Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 16.773
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(28): e2322972121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968116

RÉSUMÉ

Rapid accumulation of repair factors at DNA double-strand breaks (DSBs) is essential for DSB repair. Several factors involved in DSB repair have been found undergoing liquid-liquid phase separation (LLPS) at DSB sites to facilitate DNA repair. RNF168, a RING-type E3 ubiquitin ligase, catalyzes H2A.X ubiquitination for recruiting DNA repair factors. Yet, whether RNF168 undergoes LLPS at DSB sites remains unclear. Here, we identified K63-linked polyubiquitin-triggered RNF168 condensation which further promoted RNF168-mediated DSB repair. RNF168 formed liquid-like condensates upon irradiation in the nucleus while purified RNF168 protein also condensed in vitro. An intrinsically disordered region containing amino acids 460-550 was identified as the essential domain for RNF168 condensation. Interestingly, LLPS of RNF168 was significantly enhanced by K63-linked polyubiquitin chains, and LLPS largely enhanced the RNF168-mediated H2A.X ubiquitination, suggesting a positive feedback loop to facilitate RNF168 rapid accumulation and its catalytic activity. Functionally, LLPS deficiency of RNF168 resulted in delayed recruitment of 53BP1 and BRCA1 and subsequent impairment in DSB repair. Taken together, our finding demonstrates the pivotal effect of LLPS in RNF168-mediated DSB repair.


Sujet(s)
Cassures double-brin de l'ADN , Réparation de l'ADN , Protéine-1 liant le suppresseur de tumeur p53 , Ubiquitin-protein ligases , Ubiquitination , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Humains , Protéine-1 liant le suppresseur de tumeur p53/métabolisme , Protéine-1 liant le suppresseur de tumeur p53/génétique , Ubiquitine/métabolisme , Histone/métabolisme , Histone/génétique , Polyubiquitine/métabolisme
2.
Sci Rep ; 14(1): 15133, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956194

RÉSUMÉ

The goal of this study was to evaluate the intensity of autophagy and ubiquitin-dependent proteolysis processes occurring in myocardium of left ventricle (LV) in subsequent stages of pulmonary arterial hypertension (PAH) to determine mechanisms responsible for LV mass loss in a monocrotaline-induced PAH rat model. LV myocardium samples collected from 32 Wistar rats were analyzed in an early PAH group (n = 8), controls time-paired (n = 8), an end-stage PAH group (n = 8), and their controls (n = 8). Samples were subjected to histological analyses with immunofluorescence staining, autophagy assessment by western blotting, and evaluation of ubiquitin-dependent proteolysis in the LV by immunoprecipitation of ubiquitinated proteins. Echocardiographic, hemodynamic, and heart morphometric parameters were assessed regularly throughout the experiment. Considerable morphological and hemodynamic remodeling of the LV was observed over the course of PAH. The end-stage PAH was associated with significantly impaired LV systolic function and a decrease in LV mass. The LC3B-II expression in the LV was significantly higher in the end-stage PAH group compared to the early PAH group (p = 0.040). The measured LC3B-II/LC3B-I ratios in the end-stage PAH group were significantly elevated compared to the controls (p = 0.039). Immunofluorescence staining showed a significant increase in the abundance of LC3 puncta in the end-stage PAH group compared to the matched controls. There were no statistically significant differences in the levels of expression of all ubiquitinated proteins when comparing both PAH groups and matched controls. Autophagy may be considered as the mechanism behind the LV mass loss at the end stage of PAH.


Sujet(s)
Autophagie , Ventricules cardiaques , Protéolyse , Hypertension artérielle pulmonaire , Rat Wistar , Ubiquitine , Animaux , Ubiquitine/métabolisme , Ventricules cardiaques/métabolisme , Ventricules cardiaques/anatomopathologie , Ventricules cardiaques/physiopathologie , Rats , Mâle , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/anatomopathologie , Modèles animaux de maladie humaine , Myocarde/métabolisme , Myocarde/anatomopathologie , Échocardiographie , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Remodelage ventriculaire
3.
PLoS One ; 19(7): e0307213, 2024.
Article de Anglais | MEDLINE | ID: mdl-38990960

RÉSUMÉ

For the study of amyloid beta (Aß) associated toxicity which is supposed to be the main pathological agent in Alzheimer's disease (AD), it is important to secure Aß peptide with appropriate biological activity. However, commercial and synthetic Aß often have some pitfalls like less cell toxicity, prompt aggregation and excess price, using recombinant technology, these issues can be resolved though the method also suffered from some problems such as low yield, aggregation and prolong time to purify. Thus, we previously developed an easy, economic and convenient method for Aß42 purification using highly expressed GroES-Ubiquitin-Aß42 fusion protein. The method was efficient, but further development was performed to improve the procedure and increase the yield. Focus was on the isolation of the fusion protein (GroES-Ubiquitin) from Aß42 peptide. After a series of systematic testing with several chemicals, we found that methanol could precipitate efficiently the fusion protein, while the Aß peptide was recovered in the supernatant. By this method, Aß peptide was easily purified without tedious chromatographic steps which are main obstacles to purify the peptide in the previous method. This method yielded ~20 mg highly pure Aß42 peptide from 1-liter bacterial culture. Different biophysical characterizations and bioactivity assays indicate that the peptide purified using this method was competitive with others which have been previously reported whereas considering the simplicity, final yield and time of purification, this method is the optimal solution.


Sujet(s)
Peptides bêta-amyloïdes , Fragments peptidiques , Protéines de fusion recombinantes , Peptides bêta-amyloïdes/isolement et purification , Peptides bêta-amyloïdes/composition chimique , Peptides bêta-amyloïdes/métabolisme , Fragments peptidiques/composition chimique , Fragments peptidiques/isolement et purification , Humains , Protéines de fusion recombinantes/isolement et purification , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Ubiquitine/composition chimique , Ubiquitine/métabolisme , Ubiquitine/isolement et purification , Escherichia coli/génétique , Escherichia coli/métabolisme , Maladie d'Alzheimer/métabolisme
4.
J Chem Phys ; 161(3)2024 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-39007394

RÉSUMÉ

The unfolding dynamics of ubiquitin were studied using a combination of x-ray solution scattering (XSS) and molecular dynamics (MD) simulations. The kinetic analysis of the XSS ubiquitin signals showed that the protein unfolds through a two-state process, independent of the presence of destabilizing salts. In order to characterize the ensemble of unfolded states in atomic detail, the experimental XSS results were used as a constraint in the MD simulations through the incorporation of x-ray scattering derived potential to drive the folded ubiquitin structure toward sampling unfolded states consistent with the XSS signals. We detail how biased MD simulations provide insight into unfolded states that are otherwise difficult to resolve and underscore how experimental XSS data can be combined with MD to efficiently sample structures away from the native state. Our results indicate that ubiquitin samples unfolded in states with a high degree of loss in secondary structure yet without a collapse to a molten globule or fully solvated extended chain. Finally, we propose how using biased-MD can significantly decrease the computational time and resources required to sample experimentally relevant nonequilibrium states.


Sujet(s)
Simulation de dynamique moléculaire , Dépliement des protéines , Ubiquitine , Ubiquitine/composition chimique , Diffraction des rayons X , Cinétique
5.
Biol Direct ; 19(1): 55, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38978100

RÉSUMÉ

Ubiquitinylation of protein substrates results in various but distinct biological consequences, among which ubiquitin-mediated degradation is most well studied for its therapeutic application. Accordingly, artificially targeted ubiquitin-dependent degradation of various proteins has evolved into the therapeutically relevant PROTAC technology. This tethered ubiquitinylation of various targets coupled with a broad assortment of modifying E3 ubiquitin ligases has been made possible by rational design of bi-specific chimeric molecules that bring these proteins in proximity. However, forced ubiquitinylation inflicted by the binary warheads of a chimeric PROTAC molecule should not necessarily result in protein degradation but can be used to modulate other cellular functions. In this respect it should be noted that the ubiquitinylation of a diverse set of proteins is known to control their transport, transcriptional activity, and protein-protein interactions. This review provides examples of potential PROTAC usage based on non-degradable ubiquitinylation.


Sujet(s)
Protéolyse , Ubiquitin-protein ligases , Ubiquitine , Ubiquitination , Ubiquitine/métabolisme , Ubiquitin-protein ligases/métabolisme , Humains
6.
J Environ Pathol Toxicol Oncol ; 43(4): 13-23, 2024.
Article de Anglais | MEDLINE | ID: mdl-39016138

RÉSUMÉ

The early diagnostic methods for non-small-cell lung cancer (NSCLC) are limited, lacking effective biomarkers, and the late stage surgery is difficult and has a high recurrence rate. We investigated whether the effects of FBXO45 in arcinogenesis and metastasis of NSCLC. The up-regulation of FBXO45 expression in NSCLC patients or cell lines were observed. FBXO45 gene promoted metastasis and Warburg effect, and reduced ferroptosis of NSCLC. FBXO45 induced ZEB1 expression to promote Warburg effect and reduced ferroptosis of NSCLC. Sh-FBXO45 reduced cancer growth of NSCLC in mice model. FBXO45 decreased the ubiquitination of ZEB1, leading to increased expression of ZEB1, which in turn promoted the Warburg effect and reduced ferroptosis in NSCLC. In vivo imaging, Sh-FBXO45 also reduced ZEB1 expression levels of lung tissue in mice model. FBXO45 in NSCLC through activating the Warburg effect, and the inhibition of ferroptosis of NSCLC by the suppression of ZEB1 ubiquitin, FBXO45 may be a potential therapeutic strategy for NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Protéines F-box , Tumeurs du poumon , Facteur de transcription Zeb1 , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Facteur de transcription Zeb1/génétique , Facteur de transcription Zeb1/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Humains , Animaux , Souris , Protéines F-box/génétique , Protéines F-box/métabolisme , Lignée cellulaire tumorale , Ferroptose/génétique , Mâle , Ubiquitine/métabolisme
7.
Nat Commun ; 15(1): 5953, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009586

RÉSUMÉ

The intracellular bacterial pathogen Legionella pneumophila modulates host cell functions by secreting multiple effectors with diverse biochemical activities. In particular, effectors of the SidE family interfere with host protein ubiquitination in a process that involves production of phosphoribosyl ubiquitin (PR-Ub). Here, we show that effector LnaB converts PR-Ub into ADP-ribosylated ubiquitin, which is further processed to ADP-ribose and functional ubiquitin by the (ADP-ribosyl)hydrolase MavL, thus maintaining ubiquitin homeostasis in infected cells. Upon being activated by actin, LnaB also undergoes self-AMPylation on tyrosine residues. The activity of LnaB requires a motif consisting of Ser, His and Glu (SHxxxE) present in a large family of toxins from diverse bacterial pathogens. Thus, our study sheds light on the mechanisms by which a pathogen maintains ubiquitin homeostasis and identifies a family of enzymes capable of protein AMPylation.


Sujet(s)
Protéines bactériennes , Homéostasie , Legionella pneumophila , Ubiquitine , Ubiquitination , Ubiquitine/métabolisme , Legionella pneumophila/métabolisme , Legionella pneumophila/pathogénicité , Humains , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , ADP-Ribosylation , Interactions hôte-pathogène , Adénosine diphosphate ribose/métabolisme , Maladie des légionnaires/métabolisme , Maladie des légionnaires/microbiologie , Cellules HEK293 , Actines/métabolisme , Cellules HeLa
8.
Bioconjug Chem ; 35(7): 944-953, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38954775

RÉSUMÉ

The chemical synthesis of homogeneously ubiquitylated histones is a powerful approach to decipher histone ubiquitylation-dependent epigenetic regulation. Among the various methods, α-halogen ketone-mediated conjugation chemistry has recently been an attractive strategy to generate single-monoubiquitylated histones for biochemical and structural studies. Herein, we report the use of this strategy to prepare not only dual- and even triple-monoubiquitylated histones but also diubiquitin-modified histones. We were surprised to find that the synthetic efficiencies of multi-monoubiquitylated histones were comparable to those of single-monoubiquitylated ones, suggesting that this strategy is highly tolerant to the number of ubiquitin monomers installed onto histones. The facile generation of a series of single-, dual-, and triple-monoubiquitylated H3 proteins enabled us to evaluate the influence of ubiquitylation patterns on the binding of DNA methyltransferase 1 (DNMT1) to nucleosomes. Our study highlights the potential of site-specific conjugation chemistry to generate chemically defined histones for epigenetic studies.


Sujet(s)
Histone , Cétones , Ubiquitination , Histone/composition chimique , Histone/métabolisme , Histone/synthèse chimique , Cétones/composition chimique , Ubiquitine/composition chimique , Humains , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/composition chimique , Nucléosomes/composition chimique , Nucléosomes/métabolisme
9.
Mol Cell ; 84(11): 2011-2013, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38848689

RÉSUMÉ

In this issue of Molecular Cell, Yi et al.1 demonstrate that reduced mTORC1 activity induces the CTLH E3 ligase-dependent degradation of HMGCS1, an enzyme in the mevalonate pathway, thus revealing a unique connection between mTORC1 signaling and the degradation of a specific metabolic enzyme via the ubiquitin-proteasome system.


Sujet(s)
Complexe-1 cible mécanistique de la rapamycine , Proteasome endopeptidase complex , Transduction du signal , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/génétique , Humains , Proteasome endopeptidase complex/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéolyse , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Complexes multiprotéiques/métabolisme , Complexes multiprotéiques/génétique , Animaux , Acide mévalonique/métabolisme , Ubiquitine/métabolisme
10.
Sci Adv ; 10(24): eadm8449, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38865459

RÉSUMÉ

The accumulation of protein aggregates is a hallmark of many diseases, including Alzheimer's disease. As a major pillar of the proteostasis network, autophagy mediates the degradation of protein aggregates. The autophagy cargo receptor p62 recognizes ubiquitin on proteins and cooperates with TAX1BP1 to recruit the autophagy machinery. Paradoxically, protein aggregates are not degraded in various diseases despite p62 association. Here, we reconstituted the recognition by the autophagy receptors of physiological and pathological Tau forms. Monomeric Tau recruits p62 and TAX1BP1 via the sequential actions of the chaperone and ubiquitylation machineries. In contrast, Tau fibrils from Alzheimer's disease brains are recognized by p62 but fail to recruit TAX1BP1. This failure is due to the masking of fibrils ubiquitin moieties by p62. Tau fibrils are resistant to deubiquitylation, and, thus, this nonproductive interaction of p62 with the fibrils is irreversible. Our results shed light on the mechanism underlying autophagy evasion by protein aggregates and their consequent accumulation in disease.


Sujet(s)
Autophagie , Séquestosome-1 , Ubiquitination , Protéines tau , Humains , Protéines tau/métabolisme , Protéines tau/composition chimique , Séquestosome-1/métabolisme , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Liaison aux protéines , Agrégats de protéines , Protéines et peptides de signalisation intracellulaire/métabolisme , Ubiquitine/métabolisme , Protéines tumorales
11.
Sci Rep ; 14(1): 13037, 2024 06 06.
Article de Anglais | MEDLINE | ID: mdl-38844605

RÉSUMÉ

The proteasome-associated deubiquitinase USP14 is a potential drug target. Using an inducible USP14 knockout system in colon cancer cells, we found that USP14 depletion impedes cellular proliferation, induces cell cycle arrest, and leads to a senescence-like phenotype. Transcriptomic analysis revealed altered gene expression related to cell division and cellular differentiation. USP14 knockout cells also exhibited changes in morphology, actin distribution, and expression of actin cytoskeletal components. Increased ubiquitin turnover was observed, offset by upregulation of polyubiquitin genes UBB and UBC. Pharmacological inhibition of USP14 with IU1 increased ubiquitin turnover but did not affect cellular growth or morphology. BioGRID data identified USP14 interactors linked to actin cytoskeleton remodeling, DNA damage repair, mRNA splicing, and translation. In conclusion, USP14 loss in colon cancer cells induces a transient quiescent cancer phenotype not replicated by pharmacologic inhibition of its deubiquitinating activity.


Sujet(s)
Prolifération cellulaire , Vieillissement de la cellule , Tumeurs colorectales , Ubiquitin thiolesterase , Humains , Vieillissement de la cellule/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Lignée cellulaire tumorale , Phénotype , Proteasome endopeptidase complex/métabolisme , Points de contrôle du cycle cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Ubiquitine/métabolisme
12.
J Orthop Surg Res ; 19(1): 356, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879525

RÉSUMÉ

BACKGROUND: Ubiquitin/ubiquitin-like (Ub/UBL)-related genes have been reported to be associated with the survival of osteosarcoma patients but have not yet been systematically explored. METHODS: The prognostic value of Ub/UBL-related genes, immune cell infiltration and clinicopathological features of patients were explored by Cox and LASSO regression analyses. A prognostic model was established and then validated in the GSE21257 dataset. The differential expression of hub genes in osteosarcoma was confirmed by qRT-PCR, western blotting and immunohistochemistry. RESULTS: Tripartite Motif Containing 8 (TRIM8) and Ubiquitin Like With PHD And Ring Finger Domains 2 (UHRF2) were screened as genes with prognostic value in osteosarcoma. Kaplan-Meier analysis and scatter plots indicated that patients in the high gene significance score group tended to have a worse prognosis. The concordance index, calibration analysis and receiver operating characteristic analysis suggested that the model had good prediction accuracy and high sensitivity and specificity. Decision curve analysis revealed that patients could obtain greater net benefit from this model. Functional analyses of the differentially expressed genes indicated that they were involved in important functions and pathways. TRIM8 and UHRF2 were confirmed to be highly expressed in osteosarcoma cell lines and tissues. CONCLUSIONS: TRIM8 and UHRF2 are potential prognostic genes in osteosarcoma, and these results provide insights into the roles of these genes and their implications for patient outcomes.


Sujet(s)
Tumeurs osseuses , Ostéosarcome , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/immunologie , Ostéosarcome/mortalité , Humains , Pronostic , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/immunologie , Tumeurs osseuses/mortalité , Mâle , Femelle , Marqueurs biologiques tumoraux/génétique , Ubiquitin-protein ligases/génétique , Ubiquitine/génétique
13.
Phys Chem Chem Phys ; 26(26): 18244-18255, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38904333

RÉSUMÉ

Natural deep eutectic solvents (NADESs) comprised of osmolytes are of interest as potential biomolecular (cryo)protectants. However, the way these solvents influence the structure and dynamics of biomolecules as well as the role of water remains poorly understood. We carried out principal component analysis of various secondary structure elements of ubiquitin in water and a betaine : glycerol : water (1 : 2 : ζ; ζ = 0, 1, 2, 5, 10, 20, 45) NADES, from molecular dynamics trajectories, to gain insight into the protein dynamics as it undergoes a transition from a highly viscous anhydrous to an aqueous environment. A crossover of the protein's essential dynamics at ζ ∼ 5, induced by solvent-shell coupled fluctuations, is observed, indicating that ubiquitin might (re)fold in the NADES upon water addition at ζ > ∼5. Further, in contrast to water, the anhydrous NADES preserves ubiquitin's essential modes at high temperatures explaining the protein's seemingly enhanced thermal stability.


Sujet(s)
Simulation de dynamique moléculaire , Solvants , Ubiquitine , Eau , Ubiquitine/composition chimique , Eau/composition chimique , Solvants/composition chimique , Glycérol/composition chimique , Bétaïne/composition chimique , Analyse en composantes principales , Structure secondaire des protéines
14.
Adv Virus Res ; 119: 1-38, 2024.
Article de Anglais | MEDLINE | ID: mdl-38897707

RÉSUMÉ

The ubiquitination process is a reversible posttranslational modification involved in many essential cellular functions, such as innate immunity, cell signaling, trafficking, protein stability, and protein degradation. Viruses can use the ubiquitin system to efficiently enter host cells, replicate and evade host immunity, ultimately enhancing viral pathogenesis. Emerging evidence indicates that enveloped viruses can carry free (unanchored) ubiquitin or covalently ubiquitinated viral structural proteins that can increase the efficiency of viral entry into host cells. Furthermore, viruses continuously evolve and adapt to take advantage of the host ubiquitin machinery, highlighting its importance during virus infection. This review discusses the battle between viruses and hosts, focusing on how viruses hijack the ubiquitination process at different steps of the replication cycle, with a specific emphasis on viral entry. We discuss how ubiquitination of viral proteins may affect tropism and explore emerging therapeutics strategies targeting the ubiquitin system for antiviral drug discovery.


Sujet(s)
Ubiquitination , Pénétration virale , Réplication virale , Humains , Ubiquitine/métabolisme , Virus/métabolisme , Interactions hôte-pathogène , Protéines virales/métabolisme , Protéines virales/génétique , Maladies virales/virologie , Maladies virales/immunologie , Maladies virales/métabolisme , Animaux , Maturation post-traductionnelle des protéines
15.
Int J Mol Med ; 54(2)2024 08.
Article de Anglais | MEDLINE | ID: mdl-38940355

RÉSUMÉ

The ubiquitin (Ub)­proteasome system (UPS) plays a pivotal role in maintaining protein homeostasis and function to modulate various cellular processes including skeletal cell differentiation and bone homeostasis. The Ub ligase E3 promotes the transfer of Ub to the target protein, especially transcription factors, to regulate the proliferation, differentiation and survival of bone cells, as well as bone formation. In turn, the deubiquitinating enzyme removes Ub from modified substrate proteins to orchestrate bone remodeling. As a result of abnormal regulation of ubiquitination, bone cell differentiation exhibits disorder and then bone homeostasis is affected, consequently leading to osteoporosis. The present review discussed the role and mechanism of UPS in bone remodeling. However, the specific mechanism of UPS in the process of bone remodeling is still not fully understood and further research is required. The study of the mechanism of action of UPS can provide new ideas and methods for the prevention and treatment of osteoporosis. In addition, the most commonly used osteoporosis drugs that target ubiquitination processes in the clinic are discussed in the current review.


Sujet(s)
Ostéoporose , Ubiquitine , Ubiquitination , Humains , Ostéoporose/métabolisme , Ostéoporose/anatomopathologie , Animaux , Ubiquitine/métabolisme , Proteasome endopeptidase complex/métabolisme , Remodelage osseux , Ubiquitin-protein ligases/métabolisme
16.
Biochemistry ; 63(14): 1723-1729, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38941592

RÉSUMÉ

Protein advanced glycation end products (AGEs) can be formed via nonenzymatic glycation and accumulated intracellularly to disrupt cellular homeostasis for protein clearance. Here, we investigated the formation particulars of intracellular protein AGEs and sought to elucidate the molecular events implicated in the impact of cellular clearance systems. The formation and accumulation of intracellular protein AGEs increased protein aggregation and protease resistance, potentially overwhelming the ubiquitin-proteasome system (UPS). At high levels of protein AGEs, the abundance of many E3 ligases decreased and the overall ubiquitination level was reduced, all of which indicated decreased UPS activity. On the other hand, autophagy activity was stimulated, as evidenced by the upregulation of autophagy marker LC3II and important proteins in autophagosome and autolysosome formation, as well as downregulation of mTOR. Understanding the functional impacts of intracellular protein AGEs on the UPS and autophagy could pave the way for the future development of pharmaceutical agents targeting AGE-related diseases.


Sujet(s)
Autophagie , Produits terminaux de glycation avancée , Homéostasie , Produits terminaux de glycation avancée/métabolisme , Humains , Autophagie/physiologie , Cellules épithéliales/métabolisme , Proteasome endopeptidase complex/métabolisme , Ubiquitination , Ubiquitine/métabolisme , Ubiquitin-protein ligases/métabolisme , Animaux
17.
Cells ; 13(11)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38891087

RÉSUMÉ

Ubiquitin-specific protease 14 (USP14), one of the three major proteasome-associated deubiquitinating enzymes (DUBs), is known to be activated by the AKT-mediated phosphorylation at Ser432. Thereby, AKT can regulate global protein degradation by controlling the ubiquitin-proteasome system (UPS). However, the exact molecular mechanism of USP14 activation by AKT phosphorylation at the atomic level remains unknown. By performing the molecular dynamics (MD) simulation of the USP14 catalytic domain at three different states (inactive, active, and USP14-ubiquitin complex), we characterized the change in structural dynamics by phosphorylation. We observed that the Ser432 phosphorylation induced substantial conformational changes of USP14 in the blocking loop (BL) region to fold it from an open loop into a ß-sheet, which is critical for USP14 activation. Furthermore, phosphorylation also increased the frequency of critical hydrogen bonding and salt bridge interactions between USP14 and ubiquitin, which is essential for DUB activity. Structural dynamics insights from this study pinpoint the important local conformational landscape of USP14 by the phosphorylation event, which would be critical for understanding USP14-mediated proteasome regulation and designing future therapeutics.


Sujet(s)
Simulation de dynamique moléculaire , Protéines proto-oncogènes c-akt , Ubiquitin thiolesterase , Phosphorylation , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/composition chimique , Protéines proto-oncogènes c-akt/métabolisme , Humains , Ubiquitine/métabolisme , Activation enzymatique , Domaine catalytique , Liaison aux protéines , Conformation des protéines
18.
Cell Death Dis ; 15(6): 436, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902268

RÉSUMÉ

Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide, necessitating the identification of novel therapeutic targets. Lysosome Associated Protein Transmembrane 4B (LAPTM4B) is involved in biological processes critical to cancer progression, such as regulation of solute carrier transporter proteins and metabolic pathways, including mTORC1. However, the metabolic processes governed by LAPTM4B and its role in oncogenesis remain unknown. In this study, we conducted unbiased metabolomic screens to uncover the metabolic landscape regulated by LAPTM4B. We observed common metabolic changes in several knockout cell models suggesting of a role for LAPTM4B in suppressing ferroptosis. Through a series of cell-based assays and animal experiments, we demonstrate that LAPTM4B protects tumor cells from erastin-induced ferroptosis both in vitro and in vivo. Mechanistically, LAPTM4B suppresses ferroptosis by inhibiting NEDD4L/ZRANB1 mediated ubiquitination and subsequent proteasomal degradation of the cystine-glutamate antiporter SLC7A11. Furthermore, metabolomic profiling of cancer cells revealed that LAPTM4B knockout leads to a significant enrichment of ferroptosis and associated metabolic alterations. By integrating results from cellular assays, patient tissue samples, an animal model, and cancer databases, this study highlights the clinical relevance of the LAPTM4B-SLC7A11-ferroptosis signaling axis in NSCLC progression and identifies it as a potential target for the development of cancer therapeutics.


Sujet(s)
Système y+ de transport d'acides aminés , Carcinome pulmonaire non à petites cellules , Ferroptose , Tumeurs du poumon , Proteasome endopeptidase complex , Ubiquitine , Ferroptose/effets des médicaments et des substances chimiques , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Animaux , Proteasome endopeptidase complex/métabolisme , Ubiquitine/métabolisme , Souris , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Protéines oncogènes/métabolisme , Protéines oncogènes/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Lignée cellulaire tumorale , Ubiquitination , Souris nude , Protéolyse/effets des médicaments et des substances chimiques
19.
Cell Mol Life Sci ; 81(1): 271, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38888668

RÉSUMÉ

Cystic Fibrosis (CF) is a genetic disease caused by mutations in CFTR gene expressing the anion selective channel CFTR located at the plasma membrane of different epithelial cells. The most commonly investigated variant causing CF is F508del. This mutation leads to structural defects in the CFTR protein, which are recognized by the endoplasmic reticulum (ER) quality control system. As a result, the protein is retained in the ER and degraded via the ubiquitin-proteasome pathway. Although blocking ubiquitination to stabilize the CFTR protein has long been considered a potential pharmacological approach in CF, progress in this area has been relatively slow. Currently, no compounds targeting this pathway have entered clinical trials for CF. On the other hand, the emergence of Orkambi initially, and notably the subsequent introduction of Trikafta/Kaftrio, have demonstrated the effectiveness of molecular chaperone-based therapies for patients carrying the F508del variant and even showed efficacy against other variants. These treatments directly target the CFTR variant protein without interfering with cell signaling pathways. This review discusses the limits and potential future of targeting protein ubiquitination in CF.


Sujet(s)
Protéine CFTR , Mucoviscidose , Ubiquitination , Mucoviscidose/métabolisme , Mucoviscidose/génétique , Mucoviscidose/traitement médicamenteux , Mucoviscidose/anatomopathologie , Humains , Protéine CFTR/métabolisme , Protéine CFTR/génétique , Réticulum endoplasmique/métabolisme , Animaux , Mutation , Ubiquitine/métabolisme
20.
Anal Chem ; 96(21): 8349-8355, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38745349

RÉSUMÉ

In contrast to intracellular gene transfer, the direct delivery of expressed proteins is a significantly challenging yet essential technique for elucidating cellular functions, including protein complex structure, liquid-liquid phase separation, therapeutic applications, and reprogramming. In this study, we developed a hybrid nanotube (HyNT) stamp system that physically inserts the HyNTs into adhesive cells, enabling the injection of target molecules through HyNT ducts. This system demonstrates the capability to deliver multiple proteins, such as lactate oxidase (LOx) and ubiquitin (UQ), to approximately 1.8 × 107 adhesive cells with a delivery efficiency of 89.9% and a viability of 97.1%. The delivery of LOx enzyme into HeLa cancer cells induced cell death, while enzyme-delivered healthy cells remained viable. Furthermore, our stamp system can deliver an isotope-labeled UQ into adhesive cells for detection by nuclear magnetic resonance (NMR).


Sujet(s)
Nanotubes , Ubiquitine , Humains , Cellules HeLa , Nanotubes/composition chimique , Ubiquitine/métabolisme , Ubiquitine/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Spectroscopie par résonance magnétique , Résonance magnétique nucléaire biomoléculaire , Mixed function oxygenases
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...