Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.787
Filtrer
1.
Mol Cancer ; 23(1): 148, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39048965

RÉSUMÉ

Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin-proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.


Sujet(s)
Enzymes de désubiquitinylation , Tumeurs , Proteasome endopeptidase complex , Ubiquitination , Humains , Tumeurs/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Animaux , Proteasome endopeptidase complex/métabolisme , Enzymes de désubiquitinylation/métabolisme , Protéolyse , Ubiquitine/métabolisme , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Maturation post-traductionnelle des protéines , Thérapie moléculaire ciblée , Ubiquitin-protein ligases/métabolisme
2.
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
3.
Clin Transl Med ; 14(7): e1769, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39021054

RÉSUMÉ

Recently, there is a rise in studies that recognize the importance of targeting ubiquitin and related molecular machinery in various therapeutic contexts. Here we briefly discuss the history of ubiquitin, its biological roles in protein degradation and beyond, as well as the current state of ubiquitin-targeting therapeutics across diseases. We conclude that targeting ubiquitin machinery is approaching a renaissance, and tapping its full potential will require embracing a wholistic perspective of ubiquitin's multifaceted roles.


Sujet(s)
Proteasome endopeptidase complex , Ubiquitine , Ubiquitine/métabolisme , Humains , Proteasome endopeptidase complex/métabolisme , Protéolyse
4.
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
5.
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
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.
Int J Mol Sci ; 25(14)2024 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-39062952

RÉSUMÉ

Gastrodin (GAS) is the main chemical component of the traditional Chinese herb Gastrodia elata (called "Tianma" in Chinese), which has been used to treat neurological conditions, including headaches, epilepsy, stroke, and memory loss. To our knowledge, it is unclear whether GAS has a therapeutic effect on Huntington's disease (HD). In the present study, we evaluated the effect of GAS on the degradation of mutant huntingtin protein (mHtt) by using PC12 cells transfected with N-terminal mHtt Q74. We found that 0.1-100 µM GAS had no effect on the survival rate of Q23 and Q74 PC12 cells after 24-48 h of incubation. The ubiquitin-proteasome system (UPS) is the main system that clears misfolded proteins in eukaryotic cells. Mutated Htt significantly upregulated total ubiquitinated protein (Ub) expression, decreased chymotrypsin-like, trypsin-like and caspase-like peptidase activity, and reduced the colocalization of the 20S proteasome with mHtt. GAS (25 µM) attenuated all of the abovementioned pathological changes, and the regulatory effect of GAS on mHtt was found to be abolished by MG132, a proteasome inhibitor. The autophagy-lysosome pathway (ALP) is another system for misfolded protein degradation. Although GAS downregulated the expression of autophagy markers (LC3II and P62), it increased the colocalization of LC3II with lysosomal associated membrane protein 1 (LAMP1), which indicates that ALP was activated. Moreover, GAS prevented mHtt-induced neuronal damage in PC12 cells. GAS has a selective effect on mHtt in Q74 PC12 cells and has no effect on Q23 and proteins encoded by other genes containing long CAGs, such as Rbm33 (10 CAG repeats) and Hcn1 (>30 CAG repeats). Furthermore, oral administration of 100 mg/kg GAS increased grip strength and attenuated mHtt aggregates in B6-hHTT130-N transgenic mice. This is a high dose (100 mg/kg GAS) when compared with experiments on HD mice with other small molecules. We will design more doses to evaluate the dose-response relationship of the inhibition effect of GAS on mHtt in our next study. In summary, GAS can promote the degradation of mHtt by activating the UPS and ALP, making it a potential therapeutic agent for HD.


Sujet(s)
Autophagie , Alcools benzyliques , Glucosides , Protéine huntingtine , Lysosomes , Proteasome endopeptidase complex , Ubiquitine , Animaux , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Rats , Proteasome endopeptidase complex/métabolisme , Cellules PC12 , Autophagie/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Ubiquitine/métabolisme , Alcools benzyliques/pharmacologie , Glucosides/pharmacologie , Souris , Maladie de Huntington/métabolisme , Maladie de Huntington/traitement médicamenteux , Maladie de Huntington/génétique , Protéolyse/effets des médicaments et des substances chimiques , Mutation
8.
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
9.
Proc Natl Acad Sci U S A ; 121(32): e2319091121, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39074279

RÉSUMÉ

Understanding the normal function of the Huntingtin (HTT) protein is of significance in the design and implementation of therapeutic strategies for Huntington's disease (HD). Expansion of the CAG repeat in the HTT gene, encoding an expanded polyglutamine (polyQ) repeat within the HTT protein, causes HD and may compromise HTT's normal activity contributing to HD pathology. Here, we investigated the previously defined role of HTT in autophagy specifically through studying HTT's association with ubiquitin. We find that HTT interacts directly with ubiquitin in vitro. Tandem affinity purification was used to identify ubiquitinated and ubiquitin-associated proteins that copurify with a HTT N-terminal fragment under basal conditions. Copurification is enhanced by HTT polyQ expansion and reduced by mimicking HTT serine 421 phosphorylation. The identified HTT-interacting proteins include RNA-binding proteins (RBPs) involved in mRNA translation, proteins enriched in stress granules, the nuclear proteome, the defective ribosomal products (DRiPs) proteome and the brain-derived autophagosomal proteome. To determine whether the proteins interacting with HTT are autophagic targets, HTT knockout (KO) cells and immunoprecipitation of lysosomes were used to investigate autophagy in the absence of HTT. HTT KO was associated with reduced abundance of mitochondrial proteins in the lysosome, indicating a potential compromise in basal mitophagy, and increased lysosomal abundance of RBPs which may result from compensatory up-regulation of starvation-induced macroautophagy. We suggest HTT is critical for appropriate basal clearance of mitochondrial proteins and RBPs, hence reduced HTT proteostatic function with mutation may contribute to the neuropathology of HD.


Sujet(s)
Protéine huntingtine , Lysosomes , Mitochondries , Protéines de liaison à l'ARN , Ubiquitine , Protéine huntingtine/métabolisme , Protéine huntingtine/génétique , Lysosomes/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Humains , Ubiquitine/métabolisme , Mitochondries/métabolisme , Autophagie , Animaux , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Souris , Liaison aux protéines , Maladie de Huntington/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/anatomopathologie , Peptides/métabolisme
10.
Cell Death Dis ; 15(7): 538, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075053

RÉSUMÉ

Glioma, a malignant and infiltrative neoplasm of the central nervous system, poses a significant threat due to its high mortality rates. Branched-chain amino acid transaminase 1 (BCAT1), a key enzyme in branched-chain amino acid (BCAA) catabolism, exhibits elevated expression in gliomas and correlates strongly with poor prognosis. Nonetheless, the regulatory mechanisms underlying this increased BCAT1 expression remains incompletely understood. In this study, we reveal that ubiquitination at Lys360 facilitates BCAT1 degradation, with low ubiquitination levels contributing to high BCAT1 expression in glioma cells. The Carboxyl terminus of Hsc70-interacting protein (CHIP), an E3 ubiquitin ligase, interacts with BCAT1 via its coiled-coil (CC) domain, promoting its K48-linkage ubiquitin degradation through proteasomal pathway. Moreover, CHIP-mediated BCAT1 degradation induces metabolic reprogramming, and impedes glioma cell proliferation and tumor growth both in vitro and in vivo. Furthermore, a positive correlation is observed between low CHIP expression, elevated BCAT1 levels, and unfavorable prognosis among glioma patients. Additionally, we show that the CHIP/BCAT1 axis enhances glioma sensitivity to temozolomide by reducing glutathione (GSH) synthesis and increasing oxidative stress. These findings underscore the critical role of CHIP/BCAT1 axis in glioma cell proliferation and temozolomide sensitivity, highlighting its potential as a diagnostic marker and therapeutic target in glioma treatment.


Sujet(s)
Prolifération cellulaire , Gliome , Témozolomide , Transaminases , Ubiquitin-protein ligases , Ubiquitination , Humains , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Gliome/métabolisme , Gliome/anatomopathologie , Gliome/génétique , Gliome/traitement médicamenteux , Animaux , Lignée cellulaire tumorale , Transaminases/métabolisme , Transaminases/génétique , Souris , Souris nude , Ubiquitine/métabolisme , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Protéolyse/effets des médicaments et des substances chimiques , Mâle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Femelle
11.
Front Biosci (Landmark Ed) ; 29(7): 260, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39082341

RÉSUMÉ

Retinal degeneration (RD) is a group of chronic blinding diseases characterised by progressive retinal cell death. As the disease progresses, vision deteriorates due to retinal cell death and impaired retinal integrity, eventually leading to complete loss of vision. Therefore, the function and environmental homeostasis of the retina have an important impact on the pathogenesis and treatment of RD. Ubiquitination, as a complex post-translational modification process, plays an essential role in maintaining retinal homeostasis and normal function. It covalently combines ubiquitin with protein through a series of enzyme-mediated reactions, and participates in cell processes such as gene transcription, cell cycle process, DNA repair, apoptosis and immune response. At the same time, it plays a central role in protein degradation. There are two major protein degradation systems in eukaryotic cells: the ubiquitin-proteasome system and the autophagy-lysosomal system. The protein degradation pathway maintains retinal protein homeostasis by reducing abnormal protein accumulation in the retina through two modes of degradation. Either dysregulation of ubiquitination or disruption of protein homeostasis may lead to the development of RD. This article aims to comprehensively review recent research progress on ubiquitin-related genes, proteins and protein homeostasis in the pathogenesis of RD, and to summarize the potential targeted therapy strategies for it. The review is expected to provide valuable guidance for further development and application of ubiquitination in RD.


