Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.789
Filtrer
1.
Biomolecules ; 14(6)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38927053

RÉSUMÉ

The p53 protein is the master regulator of cellular integrity, primarily due to its tumor-suppressing functions. Approximately half of all human cancers carry mutations in the TP53 gene, which not only abrogate the tumor-suppressive functions but also confer p53 mutant proteins with oncogenic potential. The latter is achieved through so-called gain-of-function (GOF) mutations that promote cancer progression, metastasis, and therapy resistance by deregulating transcriptional networks, signaling pathways, metabolism, immune surveillance, and cellular compositions of the microenvironment. Despite recent progress in understanding the complexity of mutp53 in neoplastic development, the exact mechanisms of how mutp53 contributes to cancer development and how they escape proteasomal and lysosomal degradation remain only partially understood. In this review, we address recent findings in the field of oncogenic functions of mutp53 specifically regarding, but not limited to, its implications in metabolic pathways, the secretome of cancer cells, the cancer microenvironment, and the regulating scenarios of the aberrant proteasomal degradation. By analyzing proteasomal and lysosomal protein degradation, as well as its connection with autophagy, we propose new therapeutical approaches that aim to destabilize mutp53 proteins and deactivate its oncogenic functions, thereby providing a fundamental basis for further investigation and rational treatment approaches for TP53-mutated cancers.


Sujet(s)
Tumeurs , Protéolyse , Microenvironnement tumoral , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Tumeurs/thérapie , Microenvironnement tumoral/génétique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Autophagie/génétique , Animaux , Mutation , Lysosomes/métabolisme , Lysosomes/génétique , Carcinogenèse/génétique , Carcinogenèse/métabolisme
2.
Genes (Basel) ; 15(6)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38927717

RÉSUMÉ

We conducted transcriptome sequencing on salt-tolerant mutants X5 and X3, and a control (Ctr) strain of Gracilariopsis lemaneiformis after treatment with artificial seawater at varying salinities (30‱, 45‱, and 60‱) for 3 weeks. Differentially expressed genes were identified and a weighted co-expression network analysis was conducted. The blue, red, and tan modules were most closely associated with salinity, while the black, cyan, light cyan, and yellow modules showed a close correlation with strain attributes. KEGG enrichment of genes from the aforementioned modules revealed that the key enrichment pathways for salinity attributes included the proteasome and carbon fixation in photosynthesis, whereas the key pathways for strain attributes consisted of lipid metabolism, oxidative phosphorylation, soluble N-ethylmaleimide-sensitive factor-activating protein receptor (SNARE) interactions in vesicular transport, and porphyrin and chlorophyll metabolism. Gene expression for the proteasome and carbon fixation in photosynthesis was higher in all strains at 60‱. In addition, gene expression in the proteasome pathway was higher in the X5-60 than Ctr-60 and X3-60. Based on the above data and relevant literature, we speculated that mutant X5 likely copes with high salt stress by upregulating genes related to lysosome and carbon fixation in photosynthesis. The proteasome may be reset to adjust the organism's proteome composition to adapt to high-salt environments, while carbon fixation may aid in maintaining material and energy metabolism for normal life activities by enhancing carbon dioxide uptake via photosynthesis. The differences between the X5-30 and Ctr-30 expression of genes involved in the synthesis of secondary metabolites, oxidative phosphorylation, and SNARE interactions in vesicular transport suggested that the X5-30 may differ from Ctr-30 in lipid metabolism, energy metabolism, and vesicular transport. Finally, among the key pathways with good correlation with salinity and strain traits, the key genes with significant correlation with salinity and strain traits were identified by correlation analysis.


Sujet(s)
Tolérance au sel , Tolérance au sel/génétique , Transcriptome , Réseaux de régulation génique , Salinité , Photosynthèse/génétique , Pression osmotique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Analyse de profil d'expression de gènes/méthodes , Métabolisme lipidique/génétique
3.
Int J Mol Sci ; 25(12)2024 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-38928079

