquitin ligase Mdm2 is a new NUB1 interacting protein which induces its ubiquitination. Interestingly, we found that Mdm2-mediated ubiquitination of NUB1 is not a proteolytic signal. Instead of promoting the conjugation of polyubiquitin chains and the subsequent proteasomal degradation of NUB1, Mdm2 rather induces its di-ubiquitination on lysine 159. Importantly, mutation of lysine 159 into arginine inhibits NUB1 activity by impairing its negative regulation of Nedd8 and of neddylated proteins. We conclude that Mdm2 acts as a positive regulator of NUB1 function, by modulating NUB1 ubiquitination on lysine 159.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Lysine/metabolism , NEDD8 Protein , Proto-Oncogene Proteins c-mdm2/genetics , Transcription Factors/genetics , Ubiquitination , Ubiquitins/genetics , Ubiquitins/metabolism
17.
Sci Rep ; 7: 39732, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28054562

ABSTRACT

Intrinsically disordered proteins (IDPs) are prevalent in eukaryotes, performing signaling and regulatory functions. Often associated with human diseases, they constitute drug-development targets. NUPR1 is a multifunctional IDP, over-expressed and involved in pancreatic ductal adenocarcinoma (PDAC) development. By screening 1120 FDA-approved compounds, fifteen candidates were selected, and their interactions with NUPR1 were characterized by experimental and simulation techniques. The protein remained disordered upon binding to all fifteen candidates. These compounds were tested in PDAC-derived cell-based assays, and all induced cell-growth arrest and senescence, reduced cell migration, and decreased chemoresistance, mimicking NUPR1-deficiency. The most effective compound completely arrested tumor development in vivo on xenografted PDAC-derived cells in mice. Besides reporting the discovery of a compound targeting an intact IDP and specifically active against PDAC, our study proves the possibility to target the 'fuzzy' interface of a protein that remains disordered upon binding to its natural biological partners or to selected drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Ductal/metabolism , Intrinsically Disordered Proteins/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Trifluoperazine/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Ductal/drug therapy , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cellular Senescence , Drug Discovery , Drug Resistance, Neoplasm , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Neoplasm Proteins/genetics , Pancreatic Neoplasms/drug therapy , Protein Binding , Trifluoperazine/chemistry , Trifluoperazine/pharmacology , Trifluoperazine/therapeutic use , Xenograft Model Antitumor Assays
18.
Cell Rep ; 16(12): 3359-3372, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27653696

ABSTRACT

The anaphase promoting complex/cyclosome (APC/C) is an ubiquitin ligase and core component of the cell-cycle oscillator. During G1 phase, APC/C binds to its substrate receptor Cdh1 and APC/C(Cdh1) plays an important role in restricting S-phase entry and maintaining genome integrity. We describe a reciprocal feedback circuit between APC/C and a second ubiquitin ligase, the SCF (Skp1-Cul1-F box). We show that cyclin F, a cell-cycle-regulated substrate receptor (F-box protein) for the SCF, is targeted for degradation by APC/C. Furthermore, we establish that Cdh1 is itself a substrate of SCF(cyclin F). Cyclin F loss impairs Cdh1 degradation and delays S-phase entry, and this delay is reversed by simultaneous removal of Cdh1. These data indicate that the coordinated, temporal ordering of cyclin F and Cdh1 degradation, organized in a double-negative feedback loop, represents a fundamental aspect of cell-cycle control. This mutual antagonism could be a feature of other oscillating systems.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cyclins/metabolism , Feedback, Physiological/physiology , S Phase/physiology , HEK293 Cells , HeLa Cells , Humans
19.
Article in English | MEDLINE | ID: mdl-25774357

ABSTRACT

Response of immune and non-immune cells to pathogens infections is a very dynamic process. It involves the activation/modulation of many pathways leading to actin remodeling, membrane engulfing, phagocytosis, vesicle trafficking, phagolysosome formation, aiming at the destruction of the intruder. These sophisticated and rapid mechanisms rely on post-translational modifications (PTMs) of key host cells' factors, and bacteria have developed various strategies to manipulate them to favor their survival. Among these important PTMs, ubiquitination has emerged as a major mediator/modulator/regulator of host cells response to infections that pathogens have also learned to use for their own benefit. In this mini-review, we summarize our current knowledge about the normal functions of ubiquitination during host cell infection, and we detail its hijacking by model pathogens to escape clearance and to proliferate.


Subject(s)
Bacterial Infections/metabolism , Host-Pathogen Interactions , Ubiquitination , Animals , Bacteria/genetics , Bacterial Infections/microbiology , Bacterial Physiological Phenomena , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans
20.
J Proteome Res ; 13(5): 2478-94, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24654937

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive malignancy characterized by an excessive resistance to all known anticancer therapies, a still largely elusive phenomenon. To identify original mechanisms, we have explored the role of post-translational modifications (PTMs) mediated by members of the ubiquitin family. Although alterations of these pathways have been reported in different cancers, no methodical search for these kinds of anomalies has been performed so far. Therefore, we studied the ubiquitin-, Nedd8-, and SUMO1-specific proteomes of a pancreatic cancer cell line (MiaPaCa-2) and identified changes induced by gemcitabine, the standard PDAC's chemotherapeutic drug. These PTMs profiles contained both known major substrates of all three modifiers as well as original ones. Gemcitabine treatment altered the PTM profile of proteins involved in various biological functions, some known cancer associated genes, many potentially cancer-associated genes, and several cancer-signaling networks, including canonical and noncanonical WNT and PI3K/Akt/MTOR pathways. Some of these altered PTMs formed groups of functionally and physically associated proteins. Importantly, we could validate the gemcitabine-induced PTMs variations of relevant candidates and we could demonstrate the biological significance of such altered PTMs by studying in detail the sumoylation of SNIP1, one of these new targets.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Processing, Post-Translational/drug effects , Proteins/metabolism , Signal Transduction/drug effects , Ubiquitin/metabolism , Blotting, Western , Cell Line, Tumor , Chromatography, Liquid , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Microscopy, Fluorescence , NEDD8 Protein , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proteome/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Proteins , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Tandem Mass Spectrometry , Ubiquitin/genetics , Ubiquitins/genetics , Ubiquitins/metabolism , Wnt1 Protein/metabolism , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...