Sujet(s)
Homéostasie protéique , Dégénérescence de la rétine , Ubiquitination , Humains , Dégénérescence de la rétine/métabolisme , Animaux , Proteasome endopeptidase complex/métabolisme , Ubiquitine/métabolisme , Rétine/métabolisme , Rétine/anatomopathologie , Autophagie , Protéolyse
12.
Acta Trop ; 257: 107283, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38955322

RÉSUMÉ

Toxoplasmosis, a zoonotic parasitic disease caused by Toxoplasma gondii (T. gondii), is prevalent worldwide. The fact should be emphasized that a considerable proportion of individuals infected with T. gondii may remain asymptomatic; nevertheless, the condition can have severe implications for pregnant women or immunocompromised individuals. The current treatment of toxoplasmosis primarily relies on medication; however, traditional anti-toxoplasmosis drugs exhibit significant limitations in terms of efficacy, side effects, and drug resistance. The life cycles of T. gondii are characterized by distinct stages and its body morphology goes through dynamic alterations during the growth cycle that are intricately governed by a wide array of post-translational modifications (PTMs). Ubiquitin (Ub) signaling and ubiquitin-like (Ubl) signaling are two crucial post-translational modification pathways within cells, regulating protein function, localization, stability, or interactions by attaching Ub or ubiquitin-like proteins (Ubls) to target proteins. While these signaling mechanisms share some functional similarities, they have distinct regulatory mechanisms and effects. T. gondii possesses both Ub and Ubls and plays a significant role in regulating the parasite's life cycle and maintaining its morphology through PTMs of substrate proteins. Investigating the role and mechanism of protein ubiquitination in T. gondii will provide valuable insights for preventing and treating toxoplasmosis. This review explores the distinctive characteristics of Ub and Ubl signaling in T. gondii, with the aim of inspiring research ideas for the identification of safer and more effective drug targets against toxoplasmosis.


Sujet(s)
Transduction du signal , Toxoplasma , Toxoplasmose , Ubiquitine , Toxoplasma/métabolisme , Toxoplasma/physiologie , Toxoplasma/effets des médicaments et des substances chimiques , Ubiquitine/métabolisme , Humains , Toxoplasmose/parasitologie , Toxoplasmose/traitement médicamenteux , Toxoplasmose/métabolisme , Animaux , Protéines de protozoaire/métabolisme , Ubiquitination , Maturation post-traductionnelle des protéines , Ubiquitines/métabolisme , Étapes du cycle de vie
13.
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)
Réparation de l'ADN , Ubiquitin-protein ligases , Humains , Cassures double-brin de l'ADN , Histone/métabolisme , Histone/génétique , Polyubiquitine/métabolisme , Protéine-1 liant le suppresseur de tumeur p53/métabolisme , Protéine-1 liant le suppresseur de tumeur p53/génétique , Ubiquitine/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitination
14.
Molecules ; 29(14)2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39064934

RÉSUMÉ

Protein homeostasis is a tightly conserved process that is regulated through the ubiquitin proteasome system (UPS) in a ubiquitin-independent or ubiquitin-dependent manner. Over the past two decades, the proteasome has become an excellent therapeutic target through inhibition of the catalytic core particle, inhibition of subunits responsible for recognizing and binding ubiquitinated proteins, and more recently, through targeted protein degradation using proteolysis targeting chimeras (PROTACs). The majority of the developed inhibitors of the proteasome's core particle rely on gaining selectivity through binding interactions within the unprimed substrate channel. Although this has allowed for selective inhibitors and chemical probes to be generated for the different proteasome isoforms, much remains unknown about the interactions that could be harnessed within the primed substrate channel to increase potency or selectivity. Herein, we discuss small molecules that interact with the primed substrate pocket and how their differences may give rise to altered activity. Taking advantage of additional interactions with the primed substrate pocket of the proteasome could allow for the generation of improved chemical tools for perturbing or monitoring proteasome activity.