RÉSUMÉ

Proteasome 26S Subunit, Non-ATPase 9 (psmd9) plays an important role in the balance of protamine and the stability of the nucleolar structure during spermatogenesis. In this study, we cloned the psmd9 of Cynoglossus semilaevis and analyzed its expression pattern. psmd9 was identified on the Z chromosome of C. semilaevis, which is considered an interesting candidate gene for spermatogenesis. qRT-PCR and FISH experiments showed that the psmd9 gene was significantly highly expressed in the testes. It is worth noting that the expression level of psmd9 in male fish testes is significantly higher than that in pseudomales. In order to further explore the role of psmd9 in spermatogenesis, a male testicular cell line was used as the experimental material. The results of the psmd9-RNAi and overexpression experiments showed that psmd9 had a synergistic effect with spermatogenesis-related genes dnd1, cfap69, dnah3 and dnajb13, but had an antagonistic effect with ccne2. Our findings offer a scientific foundation for comprehending the role of psmd9 in the spermatogenesis regulatory network of C. semilaevis.


Sujet(s)
Spermatogenèse , Testicule , Animaux , Spermatogenèse/génétique , Mâle , Testicule/métabolisme , Chromosomes sexuels/génétique , Protéines de poisson/génétique , Protéines de poisson/métabolisme , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Clonage moléculaire
4.
Cancer Lett ; 596: 217020, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38849009

RÉSUMÉ

B7-H4 is an immune checkpoint crucial for inhibiting CD8+ T-cell activity. A clinical trial is underway to investigate B7-H4 as a potential immunotherapeutic agent. However, the regulatory mechanism of B7-H4 degradation via the ubiquitin-proteasome pathway (UPP) remains poorly understood. In this study, we discovered that proteasome inhibitors effectively increased B7-H4 expression, while EGFR-activating mutants promoted B7-H4 expression through the UPP. We screened B7-H4 binding proteins by co-immunoprecipitation and mass spectrometry and found that USP2a acted as a deubiquitinase of B7-H4 by removing K48- and K63-linked ubiquitin chains from B7-H4, leading to a reduction in B7-H4 degradation. EGFR mutants enhanced B7-H4 stability by upregulating USP2a expression. We further investigated the role of USP2a in tumor growth in vivo. Depletion of USP2a in L858R/LLC cells inhibited tumor cell proliferation, consequently suppressing tumor growth in immune-deficient nude mice by destabilizing downstream molecules such as Cyclin D1. In an immune-competent C57BL/6 mouse tumor model, USP2a abrogation facilitated infiltration of CD95+CD8+ effector T cells and hindered infiltration of Tim-3+CD8+ and LAG-3+CD8+ exhausted T cells by destabilizing B7-H4. Clinical lung adenocarcinoma samples showed a significant correlation between B7-H4 abundance and USP2a expression, indicating the contribution of the EGFR/USP2a/B7-H4 axis to tumor immunosuppression. In summary, this study elucidates the dual effects of USP2a in tumor growth by stabilizing Cyclin D1, promoting tumor cell proliferation, and stabilizing B7-H4, contributing to tumor immunosuppression. Therefore, USP2a represents a potential target for tumor therapy.


Sujet(s)
Adénocarcinome pulmonaire , Récepteurs ErbB , Tumeurs du poumon , Souris nude , Ubiquitin thiolesterase , V-set domain-containing T-cell activation inhibitor 1 , Animaux , Humains , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme , Souris , V-set domain-containing T-cell activation inhibitor 1/génétique , V-set domain-containing T-cell activation inhibitor 1/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/immunologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/traitement médicamenteux , Lignée cellulaire tumorale , Prolifération cellulaire , Mutation , Souris de lignée C57BL , Lymphocytes T CD8+/immunologie , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique
5.
Mol Biol Rep ; 51(1): 770, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38896168

RÉSUMÉ

BACKGROUND: MG132, a proteasome inhibitor, is widely used to inhibit nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activity by proteasome-mediated degradation of IκB. It has been marketed as a specific, reversible, cell-permeable and low-cost inhibitor. However, adverse effects of the compound have been reported in the literature. We recently discovered and characterised a point mutation in the acute phase protein serum amyloid A (SAA) in chickens, by overexpressing the protein in chicken hepatocellular carcinoma (LMH) cells. This serine to arginine exchange at amino acid position 90 (SAA.R90S) leads to intra- and extracellular accumulation of SAA, which is surprisingly counteracted by MG132 treatment, independent of SAA's intrinsic promoter. METHODS AND RESULTS: To test, whether low proteasomal degradation of SAA.R90S is responsible for the observed intra- and extracellular SAA accumulation, we intended to inhibit the proteasome in SAA wild type (SAA.WT) overexpressing cells with MG132. However, we observed an unexpected drastic decrease in SAA protein expression at the transcript level. NF-κB gene expression was unchanged by MG132 at the measured time point. CONCLUSIONS: The observed results demonstrate that MG132 inhibits SAA expression at the transcript level, independent of its endogenous promoter. Further, the data might indicate that NF-κB is not involved in the observed MG132-induced inhibition of SAA expression. We, consequently, question in this brief report whether MG132 should truly be categorised as a specific ubiquitin proteasome inhibitor and recommend the usage of alternative compounds.


