Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 993
Filtrer
1.
Cells ; 13(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38920658

RÉSUMÉ

The development of cell-type-specific dendritic arbors is integral to the proper functioning of neurons within their circuit networks. In this study, we examine the regulatory relationship between the cytosolic chaperonin CCT, key insulin pathway genes, and an E3 ubiquitin ligase (Cullin1) in dendritic development. CCT loss of function (LOF) results in dendritic hypotrophy in Drosophila Class IV (CIV) multi-dendritic larval sensory neurons, and CCT has recently been shown to fold components of the TOR (Target of Rapamycin) complex 1 (TORC1) in vitro. Through targeted genetic manipulations, we confirm that an LOF of CCT and the TORC1 pathway reduces dendritic complexity, while overexpression of key TORC1 pathway genes increases the dendritic complexity in CIV neurons. Furthermore, both CCT and TORC1 LOF significantly reduce microtubule (MT) stability. CCT has been previously implicated in regulating proteinopathic aggregation, thus, we examine CIV dendritic development in disease conditions as well. The expression of mutant Huntingtin leads to dendritic hypotrophy in a repeat-length-dependent manner, which can be rescued by Cullin1 LOF. Together, our data suggest that Cullin1 and CCT influence dendritic arborization through the regulation of TORC1 in both health and disease.


Sujet(s)
Cullines , Dendrites , Protéines de Drosophila , Drosophila melanogaster , Animaux , Cullines/métabolisme , Cullines/génétique , Dendrites/métabolisme , Drosophila melanogaster/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Protéine huntingtine/métabolisme , Protéine huntingtine/génétique , Larve/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Microtubules/métabolisme , Cellules réceptrices sensorielles/métabolisme , Transduction du signal , Facteurs de transcription , Chaperonine contenant TCP-1
2.
Sci Rep ; 14(1): 14912, 2024 06 28.
Article de Anglais | MEDLINE | ID: mdl-38942922

RÉSUMÉ

Breast cancer is a prevalent and significant cause of mortality in women, and manifests as six molecular subtypes. Its further histologic classification into non-invasive ductal or lobular carcinoma (DCIS) and invasive carcinoma (ILC or IDC) underscores its heterogeneity. The ubiquitin-proteasome system plays a crucial role in breast cancer, with inhibitors targeting the 26S proteasome showing promise in clinical treatment. The Cullin-RING ubiquitin ligases, including CUL3, have direct links to breast cancer. This study focuses on CUL3 as a potential biomarker, leveraging high-throughput sequencing, gene expression profiling, experimental and data analysis tools. Through comprehensive analysis using databases like GEPIA2 and UALCAN, as well as TCGA datasets, CUL3's expression and its association with prognostic values were assessed. Additionally, the impact of CUL3 overexpression was explored in MCF-7 and MDA-MB-231 breast cancer cell lines, revealing distinct differences in molecular and phenotypic characteristics. We further profiled its expression and localization in breast cancer tissues identifying prominent differences between luminal A and TNBC tumors. Conclusively, CUL3 was found to be associated with cell cycle progression, and DNA damage response, exhibiting diverse roles depending on the tumor's molecular type. It exhibits a tendency to act as an oncogene in triple-negative tumors and as a tumor suppressor in luminal A types, suggesting a potential significance in breast cancer progression and therapeutic directions.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du sein , Cullines , Régulation de l'expression des gènes tumoraux , Humains , Cullines/métabolisme , Cullines/génétique , Femelle , Pronostic , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes , Cellules MCF-7 , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme
3.
Int Ophthalmol ; 44(1): 288, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937308