Sujet(s)
Proteasome endopeptidase complex , Proteasome endopeptidase complex/métabolisme , Humains , Spécificité du substrat , Liaison aux protéines , Protéolyse , Inhibiteurs du protéasome/pharmacologie , Inhibiteurs du protéasome/composition chimique , Ubiquitine/métabolisme , Animaux
15.
Biomolecules ; 14(7)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-39062505

RÉSUMÉ

The ubiquitin-proteasome system (UPS) maintains intracellular protein homeostasis and cellular function by regulating various biological processes. Ubiquitination, a common post-translational modification, plays a crucial role in the regulation of protein degradation, signal transduction, and other physiological and pathological processes, and is involved in the pathogenesis of various cancers, including osteosarcoma. Osteosarcoma, the most common primary malignant bone tumor, is characterized by high metastatic potential and poor prognosis. It is a refractory bone disease, and the main treatment modalities are surgery combined with chemotherapy. Increasing evidence suggests a close association between UPS abnormalities and the progression of osteosarcoma. Due to the complexity and pleiotropy of the ubiquitination system, each step in the ubiquitination process can be targeted by drugs. In recent years, research and development of inhibitors targeting the ubiquitin system have increased gradually, showing great potential for clinical application. This article reviews the role of the ubiquitination system in the development and treatment of osteosarcoma, as well as research progress, with the hope of improving the therapeutic effects and prognosis of osteosarcoma patients by targeting effective molecules in the ubiquitination system.


Sujet(s)
Tumeurs osseuses , Ostéosarcome , Ubiquitination , Ostéosarcome/métabolisme , Ostéosarcome/traitement médicamenteux , Ostéosarcome/anatomopathologie , Ostéosarcome/génétique , Humains , Tumeurs osseuses/métabolisme , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/génétique , Ubiquitine/métabolisme , Proteasome endopeptidase complex/métabolisme , Animaux , Transduction du signal
16.
Proc Natl Acad Sci U S A ; 121(32): e2403114121, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39078678

RÉSUMÉ

Parkin is an E3 ubiquitin ligase implicated in early-onset forms of Parkinson's disease. It catalyzes a transthiolation reaction by accepting ubiquitin (Ub) from an E2 conjugating enzyme, forming a short-lived thioester intermediate, and transfers Ub to mitochondrial membrane substrates to signal mitophagy. A major impediment to the development of Parkinsonism therapeutics is the lack of structural and mechanistic detail for the essential, short-lived transthiolation intermediate. It is not known how Ub is recognized by the catalytic Rcat domain in parkin that enables Ub transfer from an E2~Ub conjugate to the catalytic site and the structure of the transthiolation complex is undetermined. Here, we capture the catalytic intermediate for the Rcat domain of parkin in complex with ubiquitin (Rcat-Ub) and determine its structure using NMR-based chemical shift perturbation experiments. We show that a previously unidentified α-helical region near the Rcat domain is unmasked as a recognition motif for Ub and guides the C-terminus of Ub toward the parkin catalytic site. Further, we apply a combination of guided AlphaFold modeling, chemical cross-linking, and single turnover assays to establish and validate a model of full-length parkin in complex with UbcH7, its donor Ub, and phosphoubiquitin, trapped in the process of transthiolation. Identification of this catalytic intermediate and orientation of Ub with respect to the Rcat domain provides important structural insights into Ub transfer by this E3 ligase and explains how the previously enigmatic Parkinson's pathogenic mutation T415N alters parkin activity.


Sujet(s)
Ubiquitin-protein ligases , Ubiquitination , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Humains , Domaine catalytique , Ubiquitine/métabolisme , Ubiquitin-conjugating enzymes/métabolisme , Ubiquitin-conjugating enzymes/génétique , Maladie de Parkinson/métabolisme , Maladie de Parkinson/génétique , Modèles moléculaires
17.
Commun Biol ; 7(1): 907, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39068227

RÉSUMÉ

Affilin proteins, artificial binding proteins based on the ubiquitin scaffold, have been generated by directed protein evolution to yield de-novo variants that bind the extra-domain B (EDB) of oncofetal fibronectin, an established marker of tumor neovasculature. The crystal structures of two EDB-specific Affilin variants reveal a striking structural plasticity of the ubiquitin scaffold, characterised by ß-strand slippage, leading to different negative register shifts of the ß5 strands. This process recruits amino acid residues from ß5 towards the N-terminus to an adjacent loop region and subsequent residues into ß5, respectively, remodeling the binding interface and leading to target specificity and affinity. Protein backbone alterations resulting from ß-strand register shifts, as seen in the ubiquitin fold, can pose additional challenges to protein engineering as structural evidence of these events is still limited and they are difficult to predict. However, they can surface under the selection pressure of directed evolution and suggest that backbone plasticity allowing ß-strand slippages can increase structural diversity, enhancing the evolutionary potential of a protein scaffold.


Sujet(s)
Fibronectines , Ubiquitine , Fibronectines/métabolisme , Fibronectines/composition chimique , Fibronectines/génétique , Ubiquitine/métabolisme , Humains , Liaison aux protéines , Structure en brin bêta , Modèles moléculaires , Cristallographie aux rayons X , Ingénierie des protéines
18.
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
19.
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
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE