Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 28.410
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(28): e2320655121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38959043

RÉSUMÉ

SLC7A11 is a cystine transporter and ferroptosis inhibitor. How the stability of SLC7A11 is coordinately regulated in response to environmental cystine by which E3 ligase and deubiquitylase (DUB) remains elusive. Here, we report that neddylation inhibitor MLN4924 increases cystine uptake by causing SLC7A11 accumulation, via inactivating Cullin-RING ligase-3 (CRL-3). We identified KCTD10 as the substrate-recognizing subunit of CRL-3 for SLC7A11 ubiquitylation, and USP18 as SLC7A11 deubiquitylase. Upon cystine deprivation, the protein levels of KCTD10 or USP18 are decreased or increased, respectively, contributing to SLC7A11 accumulation. By destabilizing or stabilizing SLC7A11, KCTD10, or USP18 inversely regulates the cystine uptake and ferroptosis. Biologically, MLN4924 combination with SLC7A11 inhibitor Imidazole Ketone Erastin (IKE) enhanced suppression of tumor growth. In human breast tumor tissues, SLC7A11 levels were negatively or positively correlated with KCTD10 or USP18, respectively. Collectively, our study defines how SLC7A11 and ferroptosis is coordinately regulated by the CRL3KCTD10/E3-USP18/DUB axis, and provides a sound rationale of drug combination to enhance anticancer efficacy.


Sujet(s)
Système y+ de transport d'acides aminés , Cystine , Ferroptose , Pyrimidines , Ubiquitin thiolesterase , Humains , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Pyrimidines/pharmacologie , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Animaux , Cystine/métabolisme , Cyclopentanes/métabolisme , Cyclopentanes/pharmacologie , Lignée cellulaire tumorale , Ubiquitination , Femelle , Souris , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Pipérazines/pharmacologie , Cellules HEK293
2.
Nat Commun ; 15(1): 5515, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951495

RÉSUMÉ

Like many other viruses, KSHV has two life cycle modes: the latent phase and the lytic phase. The RTA protein from KSHV is essential for lytic reactivation, but how this protein's activity is regulated is not fully understood. Here, we report that linear ubiquitination regulates the activity of RTA during KSHV lytic reactivation and de novo infection. Overexpressing OTULIN inhibits KSHV lytic reactivation, whereas knocking down OTULIN or overexpressing HOIP enhances it. Intriguingly, we found that RTA is linearly polyubiquitinated by HOIP at K516 and K518, and these modifications control the RTA's nuclear localization. OTULIN removes linear polyubiquitin chains from cytoplasmic RTA, preventing its nuclear import. The RTA orthologs encoded by the EB and MHV68 viruses are also linearly polyubiquitinated and regulated by OTULIN. Our study establishes that linear polyubiquitination plays a critically regulatory role in herpesvirus infection, adding virus infection to the list of biological processes known to be controlled by linear polyubiquitination.


Sujet(s)
Herpèsvirus humain de type 8 , Protéines précoces immédiates , Transactivateurs , Ubiquitination , Réplication virale , Herpèsvirus humain de type 8/physiologie , Herpèsvirus humain de type 8/génétique , Herpèsvirus humain de type 8/métabolisme , Humains , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Cellules HEK293 , Transactivateurs/métabolisme , Transactivateurs/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Activation virale , Infections à Herpesviridae/métabolisme , Infections à Herpesviridae/virologie , Noyau de la cellule/métabolisme
3.
Nat Commun ; 15(1): 5514, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951492

RÉSUMÉ

HIV-1 Vpr promotes efficient spread of HIV-1 from macrophages to T cells by transcriptionally downmodulating restriction factors that target HIV-1 Envelope protein (Env). Here we find that Vpr induces broad transcriptomic changes by targeting PU.1, a transcription factor necessary for expression of host innate immune response genes, including those that target Env. Consistent with this, we find silencing PU.1 in infected macrophages lacking Vpr rescues Env. Vpr downmodulates PU.1 through a proteasomal degradation pathway that depends on physical interactions with PU.1 and DCAF1, a component of the Cul4A E3 ubiquitin ligase. The capacity for Vpr to target PU.1 is highly conserved across primate lentiviruses. In addition to impacting infected cells, we find that Vpr suppresses expression of innate immune response genes in uninfected bystander cells, and that virion-associated Vpr can degrade PU.1. Together, we demonstrate Vpr counteracts PU.1 in macrophages to blunt antiviral immune responses and promote viral spread.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Immunité innée , Macrophages , Protéines proto-oncogènes , Transactivateurs , Produits du gène vpr du virus de l'immunodéficience humaine , Humains , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/virologie , Produits du gène vpr du virus de l'immunodéficience humaine/métabolisme , Produits du gène vpr du virus de l'immunodéficience humaine/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Transactivateurs/métabolisme , Transactivateurs/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Infections à VIH/immunologie , Infections à VIH/virologie , Infections à VIH/génétique , Cellules HEK293 , Virion/métabolisme , Protein-Serine-Threonine Kinases
4.
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
5.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38969364

RÉSUMÉ

The transmembrane E3 ligases RNF43 and ZNRF3 perform key tumour suppressor roles by inducing endocytosis of members of the Frizzled (FZD) family, the primary receptors for WNT. Loss-of-function mutations in RNF43 and ZNRF3 mediate FZD stabilisation and a WNT-hypersensitive growth state in various cancer types. Strikingly, RNF43 and ZNRF3 mutations are differentially distributed across cancer types, raising questions about their functional redundancy. Here, we compare the efficacy of RNF43 and ZNRF3 of targeting different FZDs for endocytosis. We find that RNF43 preferentially down-regulates FZD1/FZD5/FZD7, whereas ZNRF3 displays a preference towards FZD6. We show that the RNF43 transmembrane domain (TMD) is a key molecular determinant for inducing FZD5 endocytosis. Furthermore, a TMD swap between RNF43 and ZNRF3 re-directs their preference for FZD5 down-regulation. We conclude that RNF43 and ZNRF3 preferentially down-regulate specific FZDs, in part by a TMD-dependent mechanism. In accordance, tissue-specific expression patterns of FZD homologues correlate with the incidence of RNF43 or ZNRF3 cancer mutations in those tissues. Consequently, our data point to druggable vulnerabilities of specific FZD receptors in RNF43- or ZNRF3-mutant human cancers.


Sujet(s)
Endocytose , Récepteurs Frizzled , Ubiquitin-protein ligases , Récepteurs Frizzled/métabolisme , Récepteurs Frizzled/génétique , Humains , Endocytose/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Cellules HEK293 , Mutation , Voie de signalisation Wnt/génétique , Régulation négative/génétique
6.
Science ; 385(6704): 91-99, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38963839

RÉSUMÉ

Sickle cell disease (SCD) is a prevalent, life-threatening condition attributable to a heritable mutation in ß-hemoglobin. Therapeutic induction of fetal hemoglobin (HbF) can ameliorate disease complications and has been intently pursued. However, safe and effective small-molecule inducers of HbF remain elusive. We report the discovery of dWIZ-1 and dWIZ-2, molecular glue degraders of the WIZ transcription factor that robustly induce HbF in erythroblasts. Phenotypic screening of a cereblon (CRBN)-biased chemical library revealed WIZ as a previously unknown repressor of HbF. WIZ degradation is mediated by recruitment of WIZ(ZF7) to CRBN by dWIZ-1, as resolved by crystallography of the ternary complex. Pharmacological degradation of WIZ was well tolerated and induced HbF in humanized mice and cynomolgus monkeys. These findings establish WIZ degradation as a globally accessible therapeutic strategy for SCD.


Sujet(s)
Drépanocytose , Hémoglobine foetale , Hémoglobine foetale/génétique , Hémoglobine foetale/métabolisme , Animaux , Drépanocytose/génétique , Drépanocytose/métabolisme , Humains , Souris , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéolyse , Macaca fascicularis , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique , Cristallographie aux rayons X
7.
Cell Death Dis ; 15(7): 473, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956064

RÉSUMÉ

Damage to renal tubular epithelial cells (RTECs) signaled the onset and progression of sepsis-associated acute kidney injury (SA-AKI). Recent research on mitochondria has revealed that mitophagy plays a crucial physiological role in alleviating injury to RTECs and it is suppressed progressively by the inflammation response in SA-AKI. However, the mechanism by which inflammation influences mitophagy remains poorly understood. We examined how macrophage migration inhibitory factor (MIF), a pro-inflammatory protein, influences the PINK1-Parkin pathway of mitophagy by studying protein-protein interactions when MIF was inhibited or overexpressed. Surprisingly, elevated levels of MIF were found to directly bind to PINK1, disrupting its interaction with Parkin. This interference hindered the recruitment of Parkin to mitochondria and impeded the initiation of mitophagy. Furthermore, this outcome led to significant apoptosis of RTECs, which could, however, be reversed by an MIF inhibitor ISO-1 and/or a new mitophagy activator T0467. These findings highlight the detrimental impact of MIF on renal damage through its disruption of the interaction between PINK1 and Parkin, and the therapeutic potential of ISO-1 and T0467 in mitigating SA-AKI. This study offers a fresh perspective on treating SA-AKI by targeting MIF and mitophagy.


Sujet(s)
Atteinte rénale aigüe , Facteurs inhibiteurs de la migration des macrophages , Mitophagie , Protein kinases , Sepsie , Ubiquitin-protein ligases , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Facteurs inhibiteurs de la migration des macrophages/génétique , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protein kinases/métabolisme , Sepsie/complications , Sepsie/métabolisme , Animaux , Humains , Mitochondries/métabolisme , Tubules rénaux/métabolisme , Tubules rénaux/anatomopathologie , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Apoptose , Liaison aux protéines , Mâle , Intramolecular oxidoreductases/métabolisme
8.
Cell Death Dis ; 15(7): 483, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969650

RÉSUMÉ

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors, and the expression and function of an uncharacterized protein RNF214 in HCC are still unknown. Phase separation has recently been observed to participate in the progression of HCC. In this study, we investigated the expression, function, and phase separation of RNF214 in HCC. We found that RNF214 was highly expressed in HCC and associated with poor prognosis. RNF214 functioned as an oncogene to promote the proliferation, migration, and metastasis of HCC. Mechanically, RNF214 underwent phase separation, and the coiled-coil (CC) domain of RNF214 mediated its phase separation. Furthermore, the CC domain was necessary for the oncogenic function of RNF214 in HCC. Taken together, our data favored that phase separation of RNF214 promoted the progression of HCC. RNF214 may be a potential biomarker and therapeutic target for HCC.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Évolution de la maladie , Tumeurs du foie , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Humains , Lignée cellulaire tumorale , Animaux , Mouvement cellulaire/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Mâle , Souris nude , Souris , Régulation de l'expression des gènes tumoraux , Femelle , Souris de lignée BALB C , Adulte d'âge moyen ,
9.
Vet Res ; 55(1): 84, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965634

RÉSUMÉ

Pseudorabies virus (PRV) has evolved multiple strategies to evade host antiviral responses to benefit virus replication and establish persistent infection. Recently, tripartite motif 26 (TRIM26), a TRIM family protein, has been shown to be involved in a broad range of biological processes involved in innate immunity, especially in regulating viral infection. Herein, we found that the expression of TRIM26 was significantly induced after PRV infection. Surprisingly, the overexpression of TRIM26 promoted PRV production, while the depletion of this protein inhibited virus replication, suggesting that TRIM26 could positively regulate PRV infection. Further analysis revealed that TRIM26 negatively regulates the innate immune response by targeting the RIG-I-triggered type I interferon signalling pathway. TRIM26 was physically associated with MAVS independent of viral infection and reduced MAVS expression. Mechanistically, we found that NDP52 interacted with both TRIM26 and MAVS and that TRIM26-induced MAVS degradation was almost entirely blocked in NDP52-knockdown cells, demonstrating that TRIM26 degrades MAVS through NDP52-mediated selective autophagy. Our results reveal a novel mechanism by which PRV escapes host antiviral innate immunity and provide insights into the crosstalk among virus infection, autophagy, and the innate immune response.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Autophagie , Immunité innée , Animaux , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Suidae , Réplication virale , Humains , Transduction du signal , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique
10.
J Cell Biol ; 223(10)2024 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-38967608

RÉSUMÉ

Peroxisomes are membrane-bound organelles harboring metabolic enzymes. In humans, peroxisomes are required for normal development, yet the genes regulating peroxisome function remain unclear. We performed a genome-wide CRISPRi screen to identify novel factors involved in peroxisomal homeostasis. We found that inhibition of RNF146, an E3 ligase activated by poly(ADP-ribose), reduced the import of proteins into peroxisomes. RNF146-mediated loss of peroxisome import depended on the stabilization and activity of the poly(ADP-ribose) polymerases TNKS and TNKS2, which bind the peroxisomal membrane protein PEX14. We propose that RNF146 and TNKS/2 regulate peroxisome import efficiency by PARsylation of proteins at the peroxisome membrane. Interestingly, we found that the loss of peroxisomes increased TNKS/2 and RNF146-dependent degradation of non-peroxisomal substrates, including the ß-catenin destruction complex component AXIN1, which was sufficient to alter the amplitude of ß-catenin transcription. Together, these observations not only suggest previously undescribed roles for RNF146 in peroxisomal regulation but also a novel role in bridging peroxisome function with Wnt/ß-catenin signaling during development.


Sujet(s)
Axine , Péroxysomes , Ubiquitin-protein ligases , Voie de signalisation Wnt , Péroxysomes/métabolisme , Péroxysomes/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Humains , Axine/métabolisme , Axine/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Cellules HEK293 , Transport des protéines , Systèmes CRISPR-Cas
11.
Nat Commun ; 15(1): 5604, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961054

RÉSUMÉ

The CRL4-DCAF15 E3 ubiquitin ligase complex is targeted by the aryl-sulfonamide molecular glues, leading to neo-substrate recruitment, ubiquitination, and proteasomal degradation. However, the physiological function of DCAF15 remains unknown. Using a domain-focused genetic screening approach, we reveal DCAF15 as an acute myeloid leukemia (AML)-biased dependency. Loss of DCAF15 results in suppression of AML through compromised replication fork integrity and consequent accumulation of DNA damage. Accordingly, DCAF15 loss sensitizes AML to replication stress-inducing therapeutics. Mechanistically, we discover that DCAF15 directly interacts with the SMC1A protein of the cohesin complex and destabilizes the cohesin regulatory factors PDS5A and CDCA5. Loss of PDS5A and CDCA5 removal precludes cohesin acetylation on chromatin, resulting in uncontrolled chromatin loop extrusion, defective DNA replication, and apoptosis. Collectively, our findings uncover an endogenous, cell autonomous function of DCAF15 in sustaining AML proliferation through post-translational control of cohesin dynamics.


Sujet(s)
Protéines du cycle cellulaire , Protéines chromosomiques nonhistones , , Altération de l'ADN , Réplication de l'ADN , Leucémie aigüe myéloïde , Protéines chromosomiques nonhistones/métabolisme , Protéines chromosomiques nonhistones/génétique , Humains , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Lignée cellulaire tumorale , Acétylation , Animaux , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Souris , Chromatine/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Apoptose , Prolifération cellulaire , Cellules HEK293
12.
Cell Death Dis ; 15(6): 460, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38942760

RÉSUMÉ

Lung cancer stands as the leading cause of mortality among all types of tumors, with over 40% of cases being lung adenocarcinoma (LUAD). Family with sequence similarity 83 member A (FAM83A) emerges as a notable focus due to its frequent overexpression in LUAD. Despite this, the precise role of FAM83A remains elusive. This study addresses this gap by unveiling the crucial involvement of FAM83A in maintaining the cancer stem cell-like (CSC-like) phenotype of LUAD. Through a global proteomics analysis, the study identifies human epidermal growth factor receptor 2 (HER2 or ErbB2) as a crucial target of FAM83A. Mechanistically, FAM83A facilitated ErbB2 expression at the posttranslational modification level via the E3 ubiquitin ligase STUB1 (STIP1-homologous U-Box containing protein 1). More importantly, the interaction between FAM83A and ErbB2 at Arg241 promotes calcineurin (CALN)-mediated dephosphorylation of ErbB2, followed by inhibition of STUB1-mediated ubiquitin-proteasomal ErbB2 degradation. The maintenance of the CSC-like phenotype by FAM83A, achieved through the posttranslational regulation of ErbB2, offers valuable insights for identifying potential therapeutic targets for LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du poumon , Protéines tumorales , Cellules souches tumorales , Phénotype , Récepteur ErbB-2 , Humains , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Protéines tumorales/métabolisme , Protéines tumorales/génétique , Animaux , Souris , Lignée cellulaire tumorale , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique
13.
J Nutr Sci Vitaminol (Tokyo) ; 70(3): 219-227, 2024.
Article de Anglais | MEDLINE | ID: mdl-38945887

RÉSUMÉ

This study investigated the protective effect of carnosine and its components (L-histidine and ß-alanine [HA]) against dexamethasone (Dex)-induced muscle atrophy in C2C12 myotubes. Myotubes were treated with Dex (10 µM) to induce muscle atrophy manifested by decreased myotube diameter, low myosin heavy chain content, and increased expression of muscle atrophy-associated ubiquitin ligases (Atrogin-1, MuRF-1, and Cbl-b). Carnosine (20 mM) treatment significantly improved the myotube diameter and MyHC protein expression level in Dex-treated C2C12 myotubes. It also downregulated the expression of Atrogin-1, MuRF-1, and Cbl-b and suppressed the expression of forkhead box O3 (FoxO3a) mediated by Dex. Furthermore, reactive oxygen species production was increased by Dex but was ameliorated by carnosine treatment. However, HA (20 mM), the component of carnosine, treatment was found ineffective in preventing Dex-induced protein damage. Therefore, based on above results it can be suggested that carnosine could be a potential therapeutic agent to prevent Dex-induced muscle atrophy compared to its components HA.


Sujet(s)
Carnosine , Dexaméthasone , Fibres musculaires squelettiques , Protéines du muscle , Amyotrophie , Espèces réactives de l'oxygène , SKP cullin F-box protein ligases , Carnosine/pharmacologie , Dexaméthasone/pharmacologie , Amyotrophie/induit chimiquement , Amyotrophie/prévention et contrôle , Amyotrophie/métabolisme , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/métabolisme , Animaux , Souris , Protéines du muscle/métabolisme , Lignée cellulaire , Espèces réactives de l'oxygène/métabolisme , SKP cullin F-box protein ligases/métabolisme , Ubiquitin-protein ligases/métabolisme , Protéine O3 à motif en tête de fourche/métabolisme , Protéines à motif tripartite/métabolisme , Chaînes lourdes de myosine/métabolisme
14.
J Exp Clin Cancer Res ; 43(1): 177, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926803

RÉSUMÉ

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


Sujet(s)
Autophagie , Protéine NEDD8 , Paclitaxel , Protéines à motif tripartite , Tumeurs du sein triple-négatives , Ubiquitin-protein ligases , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Femelle , Souris , Animaux , Autophagie/effets des médicaments et des substances chimiques , Protéine NEDD8/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Lignée cellulaire tumorale , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Cyclopentanes/pharmacologie , Résistance aux médicaments antinéoplasiques , Tests d'activité antitumorale sur modèle de xénogreffe , Ubiquitin-conjugating enzymes/métabolisme , Ubiquitin-conjugating enzymes/génétique
15.
Genes (Basel) ; 15(6)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38927660

RÉSUMÉ

The ring finger protein 213 gene (RNF213) is involved in several vascular diseases, both intracranial and systemic ones. Some variants are common in the Asian population and are reported as a risk factor for moyamoya disease, intracranial stenosis and intracranial aneurysms. Among intracranial vascular diseases, both moyamoya disease and intracranial artery dissection are more prevalent in the Asian population. We performed a systematic review of the literature, aiming to assess the rate of RNF213 variants in patients with spontaneous intracranial dissections. Four papers were identified, providing data on 53 patients with intracranial artery dissection. The rate of RNF213 variants is 10/53 (18.9%) and it increases to 10/29 (34.5%), excluding patients with vertebral artery dissection. All patients had the RNF213 p.Arg4810Lys variant. RNF213 variants seems to be involved in intracranial dissections in Asian cohorts. The small number of patients, the inclusion of only patients of Asian descent and the small but non-negligible coexistence with moyamoya disease familiarity might be limiting factors, requiring further studies to confirm these preliminary findings and the embryological interpretation.


Sujet(s)
Adenosine triphosphatases , Ubiquitin-protein ligases , Humains , Adenosine triphosphatases/génétique , /génétique , Asiatiques/génétique , Prédisposition génétique à une maladie , Anévrysme intracrânien/génétique , Maladie de Moya-Moya/génétique , Polymorphisme de nucléotide simple , Ubiquitin-protein ligases/génétique
16.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38928146

RÉSUMÉ

Mitochondrial quality control is essential in mitochondrial function. To examine the importance of Parkin-dependent mechanisms in mitochondrial quality control, we assessed the impact of modulating Parkin on proteome flux and mitochondrial function in a context of reduced mtDNA fidelity. To accomplish this, we crossed either the Parkin knockout mouse or ParkinW402A knock-in mouse lines to the Polg mitochondrial mutator line to generate homozygous double mutants. In vivo longitudinal isotopic metabolic labeling was followed by isolation of liver mitochondria and synaptic terminals from the brain, which are rich in mitochondria. Mass spectrometry and bioenergetics analysis were assessed. We demonstrate that slower mitochondrial protein turnover is associated with loss of mtDNA fidelity in liver mitochondria but not synaptic terminals, and bioenergetic function in both tissues is impaired. Pathway analysis revealed loss of mtDNA fidelity is associated with disturbances of key metabolic pathways, consistent with its association with metabolic disorders and neurodegeneration. Furthermore, we find that loss of Parkin leads to exacerbation of Polg-driven proteomic consequences, though it may be bioenergetically protective in tissues exhibiting rapid mitochondrial turnover. Finally, we provide evidence that, surprisingly, dis-autoinhibition of Parkin (ParkinW402A) functionally resembles Parkin knockout and fails to rescue deleterious Polg-driven effects. Our study accomplishes three main outcomes: (1) it supports recent studies suggesting that Parkin dependence is low in response to an increased mtDNA mutational load, (2) it provides evidence of a potential protective role of Parkin insufficiency, and (3) it draws into question the therapeutic attractiveness of enhancing Parkin function.


Sujet(s)
DNA Polymerase gamma , ADN mitochondrial , Souris knockout , Mutation , Ubiquitin-protein ligases , Animaux , DNA Polymerase gamma/génétique , DNA Polymerase gamma/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Souris , ADN mitochondrial/génétique , ADN mitochondrial/métabolisme , Protéomique/méthodes , Protéome/métabolisme , Mitochondries/métabolisme , Mitochondries/génétique , Mitochondries du foie/métabolisme , Mitochondries du foie/génétique , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique
17.
Mol Cell ; 84(12): 2337-2352.e9, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38870935

RÉSUMÉ

Ribosome assembly requires precise coordination between the production and assembly of ribosomal components. Mutations in ribosomal proteins that inhibit the assembly process or ribosome function are often associated with ribosomopathies, some of which are linked to defects in proteostasis. In this study, we examine the interplay between several yeast proteostasis enzymes, including deubiquitylases (DUBs) Ubp2 and Ubp14, and E3 ligases Ufd4 and Hul5, and we explore their roles in the regulation of the cellular levels of K29-linked unanchored polyubiquitin (polyUb) chains. Accumulating K29-linked unanchored polyUb chains associate with maturing ribosomes to disrupt their assembly, activate the ribosome assembly stress response (RASTR), and lead to the sequestration of ribosomal proteins at the intranuclear quality control compartment (INQ). These findings reveal the physiological relevance of INQ and provide insights into mechanisms of cellular toxicity associated with ribosomopathies.


Sujet(s)
Polyubiquitine , Protéines ribosomiques , Ribosomes , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Protéines ribosomiques/métabolisme , Protéines ribosomiques/génétique , Ribosomes/métabolisme , Ribosomes/génétique , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Polyubiquitine/métabolisme , Polyubiquitine/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitination , Homéostasie protéique , Noyau de la cellule/métabolisme
18.
Cell Death Dis ; 15(6): 446, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38914543

RÉSUMÉ

Protein homeostasis is predominantly governed through post-translational modification (PTM). UBE3B, identified as an oncoprotein, exhibits elevated protein levels in breast cancer. However, the impact of PTM on UBE3B remains unexplored. In this study, we show that VHL is a bona fide E3 ligase for UBE3B. Mechanistically, VHL directly binds to UBE3B, facilitating its lysine 48 (K48)-linked polyubiquitination at K286 and K427 in a prolyl hydroxylase (PHD)-independent manner. Consequently, this promotes the proteasomal degradation of UBE3B. The K286/427R mutation of UBE3B dramatically abolishes the inhibitory effect of VHL on breast tumor growth and lung metastasis. Additionally, the protein levels of UBE3B and VHL exhibit a negative correlation in breast cancer tissues. These findings delineate an important layer of UBE3B regulation by VHL.


Sujet(s)
Tumeurs du sein , Ubiquitin-protein ligases , Ubiquitination , Protéine Von Hippel-Lindau supresseur de tumeur , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Protéine Von Hippel-Lindau supresseur de tumeur/métabolisme , Protéine Von Hippel-Lindau supresseur de tumeur/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/secondaire , Métastase tumorale , Souris nude , Souris , Cellules HEK293 , Liaison aux protéines
19.
Phytomedicine ; 130: 155537, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38823344

RÉSUMÉ

BACKGROUND: Aberrant activation of autophagy in triple-negative breast cancer (TNBC) has led researchers to investigate potential therapeutic strategies targeting this process. The regulation of autophagy is significantly influenced by METTL3. Our previous research has shown that the Panax ginseng-derived compound, 20(R)-panaxatriol (PT), has potential as an anti-tumor agent. However, it remains unclear whether PT can modulate autophagy through METTL3 to exert its anti-tumor effects. OBJECTIVE: Our objective is to investigate whether PT can regulate autophagy in TNBC cells and elucidate the molecular mechanisms. STUDY DESIGN: For in vitro experiments, we employed SUM-159-PT and MDA-MB-231 cells. While in vivo experiments involved BALB/c nude mice and NOD/SCID mice. METHODS: In vitro, TNBC cells were treated with PT, and cell lines with varying expression levels of METTL3 were established. We assessed the impact on tumor cell activity and autophagy by analyzing autophagic flux, Western Blot (WB), and methylation levels. In vivo, subcutaneous transplantation models were established in BALB/c nude and NOD/SCID mice to observe the effect of PT on TNBC growth. HE staining and immunofluorescence were employed to analyze histopathological changes in tumor tissues. MeRIP-seq and dual-luciferase reporter gene assays were used to identify key downstream targets. Additionally, the silencing of STIP1 Homology And U-Box Containing Protein 1 (STUB1) explored PT's effects. The mechanism of PT's action on STUB1 via METTL3 was elucidated through mRNA stability assays, mRNA alternative splicing analysis, and nuclear-cytoplasmic mRNA separation. RESULTS: In both in vivo and in vitro experiments, it was discovered that PT significantly upregulates the expression of METTL3, leading to autophagy inhibition and therapeutic effects in TNBC. Simultaneously, through MeRIP-seq analysis and dual-luciferase reporter gene assays, we have demonstrated that PT modulates STUB1 via METTL3, influencing autophagy in TNBC cells. Furthermore, intriguingly, PT extends the half-life of STUB1 mRNA by enhancing its methylation modification, thereby enhancing its stability. CONCLUSION: In summary, our research reveals that PT increases STUB1 m6A modification through a METTL3-mediated mechanism in TNBC cells, inhibiting autophagy and further accentuating its anti-tumor properties. Our study provides novel mechanistic insights into TNBC pathogenesis and potential drug targets for TNBC.


Sujet(s)
Autophagie , Methyltransferases , Souris de lignée BALB C , Souris nude , Tumeurs du sein triple-négatives , Ubiquitin-protein ligases , Animaux , Tumeurs du sein triple-négatives/traitement médicamenteux , Humains , Autophagie/effets des médicaments et des substances chimiques , Femelle , Lignée cellulaire tumorale , Methyltransferases/métabolisme , Ubiquitin-protein ligases/métabolisme , Souris SCID , Souris de lignée NOD , Souris , Antinéoplasiques d'origine végétale/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Panax/composition chimique , Adénosine/analogues et dérivés , Adénosine/pharmacologie
20.
Eur J Endocrinol ; 190(6): 479-488, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38857188

RÉSUMÉ

OBJECTIVES: The etiology of central precocious puberty (CPP) has expanded with identification of new genetic causes, including the monogenic deficiency of Makorin-Ring-Finger-Protein-3 (MKRN3). We aimed to assess the prevalence of CPP causes and the predictors of genetic involvement in this phenotype. DESIGN: A retrospective cohort study for an etiological survey of patients with CPP from a single academic center. METHODS: All patients with CPP had detailed medical history, phenotyping, and brain magnetic resonance imaging (MRI); those with negative brain MRI (apparently idiopathic) were submitted to genetic studies, mainly DNA sequencing studies, genomic microarray, and methylation analysis. RESULTS: We assessed 270 patients with CPP: 50 (18.5%) had CPP-related brain lesions (34 [68%] congenital lesions), whereas 220 had negative brain MRI. Of the latter, 174 (165 girls) were included for genetic studies. Genetic etiologies were identified in 22 patients (20 girls), indicating an overall frequency of genetic CPP of 12.6% (22.2% in boys and 12.1% in girls). The most common genetic defects were MKRN3, Delta-Like-Non-Canonical-Notch-Ligand-1 (DLK1), and Methyl-CpG-Binding-Protein-2 (MECP2) loss-of-function mutations, followed by 14q32.2 defects (Temple syndrome). Univariate logistic regression identified family history (odds ratio [OR] 3.3; 95% CI 1.3-8.3; P = .01) and neurodevelopmental disorders (OR 4.1; 95% CI 1.3-13.5; P = .02) as potential clinical predictors of genetic CPP. CONCLUSIONS: Distinct genetic causes were identified in 12.6% patients with apparently idiopathic CPP, revealing the genetic etiology as a relevant cause of CPP in both sexes. Family history and neurodevelopmental disorders were suggested as predictors of genetic CPP. We originally proposed an algorithm to investigate the etiology of CPP including genetic studies.


Sujet(s)
Puberté précoce , Humains , Puberté précoce/génétique , Puberté précoce/étiologie , Puberté précoce/épidémiologie , Femelle , Mâle , Enfant , Études rétrospectives , Enfant d'âge préscolaire , Imagerie par résonance magnétique , Ribonucléoprotéines/génétique , Études de cohortes , Ubiquitin-protein ligases/génétique , Mutation , Encéphale/imagerie diagnostique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...