RÉSUMÉ

PURPOSE: Age-related cataract (ARC) is the most common cause of visual impairment and blindness in older adults. However, the role of CUL4B in the ARC remains unclear. Therefore, we investigated CUL4B expression and its effects on apoptosis. MATERIALS AND METHODS: CUL4B expression levels were detected by a quantitative real-time polymerase chain reaction from the anterior lens capsules of patients with ARC and HLE-B3 cells treated with different concentrations of H2O2. CUL4B expression was silenced by siRNA transfection to evaluate apoptosis. CUL4B and apoptotic proteins B cell lymphoma 2 (Bcl-2), myeloid cell leukemia 1 (Mcl-1), caspase-3, cleaved caspase-3, Bax, Bak, and Bid were assessed using western blot analysis. Apoptosis was monitored using the TUNEL assay. RESULTS: CUL4B expression was downregulated in the anterior lens capsules (P < 0.0001) and H2O2-treated HLE-B3 cells (P = 0.0405). CUL4B protein levels were significantly lower in 100 µmol/L (P = 0.0012) and 200 µmol/L (P = 0.0041) H2O2-treated HLE-B3 cells than in the untreated cells. CUL4B expression was significantly knocked down at the mRNA (P = 0.0043) and protein levels (P = 0.0002) in HLE-B3 cells. Bcl-2 (P = 0.0199), Mcl-1 (P = 0.0042), and caspase-3 (P = 0.0142) were significantly downregulated, whereas cleaved caspase-3 (P = 0.0089) and Bak (P = 0.009) were significantly upregulated in the knockdown group. The TUNEL assay showed a greater induction of apoptosis. CONCLUSIONS: CUL4B downregulation promotes the apoptosis of lens epithelial cells. Our study may help in understanding the role of CUL4B in ARC pathogenesis.


Sujet(s)
Apoptose , Cataracte , Cullines , Humains , Cataracte/métabolisme , Cataracte/génétique , Cataracte/étiologie , Cullines/génétique , Cullines/métabolisme , Cullines/biosynthèse , Mâle , Femelle , Sujet âgé , Technique de Western , Réaction de polymérisation en chaine en temps réel , Adulte d'âge moyen , Vieillissement , Régulation de l'expression des gènes , Capsule du cristallin/métabolisme , Capsule du cristallin/anatomopathologie , Méthode TUNEL
4.
Signal Transduct Target Ther ; 9(1): 159, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38937432

RÉSUMÉ

The ORF9b protein, derived from the nucleocapsid's open-reading frame in both SARS-CoV and SARS-CoV-2, serves as an accessory protein crucial for viral immune evasion by inhibiting the innate immune response. Despite its significance, the precise regulatory mechanisms underlying its function remain elusive. In the present study, we unveil that the ORF9b protein of SARS-CoV-2, including emerging mutant strains like Delta and Omicron, can undergo ubiquitination at the K67 site and subsequent degradation via the proteasome pathway, despite certain mutations present among these strains. Moreover, our investigation further uncovers the pivotal role of the translocase of the outer mitochondrial membrane 70 (TOM70) as a substrate receptor, bridging ORF9b with heat shock protein 90 alpha (HSP90α) and Cullin 5 (CUL5) to form a complex. Within this complex, CUL5 triggers the ubiquitination and degradation of ORF9b, acting as a host antiviral factor, while HSP90α functions to stabilize it. Notably, treatment with HSP90 inhibitors such as GA or 17-AAG accelerates the degradation of ORF9b, leading to a pronounced inhibition of SARS-CoV-2 replication. Single-cell sequencing data revealed an up-regulation of HSP90α in lung epithelial cells from COVID-19 patients, suggesting a potential mechanism by which SARS-CoV-2 may exploit HSP90α to evade the host immunity. Our study identifies the CUL5-TOM70-HSP90α complex as a critical regulator of ORF9b protein stability, shedding light on the intricate host-virus immune response dynamics and offering promising avenues for drug development against SARS-CoV-2 in clinical settings.


Sujet(s)
COVID-19 , Cullines , Protéines du choc thermique HSP90 , SARS-CoV-2 , Ubiquitination , Réplication virale , Humains , Cullines/génétique , Cullines/métabolisme , SARS-CoV-2/génétique , SARS-CoV-2/métabolisme , SARS-CoV-2/effets des médicaments et des substances chimiques , Réplication virale/effets des médicaments et des substances chimiques , Réplication virale/génétique , Protéines du choc thermique HSP90/génétique , Protéines du choc thermique HSP90/métabolisme , COVID-19/virologie , COVID-19/génétique , COVID-19/métabolisme , COVID-19/immunologie , Ubiquitination/génétique , Cellules HEK293 , Benzoquinones/pharmacologie , Stabilité protéique , Cellules Vero , Protéines virales/génétique , Protéines virales/métabolisme , Lactames macrocycliques
5.
Aging (Albany NY) ; 16(10): 8898-8921, 2024 05 22.
Article de Anglais | MEDLINE | ID: mdl-38787355

RÉSUMÉ

BACKGROUND: As a member of the Cullin family, Cullin2 (CUL2) is involved in the development and spread of different types of cancers. However, the precise role of CUL2 in human cancer remains largely elusive. METHODS: In this study, various databases were applied to observe the CUL2 expression. Kaplan-Meier and Spearman correlation analyses were employed to investigate the potential links between CUL2 level, patient prognosis, and the infiltration of immune cells. In addition, the association between CUL2 and the efficacy of immunotherapy in an immunotherapy cohort was investigated. Moreover, the expression and distribution of CUL2 in cells were observed using the Human Protein Atlas (THPA) database. Finally, clinical tissue specimens and in vitro function assays were conducted to validate the expressions and effects of CUL2 on the biological functions in hepatocellular carcinoma (HCC) cells. RESULTS: While there are variations in CUL2 expression across different organs and cell types, it is notably upregulated in a majority of tumor tissues. In addition, CUL2 gene mutations are common in multiple cancers with low mutation rates and CUL2 is closely related to the prognosis of some cancer's patients, some immune regulatory factors, TMB, MSI, MMR genes, and DNA methylation. Further, our results found that downregulating CUL2 inhibits the proliferation, and migration abilities. CONCLUSIONS: The expression of CUL2 has an impact on the prognosis of various tumors, and this correlation is particularly noteworthy due to its significant association with the infiltration of immune cells within tumors. CUL2 was an oncogene contributing to the progression of HCC.


Sujet(s)
Carcinome hépatocellulaire , Cullines , Tumeurs du foie , Humains , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Cullines/génétique , Cullines/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/immunologie , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Pronostic , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Prolifération cellulaire/génétique , Mutation , Mouvement cellulaire/génétique
6.
Nat Commun ; 15(1): 3789, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38710693

RÉSUMÉ

The CUL3-RING E3 ubiquitin ligases (CRL3s) play an essential role in response to extracellular nutrition and stress stimuli. The ubiquitin ligase function of CRL3s is activated through dimerization. However, how and why such a dimeric assembly is required for its ligase activity remains elusive. Here, we report the cryo-EM structure of the dimeric CRL3KLHL22 complex and reveal a conserved N-terminal motif in CUL3 that contributes to the dimerization assembly and the E3 ligase activity of CRL3KLHL22. We show that deletion of the CUL3 N-terminal motif impairs dimeric assembly and the E3 ligase activity of both CRL3KLHL22 and several other CRL3s. In addition, we found that the dynamics of dimeric assembly of CRL3KLHL22 generates a variable ubiquitination zone, potentially facilitating substrate recognition and ubiquitination. These findings demonstrate that a CUL3 N-terminal motif participates in the assembly process and provide insights into the assembly and activation of CRL3s.


Sujet(s)
Motifs d'acides aminés , Cryomicroscopie électronique , Cullines , Récepteurs à l'interleukine-17 , Ubiquitin-protein ligases , Ubiquitination , Cullines/métabolisme , Cullines/composition chimique , Cullines/génétique , Humains , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/composition chimique , Ubiquitin-protein ligases/génétique , Cellules HEK293 , Multimérisation de protéines , Séquence conservée , Liaison aux protéines , Modèles moléculaires
7.
Neoplasia ; 53: 101005, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38761506

RÉSUMÉ

Colorectal cancer (CRC) stands as a prevalent malignancy globally. A pivotal event in CRC pathogenesis involves the loss-of-function mutation in the APC gene, leading to the formation of benign polyps. Despite the well-established role of APC, the contribution of CUL4B to CRC initiation in the pre-tumorous stage remains poorly understood. In this investigation, we generated a murine model by crossing ApcMin/+ mice with Cul4bΔIEC mice to achieve specific deletion of Cul4b in the gut epithelium against an ApcMin/+ background. By employing histological methods, RNA-sequencing (RNA-seq), and flow cytometry, we assessed alterations and characterized the immune microenvironment. Our results unveiled that CUL4B deficiency in gut epithelium expedited ApcMin/+ adenoma formation. Notably, CUL4B in adenomas restrained the accumulation of tumor-infiltrating myeloid-derived suppressor cells (MDSCs). In vivo inhibition of MDSCs significantly delayed the growth of CUL4B deleted ApcMin/+ adenomas. Furthermore, the addition of MDSCs to in vitro cultured ApcMin/+; Cul4bΔIEC adenoma organoids mitigated their alterations. Mechanistically, CUL4B directly interacted with the promoter of Csf3, the gene encoding granulocyte-colony stimulating factor (G-CSF) by coordinating with PRC2. Inhibiting CUL4B epigenetically activated the expression of G-CSF, promoting the recruitment of MDSCs. These findings offer novel insights into the tumor suppressor-like roles of CUL4B in regulating ApcMin/+ adenomas, suggesting a potential therapeutic strategy for CRC initiation and progression in the context of activated Wnt signaling.


Sujet(s)
Adénomes , Cullines , Modèles animaux de maladie humaine , Cellules myéloïdes suppressives , Animaux , Cullines/génétique , Cullines/métabolisme , Souris , Cellules myéloïdes suppressives/métabolisme , Cellules myéloïdes suppressives/anatomopathologie , Adénomes/anatomopathologie , Adénomes/génétique , Adénomes/métabolisme , Protéine de la polypose adénomateuse colique/génétique , Humains , Microenvironnement tumoral/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/étiologie , Délétion de gène , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme
8.
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 ,
9.
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
10.
Cell Mol Life Sci ; 81(1): 165, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38578457

RÉSUMÉ

The DNA methylation is gradually acquired during oogenesis, a process sustained by successful follicle development. However, the functional roles of methyl-CpG-binding protein 2 (MeCP2), an epigenetic regulator displaying specifical binding with methylated DNA, remains unknown in oogenesis. In this study, we found MeCP2 protein was highly expressed in primordial and primary follicle, but was almost undetectable in secondary follicles. However, in aged ovary, MeCP2 protein is significantly increased in both oocyte and granulosa cells. Overexpression of MeCP2 in growing oocyte caused transcription dysregulation, DNA hypermethylation, and genome instability, ultimately leading to follicle growth arrest and apoptosis. MeCP2 is targeted by DCAF13, a substrate recognition adaptor of the Cullin 4-RING (CRL4) E3 ligase, and polyubiquitinated for degradation in both cells and oocytes. Dcaf13-null oocyte exhibited an accumulation of MeCP2 protein, and the partial rescue of follicle growth arrest induced by Dcaf13 deletion was observed following MeCP2 knockdown. The RNA-seq results revealed that large amounts of genes were regulated by the DCAF13-MeCP2 axis in growing oocytes. Our study demonstrated that CRL4DCAF13 E3 ubiquitin ligase targets MeCP2 for degradation to ensure normal DNA methylome and transcription in growing oocytes. Moreover, in aged ovarian follicles, deceased DCAF13 and DDB1 protein were observed, indicating a potential novel mechanism that regulates ovary aging.


Sujet(s)
Protéine-2 de liaison au CpG méthylé , Ubiquitin-protein ligases , Femelle , Humains , Cullines/génétique , Cullines/métabolisme , ADN/métabolisme , Méthylation de l'ADN , Protéine-2 de liaison au CpG méthylé/génétique , Protéine-2 de liaison au CpG méthylé/métabolisme , Ovocytes/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme
11.
Proc Natl Acad Sci U S A ; 121(17): e2315018121, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38625940

RÉSUMÉ

Heterotrimeric G proteins can be regulated by posttranslational modifications, including ubiquitylation. KCTD5, a pentameric substrate receptor protein consisting of an N-terminal BTB domain and a C-terminal domain, engages CUL3 to form the central scaffold of a cullin-RING E3 ligase complex (CRL3KCTD5) that ubiquitylates Gßγ and reduces Gßγ protein levels in cells. The cryo-EM structure of a 5:5:5 KCTD5/CUL3NTD/Gß1γ2 assembly reveals a highly dynamic complex with rotations of over 60° between the KCTD5BTB/CUL3NTD and KCTD5CTD/Gßγ moieties of the structure. CRL3KCTD5 engages the E3 ligase ARIH1 to ubiquitylate Gßγ in an E3-E3 superassembly, and extension of the structure to include full-length CUL3 with RBX1 and an ARIH1~ubiquitin conjugate reveals that some conformational states position the ARIH1~ubiquitin thioester bond to within 10 Å of lysine-23 of Gß and likely represent priming complexes. Most previously described CRL/substrate structures have consisted of monovalent complexes and have involved flexible peptide substrates. The structure of the KCTD5/CUL3NTD/Gßγ complex shows that the oligomerization of a substrate receptor can generate a polyvalent E3 ligase complex and that the internal dynamics of the substrate receptor can position a structured target for ubiquitylation in a CRL3 complex.