Sujet(s)
Carcinome hépatocellulaire , Poulets , Leupeptines , Tumeurs du foie , Facteur de transcription NF-kappa B , Régions promotrices (génétique) , Protéine amyloïde A sérique , Animaux , Leupeptines/pharmacologie , Poulets/génétique , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/traitement médicamenteux , Lignée cellulaire tumorale , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/traitement médicamenteux , Régions promotrices (génétique)/génétique , Protéine amyloïde A sérique/génétique , Protéine amyloïde A sérique/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Inhibiteurs du protéasome/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
6.
J Biol Chem ; 300(6): 107337, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38705397

RÉSUMÉ

APE2 plays important roles in the maintenance of genomic and epigenomic stability including DNA repair and DNA damage response. Accumulating evidence has suggested that APE2 is upregulated in multiple cancers at the protein and mRNA levels and that APE2 upregulation is correlative with higher and lower overall survival of cancer patients depending on tumor type. However, it remains unknown how APE2 protein abundance is maintained and regulated in cells. Here, we provide the first evidence of APE2 regulation via the posttranslational modification ubiquitin. APE2 is poly-ubiquitinated via K48-linked chains and degraded via the ubiquitin-proteasome system where K371 is the key residue within APE2 responsible for its ubiquitination and degradation. We further characterize MKRN3 as the E3 ubiquitin ligase for APE2 ubiquitination in cells and in vitro. In summary, this study offers the first definition of the APE2 proteostasis network and lays the foundation for future studies pertaining to the posttranslational modification regulation and functions of APE2 in genome integrity and cancer etiology/treatment.


Sujet(s)
DNA-(apurinic or apyrimidinic site) lyase , Ubiquitination , Humains , DNA-(apurinic or apyrimidinic site) lyase/métabolisme , DNA-(apurinic or apyrimidinic site) lyase/génétique , Ubiquitine/métabolisme , Maturation post-traductionnelle des protéines , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Cellules HEK293 , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Protéolyse , Endonucleases , Enzymes multifonctionnelles
7.
J Biol Chem ; 300(6): 107353, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38723751

RÉSUMÉ

Recent genome-wide association studies have identified a missense variant p.A165T in mitochondrial amidoxime-reducing component 1 (mARC1) that is strongly associated with protection from all-cause cirrhosis and improved prognosis in nonalcoholic steatohepatitis. The precise mechanism of this protective effect is unknown. Substitution of alanine 165 with threonine is predicted to affect mARC1 protein stability and to have deleterious effects on its function. To investigate the mechanism, we have generated a knock-in mutant mARC1 A165T and a catalytically dead mutant C273A (as a control) in human hepatoma HepG2 cells, enabling characterization of protein subcellular distribution, stability, and biochemical functions of the mARC1 mutant protein expressed from its endogenous locus. Compared to WT mARC1, we found that the A165T mutant exhibits significant mislocalization outside of its traditional location anchored in the mitochondrial outer membrane and reduces protein stability, resulting in lower basal levels. We evaluated the involvement of the ubiquitin proteasome system in mARC1 A165T degradation and observed increased ubiquitination and faster degradation of the A165T variant. In addition, we have shown that HepG2 cells carrying the MTARC1 p.A165T variant exhibit lower N-reductive activity on exogenously added amidoxime substrates in vitro. The data from these biochemical and functional assays suggest a mechanism by which the MTARC1 p.A165T variant abrogates enzyme function which may contribute to its protective effect in liver disease.


Sujet(s)
Protéines mitochondriales , Mutation faux-sens , Humains , Cellules HepG2 , Protéines mitochondriales/génétique , Protéines mitochondriales/métabolisme , Ubiquitination , Stabilité protéique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Protéolyse , Oxidoreductases
8.
FEBS J ; 291(12): 2562-2564, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38708447

RÉSUMÉ

Around 10% of Parkinson's disease (PD) cases are associated with mutations in various genes, including FBXO7, which encodes the substrate-recognition component for the Skp1-Cullin-F-box (SCF) class of ubiquitin E3 ligases that target proteins for proteasomal degradation. In their recent study, Al Rawi et al. characterized a new mutation in FBXO7, L250P, in a pediatric patient. Their findings reveal that the L250P mutation abolishes Fbxo7 interaction with the proteasome regulator, proteasome inhibitor 31kD (PI31), affecting proteasomal activity and the ubiquitination of some of the ligase's targets. Furthermore, the authors show that this previously undescribed mutation impairs mitochondrial function and mitophagy, emphasizing the importance of mitochondrial and proteasomal dysfunction in PD pathogenesis.


Sujet(s)
Protéines F-box , Mitochondries , Maladie de Parkinson , Proteasome endopeptidase complex , Humains , Maladie de Parkinson/génétique , Maladie de Parkinson/anatomopathologie , Maladie de Parkinson/métabolisme , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Protéines F-box/génétique , Protéines F-box/métabolisme , Mitochondries/génétique , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Mutation , Mitophagie/génétique , Ubiquitination
9.
Mol Cell ; 84(11): 2166-2184.e9, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38788716

RÉSUMÉ

Mammalian target of rapamycin (mTOR) senses changes in nutrient status and stimulates the autophagic process to recycle amino acids. However, the impact of nutrient stress on protein degradation beyond autophagic turnover is incompletely understood. We report that several metabolic enzymes are proteasomal targets regulated by mTOR activity based on comparative proteome degradation analysis. In particular, 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) synthase 1 (HMGCS1), the initial enzyme in the mevalonate pathway, exhibits the most significant half-life adaptation. Degradation of HMGCS1 is regulated by the C-terminal to LisH (CTLH) E3 ligase through the Pro/N-degron motif. HMGCS1 is ubiquitylated on two C-terminal lysines during mTORC1 inhibition, and efficient degradation of HMGCS1 in cells requires a muskelin adaptor. Importantly, modulating HMGCS1 abundance has a dose-dependent impact on cell proliferation, which is restored by adding a mevalonate intermediate. Overall, our unbiased degradomics study provides new insights into mTORC1 function in cellular metabolism: mTORC1 regulates the stability of limiting metabolic enzymes through the ubiquitin system.


Sujet(s)
Prolifération cellulaire , Hydroxymethylglutaryl-coA synthase , Complexe-1 cible mécanistique de la rapamycine , Protéolyse , Ubiquitin-protein ligases , Ubiquitination , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/génétique , Humains , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Cellules HEK293 , Hydroxymethylglutaryl-coA synthase/métabolisme , Hydroxymethylglutaryl-coA synthase/génétique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Acide mévalonique/métabolisme , Complexes multiprotéiques/métabolisme , Complexes multiprotéiques/génétique , Transduction du signal , , Protéines adaptatrices de la transduction du signal
10.
Nature ; 630(8015): 149-157, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38778096

RÉSUMÉ

Accessing the natural genetic diversity of species unveils hidden genetic traits, clarifies gene functions and allows the generalizability of laboratory findings to be assessed. One notable discovery made in natural isolates of Saccharomyces cerevisiae is that aneuploidy-an imbalance in chromosome copy numbers-is frequent1,2 (around 20%), which seems to contradict the substantial fitness costs and transient nature of aneuploidy when it is engineered in the laboratory3-5. Here we generate a proteomic resource and merge it with genomic1 and transcriptomic6 data for 796 euploid and aneuploid natural isolates. We find that natural and lab-generated aneuploids differ specifically at the proteome. In lab-generated aneuploids, some proteins-especially subunits of protein complexes-show reduced expression, but the overall protein levels correspond to the aneuploid gene dosage. By contrast, in natural isolates, more than 70% of proteins encoded on aneuploid chromosomes are dosage compensated, and average protein levels are shifted towards the euploid state chromosome-wide. At the molecular level, we detect an induction of structural components of the proteasome, increased levels of ubiquitination, and reveal an interdependency of protein turnover rates and attenuation. Our study thus highlights the role of protein turnover in mediating aneuploidy tolerance, and shows the utility of exploiting the natural diversity of species to attain generalizable molecular insights into complex biological processes.


Sujet(s)
Aneuploïdie , Proteasome endopeptidase complex , Protéolyse , Protéome , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Compensation de dosage génétique , Variation génétique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Protéome/métabolisme , Protéome/génétique , Protéomique , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Ubiquitination , Analyse de profil d'expression de gènes , Génomique
11.
Biochemistry ; 63(11): 1423-1433, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38743592

RÉSUMÉ

PGM1-linked congenital disorder of glycosylation (PGM1-CDG) is an autosomal recessive disease characterized by several phenotypes, some of which are life-threatening. Research focusing on the disease-related variants of the α-D-phosphoglucomutase 1 (PGM1) protein has shown that several are insoluble in vitro and expressed at low levels in patient fibroblasts. Due to these observations, we hypothesized that some disease-linked PGM1 protein variants are structurally destabilized and subject to protein quality control (PQC) and rapid intracellular degradation. Employing yeast-based assays, we show that a disease-associated human variant, PGM1 L516P, is insoluble, inactive, and highly susceptible to ubiquitylation and rapid degradation by the proteasome. In addition, we show that PGM1 L516P forms aggregates in S. cerevisiae and that both the aggregation pattern and the abundance of PGM1 L516P are chaperone-dependent. Finally, using computational methods, we perform saturation mutagenesis to assess the impact of all possible single residue substitutions in the PGM1 protein. These analyses identify numerous missense variants with predicted detrimental effects on protein function and stability. We suggest that many disease-linked PGM1 variants are subject to PQC-linked degradation and that our in silico site-saturated data set may assist in the mechanistic interpretation of PGM1 variants.


Sujet(s)
Phosphoglucomutase , Saccharomyces cerevisiae , Humains , Phosphoglucomutase/métabolisme , Phosphoglucomutase/génétique , Phosphoglucomutase/composition chimique , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Protéolyse , Mutation faux-sens , Ubiquitination , Troubles congénitaux de la glycosylation/génétique , Troubles congénitaux de la glycosylation/métabolisme , Stabilité protéique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique
12.
Int J Mol Sci ; 25(10)2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38791597

RÉSUMÉ

Bladder cancer (BC) is a malignant tumor of the urinary system with high mortality and recurrence rates. Proteasome subunit type 4 (PSMB4) is highly expressed and has been identified as having oncogenic properties in a variety of cancer types. This study aimed to explore the effect of PSMB4 knockdown on the survival, migration, and angiogenesis of human bladder cancer cells with different degrees of malignancy. We analyzed the effects of PSMB4 knockdown in bladder cancer cells and endothelial cells in the tumor microenvironment. PSMB4 was highly expressed in patients with low- and high-grade urothelial carcinoma. Inhibition of PSMB4 reduced protein expression of focal adhesion kinase (FAK) and myosin light chain (MLC), leading to reduced migration. Furthermore, the suppression of PSMB4 decreased the levels of vascular endothelial factor B (VEGF-B), resulting in lower angiogenic abilities in human bladder cancer cells. PSMB4 inhibition affected the migratory ability of HUVECs and reduced VEGFR2 expression, consequently downregulating angiogenesis. In the metastatic animal model, PSMB4 knockdown reduced the relative volumes of lung tumors. Our findings suggest the role of PSMB4 as a potential target for therapeutic strategies against human bladder cancer.


Sujet(s)
Mouvement cellulaire , Néovascularisation pathologique , Proteasome endopeptidase complex , Tumeurs de la vessie urinaire , Humains , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/métabolisme , Mouvement cellulaire/génétique , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Lignée cellulaire tumorale , Animaux , Souris , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Techniques de knock-down de gènes , Régulation de l'expression des gènes tumoraux , Microenvironnement tumoral/génétique , Mâle , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique , Femelle , , Cysteine endopeptidases
13.
J Cell Biol ; 223(8)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38767572

RÉSUMÉ

Proteasome activity is crucial for cellular integrity, but how tissues adjust proteasome content in response to catabolic stimuli is uncertain. Here, we demonstrate that transcriptional coordination by multiple transcription factors is required to increase proteasome content and activate proteolysis in catabolic states. Using denervated mouse muscle as a model system for accelerated proteolysis in vivo, we reveal that a two-phase transcriptional program activates genes encoding proteasome subunits and assembly chaperones to boost an increase in proteasome content. Initially, gene induction is necessary to maintain basal proteasome levels, and in a more delayed phase (7-10 days after denervation), it stimulates proteasome assembly to meet cellular demand for excessive proteolysis. Intriguingly, the transcription factors PAX4 and α-PALNRF-1 control the expression of proteasome among other genes in a combinatorial manner, driving cellular adaptation to muscle denervation. Consequently, PAX4 and α-PALNRF-1 represent new therapeutic targets to inhibit proteolysis in catabolic diseases (e.g., type-2 diabetes, cancer).


Sujet(s)
Facteur nucléaire-1 respiratoire , Facteurs de transcription PAX , Proteasome endopeptidase complex , Protéolyse , Animaux , Mâle , Souris , Régulation de l'expression des gènes , Muscles squelettiques/métabolisme , Facteurs de transcription PAX/métabolisme , Facteurs de transcription PAX/génétique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Souris de lignée ICR , Facteur nucléaire-1 respiratoire/génétique , Facteur nucléaire-1 respiratoire/métabolisme
14.
Nat Commun ; 15(1): 4026, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38740822

RÉSUMÉ

Unstable proteins are prone to form non-native interactions with other proteins and thereby may become toxic. To mitigate this, destabilized proteins are targeted by the protein quality control network. Here we present systematic studies of the cytosolic aspartoacylase, ASPA, where variants are linked to Canavan disease, a lethal neurological disorder. We determine the abundance of 6152 of the 6260 ( ~ 98%) possible single amino acid substitutions and nonsense ASPA variants in human cells. Most low abundance variants are degraded through the ubiquitin-proteasome pathway and become toxic upon prolonged expression. The data correlates with predicted changes in thermodynamic stability, evolutionary conservation, and separate disease-linked variants from benign variants. Mapping of degradation signals (degrons) shows that these are often buried and the C-terminal region functions as a degron. The data can be used to interpret Canavan disease variants and provide insight into the relationship between protein stability, degradation and cell fitness.


Sujet(s)
Amidohydrolases , Maladie de Canavan , Protéolyse , Humains , Amidohydrolases/génétique , Amidohydrolases/métabolisme , Maladie de Canavan/génétique , Maladie de Canavan/métabolisme , Cellules HEK293 , Substitution d'acide aminé , Mutation , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Stabilité protéique , Ubiquitine/métabolisme , Thermodynamique
15.
Free Radic Biol Med ; 219: 31-48, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38614226

RÉSUMÉ

Hepatocellular carcinoma (HCC) is the predominant form of liver cancer, characterized by high morbidity and mortality rates, as well as unfavorable treatment outcomes. Tripartite motif-containing protein 47 (TRIM47) has been implicated in various diseases including tumor progression with the activity of E3 ubiquitin ligase. However, the precise regulatory mechanisms underlying the involvement of TRIM47 in HCC remain largely unexplored. Here, we provide evidence that TRIM47 exhibits heightened expression in tumor tissues, and its expression is in intimate association with clinical staging and patient prognosis. TRIM47 promotes HCC proliferation, migration, and invasion as an oncogene by in vitro gain- and loss-of-function experiments. TRIM47 knockdown results in HCC ferroptosis induction, primarily through CDO1 involvement to regulate GSH synthesis. Subsequent experiments confirm the interaction between TRIM47 and CDO1 dependent on B30.2 domain, wherein TRIM47 facilitates K48-linked ubiquitination, leading to a decrease in CDO1 protein abundance in HCC. Furthermore, CDO1 is able to counteract the promotional effect of TRIM47 on HCC biological functions. Overall, our research provides novel insight into the mechanism of TRIM47 in CDO1-mediated ferroptosis in HCC cells, highlighting its value as a potential target candidate for HCC therapeutic approaches.


Sujet(s)
Carcinome hépatocellulaire , Cysteine dioxygenase , Ferroptose , Tumeurs du foie , Proteasome endopeptidase complex , Protéines à motif tripartite , Animaux , Humains , Souris , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire , Évolution de la maladie , Ferroptose/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Protéines tumorales/métabolisme , Protéines nucléaires/métabolisme , Pronostic , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Protéines à motif tripartite/métabolisme , Ubiquitine/métabolisme , Ubiquitination , Cysteine dioxygenase/génétique , Cysteine dioxygenase/métabolisme
16.
Nat Cell Biol ; 26(5): 784-796, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38600234

RÉSUMÉ

DNA-protein crosslinks (DPCs) induced by aldehydes interfere with replication and transcription. Hereditary deficiencies in DPC repair and aldehyde clearance processes cause progeria, including Ruijs-Aalfs syndrome (RJALS) and AMeD syndrome (AMeDS) in humans. Although the elimination of DPC during replication has been well established, how cells overcome DPC lesions in transcription remains elusive. Here we show that endogenous aldehyde-induced DPC roadblocks are efficiently resolved by transcription-coupled repair (TCR). We develop a high-throughput sequencing technique to measure the genome-wide distribution of DPCs (DPC-seq). Using proteomics and DPC-seq, we demonstrate that the conventional TCR complex as well as VCP/p97 and the proteasome are required for the removal of formaldehyde-induced DPCs. TFIIS-dependent cleavage of RNAPII transcripts protects against transcription obstacles. Finally, a mouse model lacking both aldehyde clearance and TCR confirms endogenous DPC accumulation in actively transcribed regions. Collectively, our data provide evidence that transcription-coupled DPC repair (TC-DPCR) as well as aldehyde clearance are crucial for protecting against metabolic genotoxin, thus explaining the molecular pathogenesis of AMeDS and other disorders associated with defects in TCR, such as Cockayne syndrome.


Sujet(s)
Aldéhydes , Réparation de l'ADN , Transcription génétique , Animaux , Humains , Aldéhydes/métabolisme , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Souris , ADN/métabolisme , ADN/génétique , Altération de l'ADN , Souris knockout , Protéine contenant la valosine/métabolisme , Protéine contenant la valosine/génétique , RNA polymerase II/métabolisme , RNA polymerase II/génétique , Souris de lignée C57BL , Formaldéhyde/toxicité , Formaldéhyde/pharmacologie , Réparation par excision
17.
J Clin Immunol ; 44(4): 88, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38578475

RÉSUMÉ

The ubiquitin-proteasome system (UPS) has a critical role in post-translational protein modification that is essential for the maintenance of all cellular functions, including immune responses. The proteasome complex is ubiquitously expressed and is responsible for degradation of short-lived structurally abnormal, misfolded and not-needed proteins that are targeted for degradation via ubiquitin conjugation. Over the last 14 years, an increasing number of human diseases have been linked to pathogenic variants in proteasome subunits and UPS regulators. Defects of the proteasome complex or its chaperons - which have a regulatory role in the assembly of the proteasome - disrupt protein clearance and cellular homeostasis, leading to immune dysregulation, severe inflammation, and neurodevelopmental disorders in humans. Proteasome-associated diseases have complex inheritance, including monogenic, digenic and oligogenic disorders and can be dominantly or recessively inherited. In this review, we summarize the current known genetic causes of proteasomal disease, and discuss the molecular pathogenesis of these conditions based on the function and cellular expression of mutated proteins in the proteasome complex.


Sujet(s)
Proteasome endopeptidase complex , Ubiquitine , Humains , Proteasome endopeptidase complex/génétique , Proteasome endopeptidase complex/métabolisme , Syndrome , Ubiquitine/métabolisme
18.
Cancer Res Commun ; 4(4): 1082-1099, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38625038

RÉSUMÉ

The 26S proteasome is the major protein degradation machinery in cells. Cancer cells use the proteasome to modulate gene expression networks that promote tumor growth. Proteasome inhibitors have emerged as effective cancer therapeutics, but how they work mechanistically remains unclear. Here, using integrative genomic analysis, we discovered unexpected reprogramming of the chromatin landscape and RNA polymerase II (RNAPII) transcription initiation in breast cancer cells treated with the proteasome inhibitor MG132. The cells acquired dynamic changes in chromatin accessibility at specific genomic loci termed differentially open chromatin regions (DOCR). DOCRs with decreased accessibility were promoter proximal and exhibited unique chromatin architecture associated with divergent RNAPII transcription. Conversely, DOCRs with increased accessibility were primarily distal to transcription start sites and enriched in oncogenic superenhancers predominantly accessible in non-basal breast tumor subtypes. These findings describe the mechanisms by which the proteasome modulates the expression of gene networks intrinsic to breast cancer biology. SIGNIFICANCE: Our study provides a strong basis for understanding the mechanisms by which proteasome inhibitors exert anticancer effects. We find open chromatin regions that change during proteasome inhibition, are typically accessible in non-basal breast cancers.


Sujet(s)
Chromatine , Tumeurs , Chromatine/génétique , Proteasome endopeptidase complex/génétique , Inhibiteurs du protéasome/pharmacologie , Protéolyse , Génomique
19.
J Cell Biol ; 223(6)2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38656405

RÉSUMÉ

Cells exposed to proteotoxic stress invoke adaptive responses aimed at restoring proteostasis. Our previous studies have established a firm role for the transcription factor Nuclear factor-erythroid derived-2-related factor-1 (Nrf1) in responding to proteotoxic stress elicited by inhibition of cellular proteasome. Following proteasome inhibition, Nrf1 mediates new proteasome synthesis, thus enabling the cells to mitigate the proteotoxic stress. Here, we report that under similar circumstances, multiple components of the autophagy-lysosomal pathway (ALP) were transcriptionally upregulated in an Nrf1-dependent fashion, thus providing the cells with an additional route to cope with proteasome insufficiency. In response to proteasome inhibitors, Nrf1-deficient cells displayed profound defects in invoking autophagy and clearance of aggresomes. This phenomenon was also recapitulated in NGLY1 knockout cells, where Nrf1 is known to be non-functional. Conversely, overexpression of Nrf1 induced ALP genes and endowed the cells with an increased capacity to clear aggresomes. Overall, our results significantly expand the role of Nrf1 in shaping the cellular response to proteotoxic stress.


Sujet(s)
Autophagie , Facteur-1 apparenté à NF-E2 , Stress protéotoxique , Animaux , Humains , Souris , Autophagie/génétique , Lysosomes/métabolisme , Facteur-1 apparenté à NF-E2/métabolisme , Facteur-1 apparenté à NF-E2/génétique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Inhibiteurs du protéasome/pharmacologie , Homéostasie protéique , Stress physiologique
20.
Genet Med ; 26(6): 101120, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38469793

RÉSUMÉ

PURPOSE: Imbalances in protein homeostasis affect human brain development, with the ubiquitin-proteasome system (UPS) and autophagy playing crucial roles in neurodevelopmental disorders (NDD). This study explores the impact of biallelic USP14 variants on neurodevelopment, focusing on its role as a key hub connecting UPS and autophagy. METHODS: Here, we identified biallelic USP14 variants in 4 individuals from 3 unrelated families: 1 fetus, a newborn with a syndromic NDD and 2 siblings affected by a progressive neurological disease. Specifically, the 2 siblings from the latter family carried 2 compound heterozygous variants c.8T>C p.(Leu3Pro) and c.988C>T p.(Arg330∗), whereas the fetus had a homozygous frameshift c.899_902del p.(Lys300Serfs∗24) variant, and the newborn patient harbored a homozygous frameshift c.233_236del p.(Leu78Glnfs∗11) variant. Functional studies were conducted using sodium dodecyl-sulfate polyacrylamide gel electrophoresis, western blotting, and mass spectrometry analyses in both patient-derived and CRISPR-Cas9-generated cells. RESULTS: Our investigations indicated that the USP14 variants correlated with reduced N-terminal methionine excision, along with profound alterations in proteasome, autophagy, and mitophagy activities. CONCLUSION: Biallelic USP14 variants in NDD patients perturbed protein degradation pathways, potentially contributing to disorder etiology. Altered UPS, autophagy, and mitophagy activities underscore the intricate interplay, elucidating their significance in maintaining proper protein homeostasis during brain development.


Sujet(s)
Troubles du développement neurologique , Humains , Troubles du développement neurologique/génétique , Troubles du développement neurologique/anatomopathologie , Femelle , Mâle , Allèles , Autophagie/génétique , Ubiquitin thiolesterase/génétique , Nouveau-né , Proteasome endopeptidase complex/génétique , Pedigree , Homozygote , Prédisposition génétique à une maladie , Mutation/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...