Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 176
Filtrer
1.
J Exp Clin Cancer Res ; 43(1): 177, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926803

RÉSUMÉ

BACKGROUND: Paclitaxel (PTX) treatment resistance is an important factor leading to poor prognosis in triple-negative breast cancer (TNBC), therefore there is an urgent need to identify new target for combination therapy. Neddylation is a post-translational process that introduces a ubiquitin-like protein called neural precursor cell expressed developmentally downregulated protein 8 (NEDD8). Previous studies have found that neddylation is activated in multiple tumors, but its relationship with PTX chemotherapy sensitivity has not been reported. METHODS: Differences in UBC12 and NEDD8 expression levels between PTX-sensitive and PTX-insensitive TNBC tissues were validated using public databases and immunohistochemistry. The in vitro and in vivo functional experiments were used to observe the effect of neddylation inhibition combined with PTX therapy on tumor progression. Co-IP, western blot and PCR assays were used to investigate the molecular mechanisms. Molecular docking was used to simulate the protein binding of UBC12 and TRIM25. Molecular dynamics simulation was used to observe the changes in TRIM25 protein conformation. RESULTS: We found that in TNBC that is insensitive to PTX, NEDD8 and NEDD8 conjugating enzyme UBC12 are highly expressed. Treatment with the NEDD8-activating enzyme (NAE) inhibitor mln4924 or knockdown of UBC12 significantly increased the sensitivity of the tumor to PTX, and this increase in sensitivity is related to UBC12-mediated autophagy activation. Mechanistically, UBC12 can transfer NEDD8 to E3 ubiquitin ligase tripartite motif containing 25 (TRIM25) at K117. Molecular dynamics simulations indicate that the neddylation modification of TRIM25 reduces steric hindrance in its RING domain, facilitating the binding of TRIM25 and ubiquitylated substrates. Subsequently, TRIM25 promotes the nuclear translocation of transcription factor EB (TFEB) and transcription of autophagy related genes by increasing K63-polyubiquitination of TFEB, thereby reducing tumor sensitivity to PTX. CONCLUSIONS: Neddylation is activated in PTX-insensitive TNBC. Specifically, autophagy gene transcriptional activation mediated by the UBC12/TRIM25/TFEB axis reduces TNBC sensitivity to PTX. Neddylation suppression combination with PTX treatment shows a synergistic anti-tumor effect.


Sujet(s)
Autophagie , Protéine NEDD8 , Paclitaxel , Protéines à motif tripartite , Tumeurs du sein triple-négatives , Ubiquitin-protein ligases , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Femelle , Souris , Animaux , Autophagie/effets des médicaments et des substances chimiques , Protéine NEDD8/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Lignée cellulaire tumorale , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Cyclopentanes/pharmacologie , Résistance aux médicaments antinéoplasiques , Tests d'activité antitumorale sur modèle de xénogreffe , Ubiquitin-conjugating enzymes/métabolisme , Ubiquitin-conjugating enzymes/génétique
2.
Elife ; 122024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38865175

RÉSUMÉ

Philadelphia chromosome-positive (Ph+) leukemia is a fatal hematological malignancy. Although standard treatments with tyrosine kinase inhibitors (TKIs) have achieved remarkable success in prolonging patient survival, intolerance, relapse, and TKI resistance remain serious issues for patients with Ph+ leukemia. Here, we report a new leukemogenic process in which RAPSYN and BCR-ABL co-occur in Ph+ leukemia, and RAPSYN mediates the neddylation of BCR-ABL. Consequently, neddylated BCR-ABL enhances the stability by competing its c-CBL-mediated degradation. Furthermore, SRC phosphorylates RAPSYN to activate its NEDD8 E3 ligase activity, promoting BCR-ABL stabilization and disease progression. Moreover, in contrast to in vivo ineffectiveness of PROTAC-based degraders, depletion of RAPSYN expression, or its ligase activity decreased BCR-ABL stability and, in turn, inhibited tumor formation and growth. Collectively, these findings represent an alternative to tyrosine kinase activity for the oncoprotein and leukemogenic cells and generate a rationale of targeting RAPSYN-mediated BCR-ABL neddylation for the treatment of Ph+ leukemia.


Chronic myeloid leukemia (CML for short) accounts for about 15% of all blood cancers diagnosed in adults in the United States. The condition is characterized by the overproduction of immature immune cells that interfere with proper blood function. It is linked to a gene recombination (a type of mutation) that leads to white blood cells producing an abnormal 'BCR-ABL' enzyme which is always switched on. In turn, this overactive protein causes the cells to live longer and divide uncontrollably. Some of the most effective drugs available to control the disease today work by blocking the activity of BCR-ABL. Yet certain patients can become resistant to these treatments over time, causing them to relapse. Other approaches are therefore needed to manage this disease; in particular, a promising avenue of research consists in exploring whether it is possible to reduce the amount of the enzyme present in diseased cells. As part of this effort, Zhao, Dai, Li, Zhang et al. focused on RAPSYN, a scaffolding protein previously unknown in CML cells. In other tissues, it has recently been shown to participate in neddylation ­ a process by which proteins receive certain chemical 'tags' that change the way they behave. The experiments revealed that, compared to healthy volunteers, RAPSYN was present at much higher levels in the white blood cells of CML patients. Experimentally lowering the amount of RAPSYN in CML cells led these to divide less quickly ­ both in a dish and when injected in mice, while also being linked to decreased levels of BCR-ABL. Additional biochemical experiments indicated that RAPSYN sticks with BCR-ABL to add chemical 'tags' that protect the abnormal protein against degradation, therefore increasing its overall levels. Finally, the team showed that SRC, an enzyme often dysregulated in emerging cancers, can activate RAPSYN's ability to conduct neddylation; such mechanism could promote BCR-ABL stabilization and, in turn, disease progression. Taken together, these experiments indicate a new way by which BCR-ABL levels are controlled. Future studies should investigate whether RAPSYN also stabilizes BCR-ABL in patients whose leukemias have become resistant to existing drugs. Eventually, RAPSYN may offer a new target for overcoming drug-resistance in CML patients.


Sujet(s)
Protéines de fusion bcr-abl , Leucémie myéloïde chronique BCR-ABL positive , Protéines du muscle , Animaux , Humains , Souris , Lignée cellulaire tumorale , Protéines de fusion bcr-abl/métabolisme , Protéines de fusion bcr-abl/génétique , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Protéine NEDD8/métabolisme , Protéine NEDD8/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéines du muscle/métabolisme
3.
J Nanobiotechnology ; 22(1): 247, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38741123

RÉSUMÉ

Tyrosine kinase inhibitors have been the standard treatment for patients with Philadelphia chromosome-positive (Ph+) leukemia. However, a series of issues, including drug resistance, relapse and intolerance, are still an unmet medical need. Here, we report the targeted siRNA-based lipid nanoparticles in Ph+ leukemic cell lines for gene therapy of Ph+ leukemia, which specifically targets a recently identified NEDD8 E3 ligase RAPSYN in Ph+ leukemic cells to disrupt the neddylation of oncogenic BCR-ABL. To achieve the specificity for Ph+ leukemia therapy, a single-chain fragment variable region (scFv) of anti-CD79B monoclonal antibody was covalently conjugated on the surface of OA2-siRAPSYN lipid nanoparticles to generate the targeted lipid nanoparticles (scFv-OA2-siRAPSYN). Through effectively silencing RAPSYN gene in leukemic cell lines by the nanoparticles, BCR-ABL was remarkably degraded accompanied by the inhibition of proliferation and the promotion of apoptosis. The specific targeting, therapeutic effects and systemic safety were further evaluated and demonstrated in cell line-derived mouse models. The present study has not only addressed the clinical need of Ph+ leukemia, but also enabled gene therapy against a less druggable target.


Sujet(s)
Protéines de fusion bcr-abl , Nanoparticules , Protéines de fusion bcr-abl/génétique , Protéines de fusion bcr-abl/métabolisme , Animaux , Humains , Souris , Lignée cellulaire tumorale , Nanoparticules/composition chimique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Extinction de l'expression des gènes , Petit ARN interférent , Protéine NEDD8/métabolisme , Protéine NEDD8/génétique , Souris de lignée BALB C , Apoptose/effets des médicaments et des substances chimiques , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/thérapie , Thérapie génétique/méthodes , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle
4.
Free Radic Biol Med ; 219: 76-87, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38604315

RÉSUMÉ

Diabetic retinopathy (DR) is a highly hazardous and widespread complication of diabetes mellitus (DM). The accumulated reactive oxygen species (ROS) play a central role in DR development. The aim of this research was to examine the impact and mechanisms of mesenchymal stem cell (MSC)-derived small extracellular vesicles (sEV) on regulating ROS and retinal damage in DR. Intravitreal injection of sEV inhibited Cullin3 neddylation, stabilized Nrf2, decreased ROS, reduced retinal inflammation, suppressed Müller gliosis, and mitigated DR. Based on MSC-sEV miRNA sequencing, bioinformatics software, and dual-luciferase reporter assay, miR-143-3p was identified to be the key effector for MSC-sEV's role in regulating neural precursor cell expressed developmentally down-regulated 8 (NEDD8)-mediated neddylation. sEV were able to be internalized by Müller cells. Compared to advanced glycation end-products (AGEs)-induced Müller cells, sEV coculture decreased Cullin3 neddylation, activated Nrf2 signal pathway to combat ROS-induced inflammation. The barrier function of endothelial cells was impaired when endothelial cells were treated with the supernatant of AGEs-induced Müller cells, but was restored when treated with supernatant of AGEs-induced Müller cells cocultured with sEV. The protective effect of sEV was, however, compromised when miR-143-3p was inhibited in sEV. Moreover, the protective efficacy of sEV was diminished when NEDD8 was overexpressed in Müller cells. These findings showed MSC-sEV delivered miR-143-3p to inhibit Cullin3 neddylation, stabilizing Nrf2 to counteract ROS-induced inflammation and reducing vascular leakage. Our findings suggest that MSC-sEV may be a potential nanotherapeutic agent for DR, and that Cullin3 neddylation could be a new target for DR therapy.


Sujet(s)
Rétinopathie diabétique , Vésicules extracellulaires , Cellules souches mésenchymateuses , microARN , Protéine NEDD8 , Facteur-2 apparenté à NF-E2 , Espèces réactives de l'oxygène , Animaux , Humains , Souris , Cullines/métabolisme , Cullines/génétique , Diabète expérimental/métabolisme , Diabète expérimental/anatomopathologie , Diabète expérimental/génétique , Rétinopathie diabétique/anatomopathologie , Rétinopathie diabétique/génétique , Rétinopathie diabétique/métabolisme , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/génétique , Produits terminaux de glycation avancée/métabolisme , Cellules souches mésenchymateuses/métabolisme , Souris de lignée C57BL , microARN/génétique , microARN/métabolisme , Protéine NEDD8/métabolisme , Protéine NEDD8/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Espèces réactives de l'oxygène/métabolisme , Transduction du signal
5.
Nat Commun ; 15(1): 3558, 2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38670995

RÉSUMÉ

The E3 ligase-degron interaction determines the specificity of the ubiquitin‒proteasome system. We recently discovered that FEM1B, a substrate receptor of Cullin 2-RING ligase (CRL2), recognizes C-degrons containing a C-terminal proline. By solving several cryo-EM structures of CRL2FEM1B bound to different C-degrons, we elucidate the dimeric assembly of the complex. Furthermore, we reveal distinct dimerization states of unmodified and neddylated CRL2FEM1B to uncover the NEDD8-mediated activation mechanism of CRL2FEM1B. Our research also indicates that, FEM1B utilizes a bipartite mechanism to recognize both the C-terminal proline and an upstream aromatic residue within the substrate. These structural findings, complemented by in vitro ubiquitination and in vivo cell-based assays, demonstrate that CRL2FEM1B-mediated polyubiquitination and subsequent protein turnover depend on both FEM1B-degron interactions and the dimerization state of the E3 ligase complex. Overall, this study deepens our molecular understanding of how Cullin-RING E3 ligase substrate selection mediates protein turnover.


Sujet(s)
Cryomicroscopie électronique , Protéine NEDD8 , Récepteurs à l'interleukine-17 , Ubiquitin-protein ligases , Ubiquitination , Humains , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/composition chimique , Protéine NEDD8/métabolisme , Protéine NEDD8/génétique , Proline/métabolisme , Multimérisation de protéines , Cellules HEK293 , Liaison aux protéines , Spécificité du substrat , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/composition chimique , Modèles moléculaires , Cullines/métabolisme , Cullines/composition chimique , Cullines/génétique ,
6.
Nat Commun ; 15(1): 3581, 2024 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-38678024

RÉSUMÉ

Immune checkpoint blockade therapy aims to activate the immune system to eliminate cancer cells. However, clinical benefits are only recorded in a subset of patients. Here, we leverage genome-wide CRISPR/Cas9 screens in a Tumor-Immune co-Culture System focusing on triple-negative breast cancer (TNBC). We reveal that NEDD8 loss in cancer cells causes a vulnerability to nivolumab (anti-PD-1). Genetic deletion of NEDD8 only delays cell division initially but cell proliferation is unaffected after recovery. Since the NEDD8 gene is commonly essential, we validate this observation with additional CRISPR screens and uncover enhanced immunogenicity in NEDD8 deficient cells using proteomics. In female immunocompetent mice, PD-1 blockade lacks efficacy against established EO771 breast cancer tumors. In contrast, we observe tumor regression mediated by CD8+ T cells against Nedd8 deficient EO771 tumors after PD-1 blockade. In essence, we provide evidence that NEDD8 is conditionally essential in TNBC and presents as a synergistic drug target for PD-1/L1 blockade therapy.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Protéine NEDD8 , Tumeurs du sein triple-négatives , Animaux , Femelle , Humains , Souris , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Systèmes CRISPR-Cas , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Protéine NEDD8/métabolisme , Protéine NEDD8/génétique , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie
7.
Free Radic Biol Med ; 219: 127-140, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38614228

RÉSUMÉ

Doxorubicin (DOX) is a widely utilized chemotherapeutic agent in clinical oncology for treating various cancers. However, its clinical use is constrained by its significant side effects. Among these, the development of cardiomyopathy, characterized by cardiac remodeling and eventual heart failure, stands as a major concern following DOX chemotherapy. In our current investigation, we have showcased the efficacy of MLN4924 in mitigating doxorubicin-induced cardiotoxicity through direct inhibition of the NEDD8-activating enzyme, NAE. MLN4924 demonstrated the ability to stabilize mitochondrial function post-doxorubicin treatment, diminish cardiomyocyte apoptosis, alleviate oxidative stress-induced damage in the myocardium, enhance cardiac contractile function, mitigate cardiac fibrosis, and impede cardiac remodeling associated with heart failure. At the mechanistic level, MLN4924 intervened in the neddylation process by inhibiting the NEDD8 activating enzyme, NAE, within the murine cardiac tissue subsequent to doxorubicin treatment. This intervention resulted in the suppression of NEDD8 protein expression, reduction in neddylation activity, and consequential manifestation of cardioprotective effects. Collectively, our findings posit MLN4924 as a potential therapeutic avenue for mitigating doxorubicin-induced cardiotoxicity by attenuating heightened neddylation activity through NAE inhibition, thereby offering a viable and promising treatment modality for afflicted patients.


Sujet(s)
Cardiotoxicité , Cyclopentanes , Doxorubicine , Myocytes cardiaques , Protéine NEDD8 , Pyrimidines , Animaux , Souris , Apoptose/effets des médicaments et des substances chimiques , Cardiotoxicité/traitement médicamenteux , Cardiotoxicité/anatomopathologie , Cardiotoxicité/prévention et contrôle , Cardiotoxicité/étiologie , Cardiotoxicité/métabolisme , Cyclopentanes/pharmacologie , Cyclopentanes/usage thérapeutique , Doxorubicine/effets indésirables , Souris de lignée C57BL , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Protéine NEDD8/métabolisme , Protéine NEDD8/antagonistes et inhibiteurs , Stress oxydatif/effets des médicaments et des substances chimiques , Pyrimidines/pharmacologie , Ubiquitin-activating enzymes/antagonistes et inhibiteurs , Ubiquitin-activating enzymes/métabolisme , Ubiquitin-activating enzymes/génétique
8.
Leuk Lymphoma ; 65(7): 978-988, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38489672

RÉSUMÉ

Adult T-cell leukemia (ATL), caused by HTLV-1, is the most lethal hematological malignancy. NEDD8-activating enzyme (NAE) is a component of the NEDD8 conjunction pathway that regulates cullin-RING ubiquitin ligase (CRL) activity. HTLV-1-infected T cells expressed higher levels of NAE catalytic subunit UBA3 than normal peripheral blood mononuclear cells. NAE1 knockdown inhibited proliferation of HTLV-1-infected T cells. The NAE1 inhibitor MLN4924 suppressed neddylation of cullin and inhibited the CRL-mediated turnover of tumor suppressor proteins. MLN4924 inhibited proliferation of HTLV-1-infected T cells by inducing DNA damage, leading to S phase arrest and caspase-dependent apoptosis. S phase arrest was associated with CDK2 and cyclin A downregulation. MLN4924-induced apoptosis was mediated by the upregulation of pro-apoptotic and downregulation of anti-apoptotic proteins. Furthermore, MLN4924 inhibited NF-κB, AP-1, and Akt signaling pathways and activated JNK. Therefore, neddylation inhibition is an attractive strategy for ATL therapy. Our findings support the use of MLN4924 in ATL clinical trials.


Sujet(s)
Apoptose , Prolifération cellulaire , Cyclopentanes , Virus T-lymphotrope humain de type 1 , Protéine NEDD8 , Facteur de transcription NF-kappa B , Protéines proto-oncogènes c-akt , Pyrimidines , Transduction du signal , Facteur de transcription AP-1 , Ubiquitin-activating enzymes , Humains , Pyrimidines/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription AP-1/métabolisme , Cyclopentanes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Protéine NEDD8/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Ubiquitin-activating enzymes/antagonistes et inhibiteurs , Ubiquitin-activating enzymes/métabolisme , Ubiquitin-activating enzymes/génétique , Lymphocytes T/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques , Ubiquitines/métabolisme , Cullines/métabolisme
9.
J Biol Chem ; 300(3): 105752, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38354780

RÉSUMÉ

Cullin (CUL)-RING (Really Interesting New Gene) E3 ubiquitin (Ub) ligases (CRLs) are the largest E3 family. The E3 CRL core ligase is a subcomplex formed by the CUL C-terminal domain bound with the ROC1/RBX1 RING finger protein, which acts as a hub that mediates and organizes multiple interactions with E2, Ub, Nedd8, and the ARIH family protein, thereby resulting in Ub transfer to the E3-bound substrate. This report describes the modulation of CRL-dependent ubiquitination by small molecule compounds including KH-4-43, #33, and suramin, which target the CRL core ligases. We show that both KH-4-43 and #33 inhibit the ubiquitination of CK1α by CRL4CRBN. However, either compound's inhibitory effect on this reaction is significantly reduced when a neddylated form of CRL4CRBN is used. On the other hand, both #33 and KH-4-43 inhibit the ubiquitination of ß-catenin by CRL1ß-TrCP and Nedd8-CRL1ß-TrCP almost equally. Thus, neddylation of CRL1ß-TrCP does not negatively impact the sensitivity to inhibition by #33 and KH-4-43. These findings suggest that the effects of neddylation to alter the sensitivity of CRL inhibition by KH-4-43/#33 is dependent upon the specific CRL type. Suramin, a compound that targets CUL's basic canyon, can effectively inhibit CRL1/4-dependent ubiquitination regardless of neddylation status, in contrast to the results observed with KH-4-43/#33. This observed differential drug sensitivity of KH-4-43/#33 appears to echo CUL-specific Nedd8 effects on CRLs as revealed by recent high-resolution structural biology efforts. The highly diversified CRL core ligase structures may provide opportunities for specific targeting by small molecule modulators.


Sujet(s)
Ligands , Ubiquitin-protein ligases , Ubiquitination , Animaux , Humains , Souris , bêta-Caténine/métabolisme , Protéines à répétitions de séquences bêta-transducine/métabolisme , Cullines/métabolisme , Suramine/pharmacologie , Ubiquitine/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitination/effets des médicaments et des substances chimiques , Protéine NEDD8/métabolisme
10.
Int J Oncol ; 64(4)2024 04.
Article de Anglais | MEDLINE | ID: mdl-38391033

RÉSUMÉ

Neddylation, akin to ubiquitination, represents a post­translational modification of proteins wherein neural precursor cell­expressed developmentally downregulated protein 8 (NEDD8) is modified on the substrate protein through a series of reactions. Neddylation plays a pivotal role in the growth and proliferation of animal cells. In colorectal cancer (CRC), it predominantly contributes to the proliferation, metastasis and survival of tumor cells, decreasing overall patient survival. The strategic manipulation of the NEDD8­mediated neddylation pathway holds immense therapeutic promise in terms of the potential to modulate the growth of tumors by regulating diverse biological responses within cancer cells, such as DNA damage response and apoptosis, among others. MLN4924 is an inhibitor of NEDD8, and its combined use with platinum drugs and irinotecan, as well as cycle inhibitors and NEDD activating enzyme inhibitors screened by drug repurposing, has been found to exert promising antitumor effects. The present review summarizes the recent progress made in the understanding of the role of NEDD8 in the advancement of CRC, suggesting that NEDD8 is a promising anti­CRC target.


Sujet(s)
Tumeurs colorectales , Ubiquitines , Animaux , Humains , Apoptose , Lignée cellulaire tumorale , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Protéine NEDD8/génétique , Protéine NEDD8/métabolisme , Maturation post-traductionnelle des protéines , Ubiquitines/génétique
11.
Adv Sci (Weinh) ; 11(9): e2305907, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38126621

RÉSUMÉ

Cell cycle dysregulation is a defining feature of breast cancer. Here, 1-methyl-nicotinamide (1-MNA), metabolite of nicotinamide N-methyltransferase(NNMT) is identified, as a novel driver of cell-cycle progression in breast cancer. NNMT, highly expressed in breast cancer tissues, positively correlates with tumor grade, TNM stage, Ki-67 index, and tumor size. Ablation of NNMT expression dramatically suppresses cell proliferation and causes cell-cycle arrest in G0/G1 phase. This phenomenon predominantly stems from the targeted action of 1-MNA, resulting in a specific down-regulation of p27 protein expression. Mechanistically, 1-MNA expedites the degradation of p27 proteins by enhancing cullin-1 neddylation, crucial for the activation of Cullin-1-RING E3 ubiquitin ligase(CRL1)-an E3 ubiquitin ligase targeting p27 proteins.  NNMT/1-MNA specifically up-regulates the expression of UBC12, an E2 NEDD8-conjugating enzyme required for cullin-1 neddylation. 1-MNA showes high binding affinity to UBC12, extending the half-life of UBC12 proteins via preventing their localization to lysosome for degradation. Therefore, 1-MNA is a bioactive metabolite that promotes breast cancer progression by reinforcing neddylation pathway-mediated p27 degradation. The study unveils the link between NNMT enzymatic activity with cell-cycle progression, indicating that 1-MNA may be involved in the remodeling of tumor microenvironment.


Sujet(s)
Tumeurs du sein , Cullines , Humains , Femelle , Cullines/métabolisme , Protéine NEDD8/métabolisme , Ubiquitin-protein ligases/métabolisme , Maturation post-traductionnelle des protéines , Microenvironnement tumoral , Nicotinamide N-methyltransferase/métabolisme
12.
Arch Virol ; 169(1): 6, 2023 Dec 11.
Article de Anglais | MEDLINE | ID: mdl-38081982

RÉSUMÉ

Neddylation is a post-translational modification that plays an important role not only in cancer development but also in regulating viral infection and replication. Upregulation of neddylation occurs in viral infections, and inhibition of neddylation can suppress viral replication. Neddylation is thought to enhance viral protein stability and replication. Neddylation has been reported to enhance the stability of the regulatory hepatitis B virus (HBV) X protein, modulate viral replication, and enhance hepatocarcinogenesis. Inhibition of neddylation using the NEDD8-activating enzyme E1 inhibitor MLN4924 inhibits viral replication, including that of HBV. Understanding of the role of neddylation in viral infections is critical for developing new therapeutic targets and potential treatment strategies. In this review, we discuss recent progress in the understanding of the effects of neddylation during viral infection, particularly in HBV infection, and strategies for curing viral infection by targeting the neddylation pathway.


Sujet(s)
Tumeurs , Maladies virales , Humains , Protéine NEDD8/métabolisme , Ubiquitines/génétique , Maturation post-traductionnelle des protéines , Maladies virales/traitement médicamenteux
13.
Article de Chinois | MEDLINE | ID: mdl-37905484

RÉSUMÉ

Objective:To analyze the differential expression of neural precursor cell-expressed developmentally downregulated 8(NEDD8) protein in nasal polyp tissues of patients with different pathological types of chronic rhinorhinosinusitis with nasal polyps(CRSwNP). Methods:All specimens were obtained from the specimen library of Beijing Tongren Hospital, and were all patients who underwent nasal endoscopic surgery for chronic rhinosinusitis in Beijing Tongren Hospital. Hematoxylin-eosin staining(HE) was used to detect the number of eosinophils in nasal polyps, and CRSwNP patients were grouped according to the number of eosinophils in nasal polyps, immunohistochemistry was used to detect and analyze the expression level of NEDD8 protein in nasal polyps. Results:The expression level of NEDD8 protein in nasal polyps of patients with eosinophilic chronic rhinorhinosinusitis with nasal polyps was significantly higher than that of patients with non-eosinophilic chronic rhinosinusitis and nasal polyps(P<0.05). In addition, there was a significant positive correlation between the expression level of NEDD8 protein and the number of eosinophils in nasal polyp tissue(r=0.79, P=0.02). Conclusion:There are differences in the expression of NEDD8 protein in patients with chronic rhinosinusitis and nasal polyps of different pathological types.


Sujet(s)
Polypes du nez , Rhinite , Sinusite , Humains , Polypes du nez/métabolisme , Rhinite/diagnostic , Protéine NEDD8/métabolisme , Sinusite/diagnostic , Granulocytes éosinophiles/métabolisme , Maladie chronique
14.
Theranostics ; 13(14): 5017-5056, 2023.
Article de Anglais | MEDLINE | ID: mdl-37771770

RÉSUMÉ

Protein neddylation is a post-translational modification, and its best recognized substrates are cullin family proteins, which are the core component of Cullin-RING ligases (CRLs). Given that most neddylation pathway proteins are overactivated in different cancers and fibrotic diseases, targeting neddylation becomes an emerging approach for the treatment of these diseases. To date, numerous neddylation inhibitors have been developed, of which MLN4924 has entered phase I/II/III clinical trials for cancer treatment, such as acute myeloid leukemia, melanoma, lymphoma and solid tumors. Here, we systematically describe the structures and biological functions of the critical enzymes in neddylation, highlight the medicinal chemistry advances in the development of neddylation inhibitors and propose the perspectives concerning targeting neddylation for cancer and fibrotic diseases.


Sujet(s)
Lymphomes , Tumeurs , Humains , Cullines/métabolisme , Protéine NEDD8/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Maturation post-traductionnelle des protéines
15.
Nat Chem Biol ; 19(12): 1513-1523, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37653169

RÉSUMÉ

The cullin-RING ubiquitin ligase (CRL) network comprises over 300 unique complexes that switch from inactive to activated conformations upon site-specific cullin modification by the ubiquitin-like protein NEDD8. Assessing cellular repertoires of activated CRL complexes is critical for understanding eukaryotic regulation. However, probes surveying networks controlled by site-specific ubiquitin-like protein modifications are lacking. We developed a synthetic antibody recognizing the active conformation of NEDD8-linked cullins. Implementing the probe to profile cellular networks of activated CUL1-, CUL2-, CUL3- and CUL4-containing E3s revealed the complexes responding to stimuli. Profiling several cell types showed their baseline neddylated CRL repertoires vary, and prime efficiency of targeted protein degradation. Our probe also unveiled differential rewiring of CRL networks across distinct primary cell activation pathways. Thus, conformation-specific probes can permit nonenzymatic activity-based profiling across a system of numerous multiprotein complexes, which in the case of neddylated CRLs reveals widespread regulation and could facilitate the development of degrader drugs.


Sujet(s)
Cullines , Ubiquitin-protein ligases , Cullines/génétique , Ubiquitination , Ubiquitin-protein ligases/métabolisme , Ubiquitine/métabolisme , Ubiquitines/métabolisme , Protéine NEDD8/métabolisme
16.
Cell Death Dis ; 14(7): 438, 2023 07 17.
Article de Anglais | MEDLINE | ID: mdl-37460534

RÉSUMÉ

Natural Killer (NK) cells act as important regulators in the development and progression of hematological malignancies and their suppressor activity against Multiple Myeloma (MM) cells has been confirmed in many studies. Significant changes in the distribution of NK cell subsets and dysfunctions of NK cell effector activities were described in MM patients and correlated with disease staging. Thus, restoring or enhancing the functionality of these effectors for the treatment of MM represents a critical need. Neddylation is a post-translational modification that adds a ubiquitin-like molecule, NEDD8, to the substrate protein. One of the outcomes is the activation of the Cullin Ring Ligases (CRLs), a class of ubiquitin-ligases that controls the degradation of about 20% of proteasome-regulated proteins. Overactivation of CRLs has been described in cancer and can lead to tumor growth and progression. Thus, targeting neddylation represents an attractive approach for cancer treatment. Our group has recently described how pharmacologic inhibition of neddylation increases the expression of the NKG2D activating receptor ligands, MICA and MICB, in MM cells, making these cells more susceptible to NK cell degranulation and killing. Here, we extended our investigation to the direct role of neddylation on NK cell effector functions exerted against MM. We observed that inhibition of neddylation enhanced NK cell-mediated degranulation and killing against MM cells and improved Daratumumab/Elotuzumab-mediated response. Mechanistically, inhibition of neddylation increased the expression of Rac1 and RhoA GTPases in NK cells, critical mediators for an efficient degranulation at the immunological synapse of cytotoxic lymphocytes, and augmented the levels of F-actin and perforin polarization in NK cells contacting target cells. Moreover, inhibition of neddylation partially abrogated TGFß-mediated repression of NK cell effector activity. This study describes the role of neddylation on NK cell effector functions and highlights the positive immunomodulatory effects achieved by the inhibition of this pathway in MM.


Sujet(s)
Antinéoplasiques , Myélome multiple , Humains , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Protéine NEDD8/métabolisme , Antinéoplasiques/pharmacologie , Protéines , Cellules tueuses naturelles/métabolisme , Ligases
17.
Int J Oncol ; 62(5)2023 May.
Article de Anglais | MEDLINE | ID: mdl-37083098

RÉSUMÉ

As a protein that resembles ubiquitin, neural precursor cell expressed developmentally downregulated 8 (NEDD8) takes part in neddylation, which modifies substrates in a manner similar to ubiquitination and alters the activity of target proteins. Neddylation may affect the activity of multiple signaling pathways, have a regulatory role in tumor formation, progression and metastasis, and influence the prognosis of cancer treatment. The present review summarizes the regulatory roles of NEDD8 in the MDM2­p53, NF­κB, PI3K/AKT/mTOR, hypoxia­inducible factor, Hippo and receptor tyrosine kinase signaling pathways, as well as in the development and progression of lung cancer.


Sujet(s)
Tumeurs du poumon , Ubiquitines , Humains , Protéine NEDD8/génétique , Protéine NEDD8/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Ubiquitination
18.
Int J Biol Sci ; 19(2): 377-392, 2023.
Article de Anglais | MEDLINE | ID: mdl-36632463

RÉSUMÉ

HER2 is a transmembrane receptor with intrinsic tyrosine kinase activity that is overexpressed in almost 25% of human breast cancers. Here, we report that the neddylation of HER2 is a new post-translational modification that controls its expression and oncogenic activity in human breast cancer. Two critical members in the neddylation pathway, NEDD8 and NEDD8-activating enzyme E1 subunit 1 (NAE1), are detected in human breast specimens. Overexpressed NEDD8 and NAE1 are positively correlated with HER2 expression in human breast cancer. Subsequent structure and function experiments show that HER2 directly interacts with NEDD8 and NAE1, whereas HER2 protein expression is decreased by neddylation depletion. Mechanistically, neddylation inhibition promotes the degradation of HER2 protein by improving its ubiquitination. HER2 overexpression abrogates neddylation depletion-triggered cell growth suppression. The inhibition of neddylation synergized with trastuzumab significantly suppresses growth of HER2 positive breast cancer. Collectively, this study demonstrates a previously undiscovered role of NEDD8-dependent HER2 neddylation promotes tumor growth in breast cancer.


Sujet(s)
Tumeurs du sein , Protéine NEDD8 , Protéolyse , Récepteur ErbB-2 , Ubiquitination , Femelle , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Maturation post-traductionnelle des protéines , Récepteur ErbB-2/métabolisme , Protéine NEDD8/métabolisme , Évolution de la maladie
19.
Am J Hum Genet ; 110(1): 146-160, 2023 01 05.
Article de Anglais | MEDLINE | ID: mdl-36608681

RÉSUMÉ

Neddylation has been implicated in various cellular pathways and in the pathophysiology of numerous diseases. We identified four individuals with bi-allelic variants in NAE1, which encodes the neddylation E1 enzyme. Pathogenicity was supported by decreased NAE1 abundance and overlapping clinical and cellular phenotypes. To delineate how cellular consequences of NAE1 deficiency would lead to the clinical phenotype, we focused primarily on the rarest phenotypic features, based on the assumption that these would best reflect the pathophysiology at stake. Two of the rarest features, neuronal loss and lymphopenia worsening during infections, suggest that NAE1 is required during cellular stress caused by infections to protect against cell death. In support, we found that stressing the proteasome system with MG132-requiring upregulation of neddylation to restore proteasomal function and proteasomal stress-led to increased cell death in fibroblasts of individuals with NAE1 genetic variants. Additionally, we found decreased lymphocyte counts after CD3/CD28 stimulation and decreased NF-κB translocation in individuals with NAE1 variants. The rarest phenotypic feature-delayed closure of the ischiopubic rami-correlated with significant downregulation of RUN2X and SOX9 expression in transcriptomic data of fibroblasts. Both genes are involved in the pathophysiology of ischiopubic hypoplasia. Thus, we show that NAE1 plays a major role in (skeletal) development and cellular homeostasis during stress. Our approach suggests that a focus on rare phenotypic features is able to provide significant pathophysiological insights in diseases caused by mutations in genes with pleiotropic effects.


Sujet(s)
Déficience intellectuelle , Lymphopénie , Humains , Protéine NEDD8/génétique , Protéine NEDD8/métabolisme , Transduction du signal/génétique , Déficience intellectuelle/génétique , Facteur de transcription NF-kappa B/métabolisme , Proteasome endopeptidase complex/métabolisme , Lymphopénie/génétique
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...