Sujet(s)
Protéines de transport , Ubiquitin-protein ligases , Liaison aux protéines , Ubiquitination , Ubiquitin-protein ligases/métabolisme , Protéines de transport/métabolisme , Ubiquitine/métabolisme , Cullines/génétique , Cullines/métabolisme
12.
Sci Rep ; 14(1): 9906, 2024 04 30.
Article de Anglais | MEDLINE | ID: mdl-38689033

RÉSUMÉ

CUL4B, a crucial scaffolding protein in the largest E3 ubiquitin ligase complex CRL4B, is involved in a broad range of physiological and pathological processes. While previous research has shown that CUL4B participates in maintaining intestinal homeostasis and function, its involvement in facilitating intestinal recovery following ionizing radiation (IR) damage has not been fully elucidated. Here, we utilized in vivo and in vitro models to decipher the role of CUL4B in intestinal repair after IR-injury. Our findings demonstrated that prior to radiation exposure, CUL4B inhibited the ubiquitination modification of PSME3, which led to the accumulation of PSME3 and subsequent negative regulation of p53-mediated apoptosis. In contrast, after radiation, CUL4B dissociated from PSME3 and translocated into the nucleus at phosphorylated histones H2A (γH2AX) foci, thereby impeding DNA damage repair and augmenting p53-mediated apoptosis through inhibition of BRCA1 phosphorylation and RAD51. Our study elucidated the dynamic role of CUL4B in the repair of radiation-induced intestinal damage and uncovered novel molecular mechanisms underlying the repair process, suggesting a potential therapeutic strategy of intestinal damage after radiation therapy for cancers.


Sujet(s)
Apoptose , Cullines , Intestins , Régénération , Protéine p53 suppresseur de tumeur , Animaux , Humains , Souris , Apoptose/effets des radiations , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Cullines/métabolisme , Cullines/génétique , Altération de l'ADN , Réparation de l'ADN , Histone/métabolisme , Intestins/effets des radiations , Intestins/anatomopathologie , Souris de lignée C57BL , Phosphorylation/effets des radiations , Rad51 Recombinase/métabolisme , Rayonnement ionisant , Régénération/effets des radiations , Protéine p53 suppresseur de tumeur/métabolisme , Ubiquitination
13.
Life Sci Alliance ; 7(5)2024 May.
Article de Anglais | MEDLINE | ID: mdl-38453365

RÉSUMÉ

KRAS is a proto-oncogene encoding a small GTPase. Mutations contribute to ∼30% of human solid tumours, including lung adenocarcinoma, pancreatic, and colorectal carcinomas. Most KRAS activating mutations interfere with GTP hydrolysis, essential for its role as a molecular switch, leading to alterations in their molecular environment and oncogenic signalling. However, the precise signalling cascades these mutations affect are poorly understood. Here, APEX2 proximity labelling was used to profile the molecular environment of WT, G12D, G13D, and Q61H-activating KRAS mutants under starvation and stimulation conditions. Through quantitative proteomics, we demonstrate the presence of known KRAS interactors, including ARAF and LZTR1, which are differentially captured by WT and KRAS mutants. Notably, the KRAS mutations G12D, G13D, and Q61H abrogate their association with LZTR1, thereby affecting turnover. Elucidating the implications of LZTR1-mediated regulation of KRAS protein levels in cancer may offer insights into therapeutic strategies targeting KRAS-driven malignancies.


Sujet(s)
Tumeurs colorectales , Protéines proto-oncogènes p21(ras) , Humains , Protéines proto-oncogènes p21(ras)/génétique , Transduction du signal/génétique , Mutation , Ubiquitin-protein ligases , Cullines/génétique , Facteurs de transcription
14.
BMC Genomics ; 25(1): 293, 2024 Mar 19.
Article de Anglais | MEDLINE | ID: mdl-38504181

RÉSUMÉ

BACKGROUND: Alternative splicing (AS) is a principal mode of genetic regulation and one of the most widely used mechanisms to generate structurally and functionally distinct mRNA and protein variants. Dysregulation of AS may result in aberrant transcription and protein products, leading to the emergence of human diseases. Although considered important for regulating gene expression, genome-wide AS dysregulation, underlying mechanisms, and clinical relevance in knee osteoarthritis (OA) remain unelucidated. Therefore, in this study, we elucidated and validated AS events and their regulatory mechanisms during OA progression. RESULTS: In this study, we identified differentially expressed genes between human OA and healthy meniscus samples. Among them, the OA-associated genes were primarily enriched in biological pathways such as extracellular matrix organization and ossification. The predominant OA-associated regulated AS (RAS) events were found to be involved in apoptosis during OA development. The expression of the apoptosis-related gene BCL2L13, XAF1, and NF2 were significantly different between OA and healthy meniscus samples. The construction of a covariation network of RNA-binding proteins (RBPs) and RAS genes revealed that differentially expressed RBP genes LAMA2 and CUL4B may regulate the apoptotic genes XAF1 and BCL2L13 to undergo AS events during OA progression. Finally, RT-qPCR revealed that CUL4B expression was significantly higher in OA meniscus samples than in normal controls and that the AS ratio of XAF1 was significantly different between control and OA samples; these findings were consistent with their expected expression and regulatory relationships. CONCLUSIONS: Differentially expressed RBPs may regulate the AS of apoptotic genes during knee OA progression. XAF1 and its regulator, CUL4B, may serve as novel biomarkers and potential therapeutic targets for this disease.


Sujet(s)
Gonarthrose , Humains , Gonarthrose/génétique , Gonarthrose/métabolisme , Épissage alternatif , ARN messager/génétique , Marqueurs biologiques/métabolisme , Cullines/génétique , Cullines/métabolisme
15.
Cell Rep ; 43(2): 113749, 2024 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-38329876

RÉSUMÉ

Aberrant long interspersed element 1 (LINE-1 or L1) activity can cause insertional mutagenesis and chromosomal rearrangements and has been detected in several types of cancers. Here, we show that neddylation, a post-translational modification process, is essential for L1 transposition. The antineoplastic drug MLN4924 is an L1 inhibitor that suppresses NEDD8-activating enzyme activity. Neddylation inhibition by MLN4924 selectively impairs ORF2p-mediated L1 reverse transcription and blocks the generation of L1 cDNA. Consistent with these results, MLN4924 treatment suppresses the retrotransposition activity of the non-autonomous retrotransposons short interspersed nuclear element R/variable number of tandem repeat/Alu and Alu, which rely on the reverse transcription activity of L1 ORF2p. The E2 enzyme UBE2M in the neddylation pathway, rather than UBE2F, is required for L1 ORF2p and retrotransposition. Interference with the functions of certain neddylation-dependent Cullin-really interesting new gene E3 ligases disrupts L1 reverse transcription and transposition activity. Our findings provide insights into the regulation of L1 retrotransposition and the identification of therapeutic targets for L1 dysfunctions.


Sujet(s)
Cyclopentanes , Éléments LINE , Pyrimidines , Rétroéléments , Humains , Éléments LINE/génétique , Rétroéléments/génétique , Aberrations des chromosomes , Cullines/génétique , Ubiquitin-conjugating enzymes
16.
Cancer Genomics Proteomics ; 21(2): 166-177, 2024.
Article de Anglais | MEDLINE | ID: mdl-38423594

RÉSUMÉ

BACKGROUND/AIM: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with dismal prognosis. Genomic instability due to defects in cell-cycle regulation/mitosis or deficient DNA-damage repair is a major driver of PDAC progression with clinical relevance. Deregulation of licensing of DNA replication leads to DNA damage and genomic instability, predisposing cells to malignant transformation. While overexpression of DNA replication-licensing factors has been reported in several human cancer types, their role in PDAC remains largely unknown. We aimed here to examine the expression and prognostic significance of the DNA replication-licensing factors chromatin licensing and DNA replication factor 1 (CDT1), cell-division cycle 6 (CDC6), minichromosome maintenance complex component 7 (MCM7) and also of the ubiquitin ligase regulator of CDT1, cullin 4A (CUL4A), in PDAC. MATERIALS AND METHODS: Expression levels of CUL4, CDT1, CDC6 and MCM7 were evaluated by immunohistochemistry in 76 formalin-fixed paraffin-embedded specimens of PDAC patients in relation to DNA-damage response marker H2AX, clinicopathological parameters and survival. We also conducted bioinformatics analysis of data from online available databases to corroborate our findings. RESULTS: CUL4A and DNA replication-licensing factors were overexpressed in patients with PDAC and expression of CDT1 positively correlated with H2AX. Expression of CUL4A and CDT1 positively correlated with lymph node metastasis. Importantly, elevated CUL4A expression was associated with reduced overall survival and was an independent indicator of poor prognosis on multivariate analysis. CONCLUSION: Our findings implicate CUL4A, CDT1, CDC6 and MCM7 in PDAC progression and identify CUL4A as an independent prognostic factor for this disease.


Sujet(s)
Adénocarcinome , Tumeurs du pancréas , Humains , Adénocarcinome/génétique , Ligases/génétique , Ubiquitine , Tumeurs du pancréas/génétique , Protéines du cycle cellulaire/génétique , ADN , Instabilité du génome , Cullines/génétique , Cullines/métabolisme
17.
Cancer Lett ; 587: 216731, 2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38369005

RÉSUMÉ

Therapy resistance and metastatic progression jointly determine the fatal outcome of cancer, therefore, elucidating their crosstalk may provide new opportunities to improve therapeutic efficacy and prevent recurrence and metastasis in esophageal squamous cell carcinoma (ESCC). Here, we have established radioresistant ESCC cells with the remarkable metastatic capacity, and identified miR-494-3p (miR494) as a radioresistant activator. Mechanistically, we demonstrated that cullin 3 (CUL3) is a direct target of miR494, which is transcriptionally regulated by JunD, and highlighted that JunD-miR494-CUL3 axis promotes radioresistance and metastasis by facilitating epithelial-mesenchymal transition (EMT) and restraining programmed cell death 1 ligand 1 (PD-L1) degradation. In clinical specimens, miR494 is significantly up-regulated and positively associated with T stage and lymph node metastasis in ESCC tissues and serum. Notably, patients with higher serum miR494 expression have poor prognosis, and patients with higher CUL3 expression have more conventional dendritic cells (cDCs) and plasmacytoid DCs (pDCs), less cancer-associated fibroblasts (CAF2/4), and tumor endothelial cells (TEC2/3) infiltration than patients with lower CUL3 expression, suggesting that CUL3 may be involved in tumor microenvironment (TME). Overall, miR494 may serve as a potential prognostic predictor and therapeutic target, providing a promising strategy for ESCC treatment.


Sujet(s)
Carcinome épidermoïde , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , microARN , Humains , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/radiothérapie , Antigène CD274/génétique , Antigène CD274/métabolisme , Carcinome épidermoïde/génétique , Carcinome épidermoïde/radiothérapie , Carcinome épidermoïde/métabolisme , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/radiothérapie , Tumeurs de l'oesophage/métabolisme , Cellules endothéliales/métabolisme , Pronostic , Transition épithélio-mésenchymateuse , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Mouvement cellulaire , Microenvironnement tumoral , Protéines proto-oncogènes c-jun/métabolisme , Cullines/génétique
18.
Mol Cell ; 84(7): 1304-1320.e16, 2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38382526

RÉSUMÉ

Cullin-RING ligases (CRLs) ubiquitylate specific substrates selected from other cellular proteins. Substrate discrimination and ubiquitin transferase activity were thought to be strictly separated. Substrates are recognized by substrate receptors, such as Fbox or BCbox proteins. Meanwhile, CRLs employ assorted ubiquitin-carrying enzymes (UCEs, which are a collection of E2 and ARIH-family E3s) specialized for either initial substrate ubiquitylation (priming) or forging poly-ubiquitin chains. We discovered specific human CRL-UCE pairings governing substrate priming. The results reveal pairing of CUL2-based CRLs and UBE2R-family UCEs in cells, essential for efficient PROTAC-induced neo-substrate degradation. Despite UBE2R2's intrinsic programming to catalyze poly-ubiquitylation, CUL2 employs this UCE for geometrically precise PROTAC-dependent ubiquitylation of a neo-substrate and for rapid priming of substrates recruited to diverse receptors. Cryo-EM structures illuminate how CUL2-based CRLs engage UBE2R2 to activate substrate ubiquitylation. Thus, pairing with a specific UCE overcomes E2 catalytic limitations to drive substrate ubiquitylation and targeted protein degradation.


Sujet(s)
Cullines , Ubiquitin-protein ligases , Humains , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Cullines/génétique , Cullines/métabolisme , Ubiquitination , Ubiquitine/métabolisme , Polyubiquitine/métabolisme , Protéines de transport/métabolisme
19.
Cell Death Dis ; 15(2): 121, 2024 02 08.
Article de Anglais | MEDLINE | ID: mdl-38331954

RÉSUMÉ

Mutation in CUL4B gene is one of the most common causes for X-linked intellectual disability (XLID). CUL4B is the scaffold protein in CUL4B-RING ubiquitin ligase (CRL4B) complex. While the roles of CUL4B in cancer progression and some developmental processes like adipogenesis, osteogenesis, and spermatogenesis have been studied, the mechanisms underlying the neurological disorders in patients with CUL4B mutations are poorly understood. Here, using 2D neuronal culture and cerebral organoids generated from the patient-derived induced pluripotent stem cells and their isogenic controls, we demonstrate that CUL4B is required to prevent premature cell cycle exit and precocious neuronal differentiation of neural progenitor cells. Moreover, loss-of-function mutations of CUL4B lead to increased synapse formation and enhanced neuronal excitability. Mechanistically, CRL4B complex represses transcription of PPP2R2B and PPP2R2C genes, which encode two isoforms of the regulatory subunit of protein phosphatase 2 A (PP2A) complex, through catalyzing monoubiquitination of H2AK119 in their promoter regions. CUL4B mutations result in upregulated PP2A activity, which causes inhibition of AKT and ERK, leading to premature cell cycle exit. Activation of AKT and ERK or inhibition of PP2A activity in CUL4B mutant organoids rescues the neurogenesis defect. Our work unveils an essential role of CUL4B in human cortical development.


Sujet(s)
Protein Phosphatase 2 , Protéines proto-oncogènes c-akt , Mâle , Humains , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protein Phosphatase 2/génétique , Cullines/génétique , Cullines/métabolisme , Mutation/génétique , Neurogenèse/génétique
20.
Ann Anat ; 253: 152224, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38367951

RÉSUMÉ

BACKGROUND: 3 M syndrome is first reported in 1975,which characterized by severe pre- and postnatal growth retardation, skeletal malformation and facial dysmorphism. These three genes (CUL7, OBSL1 and CCDC8) have been identified to be respond for 3 M syndrome, of which CUL7 is accounting for approximately 70%. To date, the molecular mechanism underlying the pathogenesis of 3 M syndrome remains poorly understood. Previous studies showed that no Cul7-/- mice could survive after birth, because of growth retardation at late gestational stage and respiratory distress after birth. The establishment of the animal model of cartilage specific Cul7 knockout mice (Cul7fl/fl;Col2a1-CreERT2 mice) has confirmed that Cul7fl/fl;Col2a1-CreERT2 mice can be selective in a time- and tissue-dependent manner, which can provide an experimental basis for further research on severe genetic diseases related to growth plates. OBJECTIVE: To establish a model of Cul7fl/fl;Col2a1-CreERT2 mice based on Cre/LoxP system, and to further observe its phenotype and morphological changes in growth plate. METHODS: The Cul7fl/fl;Col2a1-CreERT2 mice were taken as the experimental group, while the genotype of Cul7fl/+;Col2a1-CreERT2 mice were used as the control group. The gross morphological features and X-ray films of limbs in the two groups were observed every week for 3-6 consecutive weeks, and the length of the mice from nose to the tail, the length of femur and tibia were recorded. In the meantime, The histological morphology of tibial growth plates was compared between the two groups. RESULTS: A preliminary model of Cul7fl/fl;Col2a1-CreERT2 mice was established. The Cul7fl/fl;Col2a1-CreERT2 mice had abnormally short and deformed limbs (P<0.05), increased thickness of growth plate, the disorderly arranged chondrocyte columns, decreased number of cells in the proliferation zone, changes in the shape from flat to round, obviously expanded extracellular matrix, and disordered arrangement, thickening and loosening of bone trabecula at the proximal metaphysis of the femur. CONCLUSIONS: The knockout of Cul7 gene may affect both the proliferation of chondrocytes and the endochondral osteogenesis, confirming that Cul7 is essential for the normal development of bone in the body.


Sujet(s)
Malformations multiples , Nanisme , Lame épiphysaire , Déficience intellectuelle , Hypotonie musculaire , Rétinite pigmentaire , Rachis/malformations , Souris , Animaux , Souris knockout , Chondrocytes , Troubles de la croissance , Cullines/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE