Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 143
Filtrer
1.
Biomed Pharmacother ; 179: 117356, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39214012

RÉSUMÉ

Protein neddylation, a type of post-translational modifications, involves the transfer of the ubiquitin-like protein NEDD8 to the lysine residues of a target substrate, which is catalyzed by the NEDD8 activating enzyme (E1), NEDD8 conjugating enzyme (E2), and NEDD8 ligase (E3). Cullin family proteins, core components of Cullin-RING E3 ubiquitin ligases (CRLs), are the most well-known physiological substrates of neddylation. CRLs, activated upon cullin neddylation, promote the ubiquitination of a variety of key signaling proteins for proteasome degradation, thereby regulating many critical biological functions. Abnormal activation of neddylation enzymes as well as CRLs has been frequently observed in various human cancers and is associated with poor prognosis for cancer patients. Consequently, targeting neddylation has emerged as a promising strategy for the development of novel anticancer therapeutics. This review first briefly introduces the properties of protein neddylation and its role in cancer, and then systematically summarizes all reported chemical inhibitors of the three neddylation enzymes, providing a focused, up to date, and comprehensive resource in the discovery and development of these small molecule inhibitors.


Sujet(s)
Antinéoplasiques , Protéine NEDD8 , Tumeurs , Humains , Protéine NEDD8/métabolisme , Protéine NEDD8/antagonistes et inhibiteurs , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Tumeurs/traitement médicamenteux , Tumeurs/enzymologie , Tumeurs/métabolisme , Animaux , Ubiquitin-activating enzymes/antagonistes et inhibiteurs , Ubiquitin-activating enzymes/métabolisme , Découverte de médicament , Maturation post-traductionnelle des protéines/effets des médicaments et des substances chimiques , Ubiquitin-conjugating enzymes/métabolisme , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Antienzymes/pharmacologie , Antienzymes/usage thérapeutique , Bibliothèques de petites molécules/pharmacologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/antagonistes et inhibiteurs
2.
Pain ; 165(3): 573-588, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-37751532

RÉSUMÉ

ABSTRACT: Dysregulation of voltage-gated sodium Na V 1.7 channels in sensory neurons contributes to chronic pain conditions, including trigeminal neuropathic pain. We previously reported that chronic pain results in part from increased SUMOylation of collapsin response mediator protein 2 (CRMP2), leading to an increased CRMP2/Na V 1.7 interaction and increased functional activity of Na V 1.7. Targeting this feed-forward regulation, we developed compound 194 , which inhibits CRMP2 SUMOylation mediated by the SUMO-conjugating enzyme Ubc9. We further demonstrated that 194 effectively reduces the functional activity of Na V 1.7 channels in dorsal root ganglia neurons and alleviated inflammatory and neuropathic pain. Here, we used a comprehensive array of approaches, encompassing biochemical, pharmacological, genetic, electrophysiological, and behavioral analyses, to assess the functional implications of Na V 1.7 regulation by CRMP2 in trigeminal ganglia (TG) neurons. We confirmed the expression of Scn9a , Dpysl2 , and UBE2I within TG neurons. Furthermore, we found an interaction between CRMP2 and Na V 1.7, with CRMP2 being SUMOylated in these sensory ganglia. Disrupting CRMP2 SUMOylation with compound 194 uncoupled the CRMP2/Na V 1.7 interaction, impeded Na V 1.7 diffusion on the plasma membrane, and subsequently diminished Na V 1.7 activity. Compound 194 also led to a reduction in TG neuron excitability. Finally, when intranasally administered to rats with chronic constriction injury of the infraorbital nerve, 194 significantly decreased nociceptive behaviors. Collectively, our findings underscore the critical role of CRMP2 in regulating Na V 1.7 within TG neurons, emphasizing the importance of this indirect modulation in trigeminal neuropathic pain.


Sujet(s)
Douleur chronique , Protéines et peptides de signalisation intercellulaire , Protéines de tissu nerveux , Névralgie essentielle du trijumeau , Ubiquitin-conjugating enzymes , Animaux , Rats , Douleur chronique/traitement médicamenteux , Douleur chronique/métabolisme , Ganglions sensitifs des nerfs spinaux , Rat Sprague-Dawley , Cellules réceptrices sensorielles/métabolisme , Névralgie essentielle du trijumeau/traitement médicamenteux , Névralgie essentielle du trijumeau/métabolisme , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Administration par voie nasale , Protéines de tissu nerveux/antagonistes et inhibiteurs
3.
Acta Pharmacol Sin ; 44(3): 661-669, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36138144

RÉSUMÉ

Neddylation is a type of posttranslational protein modification that has been observed to be overactivated in various cancers. UBC12 is one of two key E2 enzymes in the neddylation pathway. Reports indicate that UBC12 deficiency may suppress lung cancer cells, such that UBC12 could play an important role in tumor progression. However, systematic studies regarding the expression profile of UBC12 in cancers and its relationship to cancer prognosis are lacking. In this study, we comprehensively analyzed UBC12 expression in diverse cancer types and found that UBC12 is markedly overexpressed in most cancers (17/21), a symptom that negatively correlates with the survival rates of cancer patients, including gastric cancer. These results demonstrate the suitability of UBC12 as a potential target for cancer treatment. Currently, no effective inhibitor targeting UBC12 has been discovered. We screened a natural product library and found, for the first time, that arctigenin has been shown to significantly inhibit UBC12 enzyme activity and cullin neddylation. The inhibition of UBC12 enzyme activity was newly found to contribute to the effects of arctigenin on suppressing the malignant phenotypes of cancer cells. Furthermore, we performed proteomics analysis and found that arctigenin intervened with cullin downstream signaling pathways and substrates, such as the tumor suppressor PDCD4. In summary, these results demonstrate the importance of UBC12 as a potential therapeutic target for cancer treatment, and, for the first time, the suitability of arctigenin as a potential compound targeting UBC12 enzyme activity. Thus, these findings provide a new strategy for inhibiting neddylation-overactivated cancers.


Sujet(s)
Cullines , Tumeurs du poumon , Ubiquitin-conjugating enzymes , Humains , Protéines régulatrices de l'apoptose/métabolisme , Cullines/effets des médicaments et des substances chimiques , Furanes/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Protéine NEDD8/métabolisme , Protéines de liaison à l'ARN , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitin-conjugating enzymes/effets des médicaments et des substances chimiques
4.
Biochem Cell Biol ; 100(4): 309-324, 2022 08 01.
Article de Anglais | MEDLINE | ID: mdl-35544948

RÉSUMÉ

Liver fibrosis is a very common health problem and currently lacks effective treatments. Cullin RING E3 ligases (CRLs) regulate the turnover of ∼20% of mammalian cell proteins. Neddylation, the process by which NEDD8 is covalently attached to cullin proteins through sequential enzymatic reactions, is critical for the activation of CRLs and was recently found to be elevated in liver fibrosis. NEDD8-activating enzyme E1-specific inhibition led to the reduced liver damage characterized by decreased apoptosis, inflammation, and fibrosis. However, the relevance of a co-E3 ligase, DCN1, in liver fibrosis remains unclear. Here, a novel and potent DCN1-UBC12 interaction inhibitor HZX-960 was discovered with an IC50 value of 9.37 nmol/L, which could inhibit the neddylation of cullin3. Importantly, we identified that HZX-960 treatment could attenuate transforming growth factor ß-induced liver fibrotic responses by reducing the deposition of collagen I and α-smooth muscle actin, and upregulating cellular NF-E2-related factor 2, hemeoxygenase 1, and NADPH quinone oxidoreductase-1 levels in two hepatic stellate cell lines. Additionally, DCN1 was shown to be unregulated in CCl4-induced mice liver tissue, and liver fibrotic signaling in mice was reduced by HZX-960. Therefore, our data demonstrated that HZX-960 possessed anti-liver fibrosis ability and that DCN1 may be a potential therapeutic target for liver fibrosis treatment.


Sujet(s)
Antienzymes , Cirrhose du foie , Ubiquitin-conjugating enzymes , Ubiquitin-protein ligases , Animaux , Cullines/métabolisme , Antienzymes/pharmacologie , Cirrhose du foie/traitement médicamenteux , Souris , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitin-protein ligases/antagonistes et inhibiteurs , Ubiquitination
5.
Sci Transl Med ; 14(635): eabb7695, 2022 03 09.
Article de Anglais | MEDLINE | ID: mdl-35263148

RÉSUMÉ

Dysregulation of innate immune signaling pathways is implicated in various hematologic malignancies. However, these pathways have not been systematically examined in acute myeloid leukemia (AML). We report that AML hematopoietic stem and progenitor cells (HSPCs) exhibit a high frequency of dysregulated innate immune-related and inflammatory pathways, referred to as oncogenic immune signaling states. Through gene expression analyses and functional studies in human AML cell lines and patient-derived samples, we found that the ubiquitin-conjugating enzyme UBE2N is required for leukemic cell function in vitro and in vivo by maintaining oncogenic immune signaling states. It is known that the enzyme function of UBE2N can be inhibited by interfering with thioester formation between ubiquitin and the active site. We performed in silico structure-based and cellular-based screens and identified two related small-molecule inhibitors UC-764864/65 that targeted UBE2N at its active site. Using these small-molecule inhibitors as chemical probes, we further revealed the therapeutic efficacy of interfering with UBE2N function. This resulted in the blocking of ubiquitination of innate immune- and inflammatory-related substrates in human AML cell lines. Inhibition of UBE2N function disrupted oncogenic immune signaling by promoting cell death of leukemic HSPCs while sparing normal HSPCs in vitro. Moreover, baseline oncogenic immune signaling states in leukemic cells derived from discrete subsets of patients with AML exhibited a selective dependency on UBE2N function in vitro and in vivo. Our study reveals that interfering with UBE2N abrogates leukemic HSPC function and underscores the dependency of AML cells on UBE2N-dependent oncogenic immune signaling states.


Sujet(s)
Leucémie aigüe myéloïde , Ubiquitin-conjugating enzymes , Prolifération cellulaire/génétique , Humains , Leucémie aigüe myéloïde/métabolisme , Oncogènes , Transduction du signal/génétique , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitin-conjugating enzymes/génétique , Ubiquitin-conjugating enzymes/métabolisme
6.
J Mol Biol ; 434(8): 167524, 2022 04 30.
Article de Anglais | MEDLINE | ID: mdl-35248542

RÉSUMÉ

A general approach for the rapid and selective inhibition of enzymes in cells using a common tool compound would be of great value for research and therapeutic development. We previously reported a chemogenetic strategy that addresses this challenge for kinases, relying on bioorthogonal tethering of a pan inhibitor to a target kinase through a genetically encoded non-canonical amino acid. However, pan inhibitors are not available for many enzyme classes. Here, we expand the scope of the chemogenetic strategy to cysteine-dependent enzymes by bioorthogonal tethering of electrophilic warheads. For proof of concept, selective inhibition of two E2 ubiquitin-conjugating enzymes, UBE2L3 and UBE2D1, was demonstrated in biochemical assays. Further development and optimization of this approach should enable its use in cells as well as with other cysteine-dependent enzymes, facilitating the investigation of their cellular function and validation as therapeutic targets.


Sujet(s)
Cystéine , Ubiquitin-conjugating enzymes , Cystéine/composition chimique , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitin-conjugating enzymes/génétique
7.
SLAS Discov ; 27(4): 266-271, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35342035

RÉSUMÉ

UBCH10 is an ubiquitin-conjugating enzyme (E2) of the anaphase-promoting complex E3 ligase, a key regulator of the cell cycle. The UBCH10 gene and protein are frequently upregulated in multiple solid tumors, associated with an unfavorable outcome. Accumulating evidence from studies of human cancer cell lines, mouse transgenic models, and analyses of clinical samples suggest that UBCH10 is a potential cancer drug target. No small molecule inhibitor of UBCH10 has been reported in the literature. Here, we described the development and optimization of a novel time-resolved fluorescence resonance energy transfer (TR-FRET) UBCH10 assay based on the self-polyubiquitination of the enzyme in the absence of E3. The homogenous assay is robust, sensitive, and scalable to different multi-well formats for high-throughput screening (HTS). We demonstrate the suitability of the TR-FRET assay to identify chemical inhibitors of UBCH10 in a pilot HTS campaign.


Sujet(s)
Tests de criblage à haut débit , Ubiquitin-conjugating enzymes , Animaux , Humains , Souris , Tumeurs , Ubiquitin-conjugating enzymes/analyse , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitin-protein ligases , Ubiquitination
8.
J Hepatol ; 76(1): 11-24, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34555423

RÉSUMÉ

BACKGROUND & AIMS: Autophagy-related gene 3 (ATG3) is an enzyme mainly known for its actions in the LC3 lipidation process, which is essential for autophagy. Whether ATG3 plays a role in lipid metabolism or contributes to non-alcoholic fatty liver disease (NAFLD) remains unknown. METHODS: By performing proteomic analysis on livers from mice with genetic manipulation of hepatic p63, a regulator of fatty acid metabolism, we identified ATG3 as a new target downstream of p63. ATG3 was evaluated in liver samples from patients with NAFLD. Further, genetic manipulation of ATG3 was performed in human hepatocyte cell lines, primary hepatocytes and in the livers of mice. RESULTS: ATG3 expression is induced in the liver of animal models and patients with NAFLD (both steatosis and non-alcoholic steatohepatitis) compared with those without liver disease. Moreover, genetic knockdown of ATG3 in mice and human hepatocytes ameliorates p63- and diet-induced steatosis, while its overexpression increases the lipid load in hepatocytes. The inhibition of hepatic ATG3 improves fatty acid metabolism by reducing c-Jun N-terminal protein kinase 1 (JNK1), which increases sirtuin 1 (SIRT1), carnitine palmitoyltransferase 1a (CPT1a), and mitochondrial function. Hepatic knockdown of SIRT1 and CPT1a blunts the effects of ATG3 on mitochondrial activity. Unexpectedly, these effects are independent of an autophagic action. CONCLUSIONS: Collectively, these findings indicate that ATG3 is a novel protein implicated in the development of steatosis. LAY SUMMARY: We show that autophagy-related gene 3 (ATG3) contributes to the progression of non-alcoholic fatty liver disease in humans and mice. Hepatic knockdown of ATG3 ameliorates the development of NAFLD by stimulating mitochondrial function. Thus, ATG3 is an important factor implicated in steatosis.


Sujet(s)
Protéines associées à l'autophagie/antagonistes et inhibiteurs , Stéatose hépatique/prévention et contrôle , Mitochondries du foie/métabolisme , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Animaux , Protéines associées à l'autophagie/pharmacologie , Modèles animaux de maladie humaine , Stéatose hépatique/physiopathologie , Métabolisme lipidique/génétique , Souris , Mitochondries du foie/physiologie , Protéomique/méthodes , Ubiquitin-conjugating enzymes/pharmacologie
9.
J Med Chem ; 65(1): 163-190, 2022 01 13.
Article de Anglais | MEDLINE | ID: mdl-34939411

RÉSUMÉ

DCN1, a co-E3 ligase, interacts with UBC12 and activates cullin-RING ligases (CRLs) by catalyzing cullin neddylation. Although DCN1 has been recognized as an important therapeutic target for human diseases, its role in the cardiovascular area remains unknown. Here, we first found that DCN1 was upregulated in isolated cardiac fibroblasts (CFs) treated by angiotensin (Ang) II and in mouse hearts after pressure overload. Then, structure-based optimizations for DCN1-UBC12 inhibitors were performed based on our previous work, yielding compound DN-2. DN-2 specifically targeted DCN1 at molecular and cellular levels as shown by molecular modeling studies, HTRF, cellular thermal shift and co-immunoprecipitation assays. Importantly, DN-2 effectively reversed Ang II-induced cardiac fibroblast activation, which was associated with the inhibition of cullin 3 neddylation. Our findings indicate a potentially unrecognized role of DCN1 inhibition for anticardiac fibrotic effects. DN-2 may be used as a lead compound for further development.


Sujet(s)
Antifibrotiques , Découverte de médicament , Antienzymes , Fibrose , Cardiopathies , Protéines et peptides de signalisation intracellulaire , Pyrimidines , Ubiquitin-conjugating enzymes , Animaux , Mâle , Souris , Rats , Antifibrotiques/composition chimique , Antifibrotiques/pharmacologie , Cullines/métabolisme , Antienzymes/composition chimique , Antienzymes/pharmacologie , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Fibrose/traitement médicamenteux , Fibrose/métabolisme , Fibrose/anatomopathologie , Cardiopathies/traitement médicamenteux , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Protéines et peptides de signalisation intracellulaire/antagonistes et inhibiteurs , Souris de lignée C57BL , Protéine NEDD8/métabolisme , Pyrimidines/composition chimique , Rat Sprague-Dawley , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitines
10.
Cells ; 10(8)2021 08 03.
Article de Anglais | MEDLINE | ID: mdl-34440743

RÉSUMÉ

The ubiquitin proteasome system (UPS) is the main player of skeletal muscle wasting, a common characteristic of many diseases (cancer, etc.) that negatively impacts treatment and life prognosis. Within the UPS, the E3 ligase MuRF1/TRIM63 targets for degradation several myofibrillar proteins, including the main contractile proteins alpha-actin and myosin heavy chain (MHC). We previously identified five E2 ubiquitin-conjugating enzymes interacting with MuRF1, including UBE2L3/UbcH7, that exhibited a high affinity for MuRF1 (KD = 50 nM). Here, we report a main effect of UBE2L3 on alpha-actin and MHC degradation in catabolic C2C12 myotubes. Consistently UBE2L3 knockdown in Tibialis anterior induced hypertrophy in dexamethasone (Dex)-treated mice, whereas overexpression worsened the muscle atrophy of Dex-treated mice. Using combined interactomic approaches, we also characterized the interactions between MuRF1 and its substrates alpha-actin and MHC and found that MuRF1 preferentially binds to filamentous F-actin (KD = 46.7 nM) over monomeric G-actin (KD = 450 nM). By contrast with actin that did not alter MuRF1-UBE2L3 affinity, binding of MHC to MuRF1 (KD = 8 nM) impeded UBE2L3 binding, suggesting that differential interactions prevail with MuRF1 depending on both the substrate and the E2. Our data suggest that UBE2L3 regulates contractile proteins levels and skeletal muscle atrophy.


Sujet(s)
Actines/métabolisme , Ubiquitin-conjugating enzymes/métabolisme , Animaux , Lignée cellulaire , Dexaméthasone/pharmacologie , Antigènes d'histocompatibilité de classe II/métabolisme , Mâle , Souris , Souris de lignée C57BL , Fibres musculaires squelettiques/cytologie , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/métabolisme , Protéines du muscle/métabolisme , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Liaison aux protéines , Interférence par ARN , Petit ARN interférent/métabolisme , Protéines à motif tripartite/métabolisme , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitin-conjugating enzymes/génétique , Ubiquitin-protein ligases/métabolisme
11.
ACS Chem Biol ; 16(9): 1745-1756, 2021 09 17.
Article de Anglais | MEDLINE | ID: mdl-34397214

RÉSUMÉ

Transfer of ubiquitin to substrate proteins regulates most processes in eukaryotic cells. E2 enzymes are a central component of the ubiquitin machinery, and generally determine the type of ubiquitin signal generated and thus the ultimate fate of substrate proteins. The E2, Ube2k, specifically builds degradative ubiquitin chains on diverse substrates. Here we have identified protein-based reagents, called ubiquitin variants (UbVs), that bind tightly and specifically to Ube2k. Crystal structures reveal that the UbVs bind to the E2 enzyme at a hydrophobic cleft that is distinct from the active site and previously identified ubiquitin binding sites. We demonstrate that the UbVs are potent inhibitors of Ube2k and block both ubiquitin charging of the E2 enzyme and E3-catalyzed ubiquitin transfer. The binding site of the UbVs suggests they directly clash with the ubiquitin activating enzyme, while potentially disrupting interactions with E3 ligases via allosteric effects. Our data reveal the first protein-based inhibitors of Ube2k and unveil a hydrophobic groove that could be an effective target for inhibiting Ube2k and other E2 enzymes.


Sujet(s)
Protéines mutantes/métabolisme , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitine/métabolisme , Catalyse , Domaine catalytique , Escherichia coli/génétique , Interactions hydrophobes et hydrophiles , Modèles moléculaires , Protéines mutantes/génétique , Liaison aux protéines , Domaines protéiques , Maturation post-traductionnelle des protéines , Relation structure-activité , Spécificité du substrat , Ubiquitine/génétique , Ubiquitin-activating enzymes/métabolisme
12.
Bioorg Chem ; 114: 105092, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34147881

RÉSUMÉ

A collection of 9050 natural products, their derivatives, and mimetics, was virtually screened against the human Atg3-Atg8 (Atg - autophagy) binding scaffold. By blocking this interaction, the lipidation of Atg8 does not occur and the formation of autophagosomes is inhibited. Forty-three (43) potential ligands were tested using enhanced Green Fluorescent Protein (eGFP) tagged LC3, the human ortholog of Atg8, in MCF7 breast cancer cells. Three hits showed single digit µM IC50 values with AT110, an isoflavone derivative, being the best at 1.2 ± 0.6 µM. Molecular modelling against Atg8 in conjunction with structural activity relationship (SAR) strongly supports the binding to this target. Testing in a panel of cancer cell lines showed little cytotoxic effect as compared to chloroquine. However, same concentration of AT110 was shown to be toxic to young zebrafish embryos. This can be explained in terms of the autophagy process being very active in the zebrafish embryos rendering them susceptible to AT110 whereas in the cancer cells tested the autophagy is not usually active. Nevertheless, AT110 blocks autophagy flux in the zebrafish confirming that the ligand is modulating autophagy. A small molecule non-cytotoxic autophagy inhibitor would open the door for adjunct therapies to bolster many established anticancer drugs, reducing their efficacious concentration thus limiting undesirable site effects. In addition, since many cancer types rely on the autophagy mechanism to survive a therapeutic regime, recurrence can potentially be reduced. The discovery of AT110 is an important step in establishing such an adjunct therapy.


Sujet(s)
Antinéoplasiques/pharmacologie , Famille de la protéine-8 associée à l'autophagie/antagonistes et inhibiteurs , Protéines associées à l'autophagie/antagonistes et inhibiteurs , Autophagie/effets des médicaments et des substances chimiques , Isoflavones/pharmacologie , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/composition chimique , Famille de la protéine-8 associée à l'autophagie/métabolisme , Protéines associées à l'autophagie/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Évaluation préclinique de médicament , Tests de criblage d'agents antitumoraux , Développement embryonnaire/effets des médicaments et des substances chimiques , Humains , Isoflavones/composition chimique , Structure moléculaire , Relation structure-activité , Ubiquitin-conjugating enzymes/métabolisme , Danio zébré/embryologie
13.
J Med Chem ; 64(9): 5850-5862, 2021 05 13.
Article de Anglais | MEDLINE | ID: mdl-33945681

RÉSUMÉ

The cullin-RING ubiquitin ligases (CRLs) are ubiquitin E3 enzymes that play a key role in controlling proteasomal degradation and are activated by neddylation. We previously reported inhibitors that target CRL activation by disrupting the interaction of defective in cullin neddylation 1 (DCN1), a CRL neddylation co-E3, and UBE2M, a neddylation E2. Our first-generation inhibitors possessed poor oral bioavailability and fairly rapid clearance that hindered the study of acute inhibition of DCN-controlled CRL activity in vivo. Herein, we report studies to improve the pharmacokinetic performance of the pyrazolo-pyridone inhibitors. The current best inhibitor, 40, inhibits the interaction of DCN1 and UBE2M, blocks NEDD8 transfer in biochemical assays, thermally stabilizes cellular DCN1, and inhibits anchorage-independent growth in a DCN1 amplified squamous cell carcinoma cell line. Additionally, we demonstrate that a single oral 50 mg/kg dose sustains plasma exposures above the biochemical IC90 for 24 h in mice.


Sujet(s)
Protéines et peptides de signalisation intracellulaire/métabolisme , Pyrazoles/composition chimique , Pyridines/composition chimique , Ubiquitin-conjugating enzymes/métabolisme , Administration par voie orale , Animaux , Sites de fixation , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cristallographie aux rayons X , Conception de médicament , Stabilité de médicament , Période , Humains , Protéines et peptides de signalisation intracellulaire/antagonistes et inhibiteurs , Souris , Simulation de dynamique moléculaire , Pyrazoles/métabolisme , Pyrazoles/pharmacologie , Pyridines/métabolisme , Pyridines/pharmacologie , Relation structure-activité , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs
14.
J Immunol ; 206(10): 2376-2385, 2021 05 15.
Article de Anglais | MEDLINE | ID: mdl-33893171

RÉSUMÉ

NLRP3 inflammasome plays an important role in innate immune system through recognizing pathogenic microorganisms and danger-associated molecules. Deubiquitination of NLRP3 has been shown to be essential for its activation, yet the functions of Ubc13, the K63-linked specific ubiquitin-conjugating enzyme E2, in NLRP3 inflammasome activation are not known. In this study, we found that in mouse macrophages, Ubc13 knockdown or knockout dramatically impaired NLRP3 inflammasome activation. Catalytic activity is required for Ubc13 to control NLRP3 activation, and Ubc13 pharmacological inhibitor significantly attenuates NLRP3 inflammasome activation. Mechanistically, Ubc13 associates with NLRP3 and promotes its K63-linked polyubiquitination. Through mass spectrum and biochemical analysis, we identified lysine 565 and lysine 687 as theK63-linked polyubiquitination sites of NLRP3. Collectively, our data suggest that Ubc13 potentiates NLRP3 inflammasome activation via promoting site-specific K63-linked ubiquitination of NLRP3. Our study sheds light on mechanisms of NLRP3 inflammasome activation and identifies that targeting Ubc13 could be an effective therapeutic strategy for treating aberrant NLRP3 inflammasome activation-induced pathogenesis.


Sujet(s)
Inflammasomes/métabolisme , Lysine/métabolisme , Macrophages/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Polyubiquitine/métabolisme , Ubiquitin-conjugating enzymes/déficit , Ubiquitination/génétique , Animaux , Cellules HEK293 , Humains , Inflammasomes/immunologie , Macrophages/immunologie , Souris , Souris transgéniques , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Liaison aux protéines , Transfection , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitin-conjugating enzymes/génétique , Ubiquitination/effets des médicaments et des substances chimiques
15.
Exp Cell Res ; 403(2): 112614, 2021 06 15.
Article de Anglais | MEDLINE | ID: mdl-33905671

RÉSUMÉ

In this study, we explored the regulatory effects of nitrogen permease regulator 2-like (NPRL2) on niraparib sensitivity, a PARP inhibitor (PARPi) in castrate-resistant prostate cancer (CRPC). Data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) program were retrospectively examined. Gene-set enrichment analysis (GSEA) was conducted between high and low NRPL2 expression prostate adenocarcinoma (PRAD) cases in TCGA. CCK-8 assay, Western blot analysis of apoptotic proteins, and flow cytometric analysis of apoptosis were applied to test niraparib sensitivity. Immunofluorescent (IF) staining and co-immunoprecipitation (co-IP) were conducted to explore the proteins interacting with NPRL2. Results showed that the upregulation of a canonical protein-coding transcript of NPRL2 (ENST00000232501.7) is associated with an unfavorable prognosis. Bioinformatic analysis predicts a physical interaction between NPRL2 and UBE2M, which is validated by a following Co-IP assay. This interaction increases NPRL2 stability by reducing polyubiquitination and proteasomal degradation. Depletion of NPRL2 or UBE2M significantly increases the niraparib sensitivity of CRPC cells and enhances niraparib-induced tumor growth inhibition in vivo. NPRL2 cooperatively enhances UBE2M-mediated neddylation and facilitates the degradation of multiple substrates of Cullin-RING E3 ubiquitin ligases (CRLs). In conclusion, this study identified a novel NPRL2-UBE2M complex in modulating neddylation and niraparib sensitivity of CRPC cells. Therefore, targeting NPRL2 might be considered as an adjuvant strategy for PARPi therapy.


Sujet(s)
Adénocarcinome/génétique , Antinéoplasiques/usage thérapeutique , Régulation de l'expression des gènes tumoraux , Indazoles/usage thérapeutique , Pipéridines/usage thérapeutique , Tumeurs prostatiques résistantes à la castration/génétique , Protéines suppresseurs de tumeurs/génétique , Ubiquitin-conjugating enzymes/génétique , Adénocarcinome/traitement médicamenteux , Adénocarcinome/mortalité , Adénocarcinome/anatomopathologie , Animaux , Atlas comme sujet , Caspase-3/génétique , Caspase-3/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Bases de données génétiques , Humains , Mâle , Souris , Souris de lignée BALB C , Souris nude , Protéine NEDD8/métabolisme , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/mortalité , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Maturation post-traductionnelle des protéines , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction du signal , Analyse de survie , Protéines suppresseurs de tumeurs/métabolisme , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitin-conjugating enzymes/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Protéine Bax/génétique , Protéine Bax/métabolisme
16.
Eur J Med Chem ; 217: 113326, 2021 May 05.
Article de Anglais | MEDLINE | ID: mdl-33756127

RÉSUMÉ

Defective in cullin neddylation 1(DCN1) is a co-E3 ligase that is important for cullin neddylation. Dysregulation of DCN1 highly correlates with the development of various cancers. Herein, from the initial high-throughput screening, a novel hit compound 5a containing a phenyltriazole thiol core (IC50 value of 0.95 µM for DCN1-UBC12 interaction) was discovered. Further structure-based optimization leads to the development of SK-464 (IC50 value of 26 nM). We found that SK-464 not only directly bound to DCN1 in vitro, but also engaged cellular DCN1, suppressed the neddylation of cullin3, and hindered the migration and invasion of two DCN1-overexpressed squamous carcinoma cell lines (KYSE70 and H2170). These findings indicate that SK-464 may be a novel lead compound targeting DCN1-UBC12 interaction.


Sujet(s)
Développement de médicament , Antienzymes/pharmacologie , Protéines et peptides de signalisation intracellulaire/antagonistes et inhibiteurs , Thiols/pharmacologie , Triazoles/pharmacologie , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Humains , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Simulation de docking moléculaire , Structure moléculaire , Relation structure-activité , Thiols/synthèse chimique , Thiols/composition chimique , Triazoles/synthèse chimique , Triazoles/composition chimique , Ubiquitin-conjugating enzymes/génétique , Ubiquitin-conjugating enzymes/métabolisme
17.
Biochem Biophys Res Commun ; 554: 1-6, 2021 05 21.
Article de Anglais | MEDLINE | ID: mdl-33770685

RÉSUMÉ

Exposure to fine particulate matter (PM2.5) increases the incidence of allergic rhinitis (AR). microRNA (miRNA) can regulate cell proliferation, invasion and apoptosis. However, the mechanism of miR-338-3p in mediating PM2.5-induced autophagy in AR animal models remains unknown. To explore the mechanism of miR-338-3p in PM2.5-induced autophagy in AR, the human nasal epithelium cells and AR model exposed to PM2.5 were deployed. The results showed that miR-338-3p was down-regulated in both nasal mucosa of PM2.5-exacerbated AR rat models and PM2.5-treated RPMI-2650 cells. Forced expression of miR-338-3p could inhibit autophagy in vitro. miR-338-3p specifically bound to UBE2Q1 3'-untranslated region (3' UTR) and negatively regulated its expression. Overexpression of UBE2Q1 attenuated the inhibitory effects of miR-338-3p on PM2.5-induced autophagy of RPMI-2650 cells through AKT/mTOR pathway. Moreover, our in vivo study found that after administration of agomiR-338-3p in AR rats model, the expression of autophagy-related proteins decreased and nasal symptoms alleviated. In conclusion, this study revealed that miR-338-3p acts as an autophagy suppressor in PM2.5-exacerbated AR by directly targeting UBE2Q1 and affecting AKT/mTOR pathway.


Sujet(s)
microARN/génétique , Muqueuse nasale/métabolisme , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Rhinite allergique/prévention et contrôle , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Polluants atmosphériques/analyse , Animaux , Autophagie/physiologie , Lignée cellulaire , Modèles animaux de maladie humaine , Humains , Muqueuse nasale/effets des médicaments et des substances chimiques , Muqueuse nasale/anatomopathologie , Matière particulaire/administration et posologie , Protéines proto-oncogènes c-akt/métabolisme , Rats , Rat Sprague-Dawley , Rhinite allergique/étiologie , Rhinite allergique/génétique , Rhinite allergique/métabolisme , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/métabolisme , Ubiquitin-conjugating enzymes/métabolisme
18.
Oncogene ; 40(5): 1027-1042, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33323973

RÉSUMÉ

Dysregulation of the Wnt/ß-catenin signaling pathway is critically involved in gastric cancer (GC) progression. However, current Wnt pathway inhibitors being studied in preclinical or clinical settings for other cancers such as colorectal and pancreatic cancers are either too cytotoxic or insufficiently efficacious for GC. Thus, we screened new potent targets from ß-catenin destruction complex associated with GC progression from clinical samples, and found that scaffolding protein RACK1 deficiency plays a significant role in GC progression, but not APC, AXIN, and GSK3ß. Then, we identified its upstream regulator UBE2T which promotes GC progression via hyperactivating the Wnt/ß-catenin signaling pathway through the ubiquitination and degradation of RACK1 at the lysine K172, K225, and K257 residues independent of an E3 ligase. Indeed, UBE2T protein level is negatively associated with prognosis in GC patients, suggesting that UBE2T is a promising target for GC therapy. Furthermore, we identified a novel UBE2T inhibitor, M435-1279, and suggested that M435-1279 acts inhibit the Wnt/ß-catenin signaling pathway hyperactivation through blocking UBE2T-mediated degradation of RACK1, resulting in suppression of GC progression with lower cytotoxicity in the meantime. Overall, we found that increased UBE2T levels promote GC progression via the ubiquitination of RACK1 and identified a novel potent inhibitor providing a balance between growth inhibition and cytotoxicity as well, which offer a new opportunity for the specific GC patients with aberrant Wnt/ß-catenin signaling.


Sujet(s)
Protéines tumorales/génétique , Récepteurs de kinase-C activée/génétique , Tumeurs de l'estomac/traitement médicamenteux , Ubiquitin-conjugating enzymes/génétique , bêta-Caténine/génétique , Animaux , Complexe de signalisation de l'axine/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mâle , Souris , Protéines tumorales/antagonistes et inhibiteurs , Récepteurs de kinase-C activée/antagonistes et inhibiteurs , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitination/effets des médicaments et des substances chimiques , Voie de signalisation Wnt/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Cell Death Differ ; 28(2): 671-684, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-32901121

RÉSUMÉ

UBE2O, an E2/E3 hybrid ubiquitin-protein ligase, has been implicated in the regulation of adipogenesis, erythroid differentiation, and tumor proliferation. However, its role in cancer radioresistance remains completely unknown. Here, we uncover that UBE2O interacts and targets Mxi1 for ubiquitination and degradation at the K46 residue. Furthermore, we show that genetical or pharmacological blockade of UBE2O impairs tumor progression and radioresistance in lung cancer in vitro and in vivo, and these effects can be restored by Mxi1 inhibition. Moreover, we demonstrate that UBE2O is overexpressed and negatively correlated with Mxi1 protein levels in lung cancer tissues. Collectively, our work reveals that UBE2O facilitates tumorigenesis and radioresistance by promoting Mxi1 ubiquitination and degradation, suggesting that UBE2O is an attractive radiosensitization target for the treatment of lung cancer.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Tumeurs du poumon/métabolisme , Radiotolérance , Protéines suppresseurs de tumeurs/métabolisme , Ubiquitin-conjugating enzymes/métabolisme , Ubiquitination , Animaux , Lignée cellulaire tumorale , Évolution de la maladie , Humains , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/thérapie , Souris , Souris de lignée BALB C , Souris nude , Protéolyse , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Tests d'activité antitumorale sur modèle de xénogreffe
20.
Br J Cancer ; 124(3): 531-538, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33071285

RÉSUMÉ

An urgent medical need to develop novel treatment strategies for patients with pancreatic ductal adenocarcinoma (PDAC) exists. However, despite various efforts in the histopathological and molecular subtyping of PDAC, novel targeted or specific therapies have not been established. Posttranslational modifications (PTMs) with ubiquitin-like proteins, including small ubiquitin-like modifiers (SUMOs), mediate numerous processes that can contribute to the fitness and survival of cancer cells. The contribution of SUMOylation to transcriptional control, DNA repair pathways, mitotic progression, and oncogenic signalling has been described. Here we review functions of the SUMO pathway in PDAC, with a special focus on its connection to an aggressive subtype of the disease characterised by high MYC activity, and discuss SUMOylation inhibitors under development for precise PDAC therapies.


Sujet(s)
Carcinome du canal pancréatique/métabolisme , Tumeurs du pancréas/métabolisme , Maturation post-traductionnelle des protéines , Protéines proto-oncogènes c-myc/métabolisme , Petites protéines modificatrices apparentées à l'ubiquitine/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Survie cellulaire/physiologie , Cysteine endopeptidases/effets des médicaments et des substances chimiques , Cysteine endopeptidases/métabolisme , Réparation de l'ADN/physiologie , Antienzymes/pharmacologie , Humains , Mitose/physiologie , Tumeurs du pancréas/anatomopathologie , Protéines liant le poly-adp-ribose/métabolisme , Protéine de la leucémie promyélocytaire/métabolisme , Inhibiteurs de STAT activés/métabolisme , Protéines proto-oncogènes c-myc/génétique , Protéine SUMO-1/antagonistes et inhibiteurs , Protéine SUMO-1/métabolisme , Transduction du signal/physiologie , Petites protéines modificatrices apparentées à l'ubiquitine/antagonistes et inhibiteurs , Sumoylation/effets des médicaments et des substances chimiques , Mutations synthétiques létales , Ubiquitin-conjugating enzymes/antagonistes et inhibiteurs , Ubiquitin-conjugating enzymes/métabolisme , Ubiquitines/antagonistes et inhibiteurs , Ubiquitines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE