Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 233
Filtrer
1.
Cell Commun Signal ; 22(1): 302, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38831335

RÉSUMÉ

The ubiquitination-mediated protein degradation exerts a vital role in the progression of multiple tumors. NEDD4L, which belongs to the E3 ubiquitin ligase NEDD4 family, is related to tumor genesis, metastasis and drug resistance. However, the anti-tumor role of NEDD4L in esophageal carcinoma, and the potential specific recognition substrate remain unclear. Based on public esophageal carcinoma database and clinical sample data, it was discovered in this study that the expression of NEDD4L in esophageal carcinoma was apparently lower than that in atypical hyperplastic esophageal tissue and esophageal squamous epithelium. Besides, patients with high expression of NEDD4L in esophageal carcinoma tissue had longer progression-free survival than those with low expression. Experiments in vivo and in vitro also verified that NEDD4L suppressed the growth and metastasis of esophageal carcinoma. Based on co-immunoprecipitation and proteome analysis, the NEDD4L ubiquitination-degraded protein ITGB4 was obtained. In terms of the mechanism, the HECT domain of NEDD4L specifically bound to the Galx-ß domain of ITGB4, which modified the K915 site of ITGB4 in an ubiquitination manner, and promoted the ubiquitination degradation of ITGB4, thus suppressing the malignant phenotype of esophageal carcinoma.


Sujet(s)
Évolution de la maladie , Tumeurs de l'oesophage , Intégrine bêta4 , Ubiquitine protéine ligases NEDD4 , Protéolyse , Ubiquitination , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'oesophage/génétique , Humains , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Animaux , Lignée cellulaire tumorale , Intégrine bêta4/métabolisme , Intégrine bêta4/génétique , Souris nude , Souris , Prolifération cellulaire , Mâle , Régulation de l'expression des gènes tumoraux , Femelle
2.
Invest Ophthalmol Vis Sci ; 65(6): 29, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38888282

RÉSUMÉ

Purpose: Ubiquitination serves as a fundamental post-translational modification in numerous cellular events. Yet, its role in regulating corneal epithelial wound healing (CEWH) remains elusive. This study endeavored to determine the function and mechanism of ubiquitination in CEWH. Methods: Western blot and immunoprecipitation were used to discern ubiquitination alterations during CEWH in mice. Interventions, including neuronally expressed developmentally downregulated 4 (Nedd4) siRNA and proteasome/lysosome inhibitor, assessed their impact on CEWH. In vitro analyses, such as the scratch wound assay, MTS assay, and EdU staining, were conducted to gauge cell migration and proliferation in human corneal epithelial cells (HCECs). Moreover, transfection of miR-30/200 coupled with a luciferase activity assay ascertained their regulatory mechanism on Nedd4. Results: Global ubiquitination levels were markedly increased during the mouse CEWH. Importantly, the application of either proteasomal or lysosomal inhibitors notably impeded the healing process both in vivo and in vitro. Furthermore, Nedd4 was identified as an essential E3 ligase for CEWH. Nedd4 expression was significantly upregulated during CEWH. In vivo studies revealed that downregulation of Nedd4 substantially delayed CEWH, whereas further investigations underscored its role in regulating cell proliferation and migration, through the Stat3 pathway by targeting phosphatase and tensin homolog (PTEN). Notably, our findings pinpointed miR-30/200 family members as direct regulators of Nedd4. Conclusions: Ubiquitination holds pivotal significance in orchestrating CEWH. The critical E3 ligase Nedd4, under the regulatory purview of miR-30 and miR-200, facilitates CEWH through PTEN-mediated Stat3 signaling. This revelation sheds light on a prospective therapeutic target within the realm of CEWH.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Épithélium antérieur de la cornée , Ubiquitine protéine ligases NEDD4 , Phosphohydrolase PTEN , Ubiquitin-protein ligases , Ubiquitination , Cicatrisation de plaie , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Animaux , Souris , Mouvement cellulaire/physiologie , Prolifération cellulaire/physiologie , Cicatrisation de plaie/physiologie , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Épithélium antérieur de la cornée/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Humains , Souris de lignée C57BL , Complexes de tri endosomique requis pour le transport/métabolisme , Technique de Western , Facteur de transcription STAT-3/métabolisme , Cellules cultivées , Modèles animaux de maladie humaine , microARN/génétique , Immunoprécipitation , Mâle , Régulation de l'expression des gènes/physiologie
3.
Int J Biol Macromol ; 269(Pt 2): 131976, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38697427

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a chronic and lethal lung disease characterized by progressive lung scarring. This study aims to elucidate the role of the E3 ubiquitin ligase NEDD4 in the ubiquitination of YY1 and its subsequent impact on TAB1 transcription, revealing a possible molecular mechanism in the development of IPF. Through bioinformatics analysis and both in vitro and in vivo experiments, we observed differential expression levels of NEDD4 and YY1 between normal and IPF samples, identifying NEDD4 as an upstream E3 ubiquitin ligase of YY1. Furthermore, binding sites for the transcription factor YY1 on the promoter region of TAB1 were discovered, indicating a direct interaction. In vitro experiments using HEPF cells showed that NEDD4 mediates the ubiquitination and degradation of YY1, leading to suppressed TAB1 transcription, thereby inhibiting cell proliferation and fibrogenesis. These findings were corroborated by in vivo experiments in an IPF mouse model, where the ubiquitination pathway facilitated by NEDD4 attenuated IPF progression through the downregulation of YY1 and TAB1 transcription. These results suggest that NEDD4 plays a crucial role in the development of IPF by modulating YY1 ubiquitination and TAB1 transcription, providing new insights into potential therapeutic targets for treating IPF.


Sujet(s)
Fibrose pulmonaire idiopathique , Ubiquitine protéine ligases NEDD4 , Ubiquitination , Facteur de transcription YY1 , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Facteur de transcription YY1/métabolisme , Facteur de transcription YY1/génétique , Humains , Animaux , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/génétique , Souris , Prolifération cellulaire , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Modèles animaux de maladie humaine , Mâle
4.
Biomolecules ; 14(5)2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38785984

RÉSUMÉ

Protein ubiquitination is an enzymatic cascade reaction and serves as an important protein post-translational modification (PTM) that is involved in the vast majority of cellular life activities. The key enzyme in the ubiquitination process is E3 ubiquitin ligase (E3), which catalyzes the binding of ubiquitin (Ub) to the protein substrate and influences substrate specificity. In recent years, the relationship between the subfamily of neuron-expressed developmental downregulation 4 (NEDD4), which belongs to the E3 ligase system, and digestive diseases has drawn widespread attention. Numerous studies have shown that NEDD4 and NEDD4L of the NEDD4 family can regulate the digestive function, as well as a series of related physiological and pathological processes, by controlling the subsequent degradation of proteins such as PTEN, c-Myc, and P21, along with substrate ubiquitination. In this article, we reviewed the appropriate functions of NEDD4 and NEDD4L in digestive diseases including cell proliferation, invasion, metastasis, chemotherapeutic drug resistance, and multiple signaling pathways, based on the currently available research evidence for the purpose of providing new ideas for the prevention and treatment of digestive diseases.


Sujet(s)
Ubiquitine protéine ligases NEDD4 , Ubiquitination , Humains , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Maladies de l'appareil digestif/métabolisme , Maladies de l'appareil digestif/anatomopathologie , Animaux , Transduction du signal , Prolifération cellulaire , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique
5.
J Transl Med ; 22(1): 465, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755664

RÉSUMÉ

Disturbance in mitochondrial homeostasis within proximal tubules is a critical characteristic associated with diabetic kidney disease (DKD). CaMKKß/AMPK signaling plays an important role in regulating mitochondrial homeostasis. Despite the downregulation of CaMKKß in DKD pathology, the underlying mechanism remains elusive. The expression of NEDD4L, which is primarily localized to renal proximal tubules, is significantly upregulated in the renal tubules of mice with DKD. Coimmunoprecipitation (Co-IP) assays revealed a physical interaction between NEDD4L and CaMKKß. Moreover, deletion of NEDD4L under high glucose conditions prevented rapid CaMKKß protein degradation. In vitro studies revealed that the aberrant expression of NEDD4L negatively influences the protein stability of CaMKKß. This study also explored the role of NEDD4L in DKD by using AAV-shNedd4L in db/db mice. These findings confirmed that NEDD4L inhibition leads to a decrease in urine protein excretion, tubulointerstitial fibrosis, and oxidative stress, and mitochondrial dysfunction. Further in vitro studies demonstrated that si-Nedd4L suppressed mitochondrial fission and reactive oxygen species (ROS) production, effects antagonized by si-CaMKKß. In summary, the findings provided herein provide strong evidence that dysregulated NEDD4L disturbs mitochondrial homeostasis by negatively modulating CaMKKß in the context of DKD. This evidence underscores the potential of therapeutic interventions targeting NEDD4L and CaMKKß to safeguard renal tubular function in the management of DKD.


Sujet(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase , Néphropathies diabétiques , Régulation négative , Homéostasie , Mitochondries , Ubiquitine protéine ligases NEDD4 , Animaux , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Mitochondries/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Kinase/métabolisme , Souris de lignée C57BL , Souris , Humains , Espèces réactives de l'oxygène/métabolisme , Mâle , Stress oxydatif , Dynamique mitochondriale , Stabilité protéique , Protéolyse
6.
Mol Med ; 30(1): 69, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38783226

RÉSUMÉ

BACKGROUND: The Enoyl-CoA hydratase/isomerase family plays a crucial role in the metabolism of tumors, being crucial for maintaining the energy balance and biosynthetic needs of cancer cells. However, the enzymes within this family that are pivotal in gastric cancer (GC) remain unclear. METHODS: We employed bioinformatics techniques to identify key Enoyl-CoA hydratase/isomerase in GC. The expression of ECHDC2 and its clinical significance were validated through tissue microarray analysis. The role of ECHDC2 in GC was further assessed using colony formation assays, CCK8 assay, EDU assay, Glucose and lactic acid assay, and subcutaneous tumor experiments in nude mice. The mechanism of action of ECHDC2 was validated through Western blotting, Co-immunoprecipitation, and immunofluorescence experiments. RESULTS: Our analysis of multiple datasets indicates that low expression of ECHDC2 in GC is significantly associated with poor prognosis. Overexpression of ECHDC2 notably inhibits aerobic glycolysis and proliferation of GC cells both in vivo and in vitro. Further experiments revealed that overexpression of ECHDC2 suppresses the P38 MAPK pathway by inhibiting the protein level of MCCC2, thereby restraining glycolysis and proliferation in GC cells. Ultimately, it was discovered that ECHDC2 promotes the ubiquitination and subsequent degradation of MCCC2 protein by binding with NEDD4. CONCLUSIONS: These findings underscore the pivotal role of the ECHDC2 in regulating aerobic glycolysis and proliferation in GC cells, suggesting ECHDC2 as a potential therapeutic target in GC.


Sujet(s)
Prolifération cellulaire , Ubiquitine protéine ligases NEDD4 , Tumeurs de l'estomac , Animaux , Femelle , Humains , Mâle , Souris , Lignée cellulaire tumorale , Énoyl-CoA hydratases/métabolisme , Énoyl-CoA hydratases/génétique , Régulation de l'expression des gènes tumoraux , Glycolyse , Souris nude , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Liaison aux protéines , Protéolyse , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Ubiquitination , Effet Warburg en oncologie
7.
Adv Sci (Weinh) ; 11(21): e2309002, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38569496

RÉSUMÉ

Preeclampsia (PE) is considered as a disease of placental origin. However, the specific mechanism of placental abnormalities remains elusive. This study identified thrombospondin-1 (THBS1) is downregulated in preeclamptic placentae and negatively correlated with blood pressure. Functional studies show that THBS1 knockdown inhibits proliferation, migration, and invasion and increases the cycle arrest and apoptosis rate of HTR8/SVneo cells. Importantly, THBS1 silencing induces necroptosis in HTR8/SVneo cells, accompanied by the release of damage-associated molecular patterns (DAMPs). Necroptosis inhibitors necrostatin-1 and GSK'872 restore the trophoblast survival while pan-caspase inhibitor Z-VAD-FMK has no effect. Mechanistically, the results show that THBS1 interacts with transforming growth factor B-activated kinase 1 (TAK1), which is a central modulator of necroptosis quiescence and affects its stability. Moreover, THBS1 silencing up-regulates the expression of neuronal precursor cell-expressed developmentally down-regulated 4 (NEDD4), which acts as an E3 ligase of TAK1 and catalyzes K48-linked ubiquitination of TAK1 in HTR8/SVneo cells. Besides, THBS1 attenuates PE phenotypes and improves the placental necroptosis in vivo. Taken together, the down-regulation of THBS1 destabilizes TAK1 by activating NEDD4-mediated, K48-linked TAK1 ubiquitination and promotes necroptosis and DAMPs release in trophoblast cells, thus participating in the pathogenesis of PE.


Sujet(s)
MAP Kinase Kinase Kinases , Nécroptose , Ubiquitine protéine ligases NEDD4 , Pré-éclampsie , Thrombospondine-1 , Trophoblastes , Ubiquitination , Humains , Pré-éclampsie/métabolisme , Pré-éclampsie/génétique , Femelle , Grossesse , Trophoblastes/métabolisme , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Nécroptose/génétique , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Thrombospondine-1/métabolisme , Thrombospondine-1/génétique , Adulte , Placenta/métabolisme
8.
Mol Syst Biol ; 20(6): 676-701, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38664594

RÉSUMÉ

Splice-switching oligonucleotides (SSOs) are antisense compounds that act directly on pre-mRNA to modulate alternative splicing (AS). This study demonstrates the value that artificial intelligence/machine learning (AI/ML) provides for the identification of functional, verifiable, and therapeutic SSOs. We trained XGboost tree models using splicing factor (SF) pre-mRNA binding profiles and spliceosome assembly information to identify modulatory SSO binding sites on pre-mRNA. Using Shapley and out-of-bag analyses we also predicted the identity of specific SFs whose binding to pre-mRNA is blocked by SSOs. This step adds considerable transparency to AI/ML-driven drug discovery and informs biological insights useful in further validation steps. We applied this approach to previously established functional SSOs to retrospectively identify the SFs likely to regulate those events. We then took a prospective validation approach using a novel target in triple negative breast cancer (TNBC), NEDD4L exon 13 (NEDD4Le13). Targeting NEDD4Le13 with an AI/ML-designed SSO decreased the proliferative and migratory behavior of TNBC cells via downregulation of the TGFß pathway. Overall, this study illustrates the ability of AI/ML to extract actionable insights from RNA-seq data.


Sujet(s)
Épissage alternatif , Intelligence artificielle , Apprentissage machine , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/génétique , Lignée cellulaire tumorale , Ubiquitine protéine ligases NEDD4/génétique , Ubiquitine protéine ligases NEDD4/métabolisme , Précurseurs des ARN/génétique , Précurseurs des ARN/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Facteurs d'épissage des ARN/génétique , Facteurs d'épissage des ARN/métabolisme , Oligonucléotides antisens/génétique , Mouvement cellulaire/génétique , Splicéosomes/métabolisme , Splicéosomes/génétique , Oligonucléotides/génétique , Femelle
9.
CNS Neurosci Ther ; 30(4): e14685, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38634270

RÉSUMÉ

OBJECTIVE: Neuronal precursor cells expressed developmentally down-regulated 4 (Nedd4) are believed to play a critical role in promoting the degradation of substrate proteins and are involved in numerous biological processes. However, the role of Nedd4 in intracerebral hemorrhage (ICH) remains unknown. This study aims to investigate the regulatory role of Nedd4 in the ICH model. METHODS: Male C57BL/6J mice were induced with ICH. Subsequently, the levels of glutathione peroxidase 4 (GPX4), malondialdehyde (MDA) concentration, iron content, mitochondrial morphology, as well as the expression of divalent metal transporter 1 (DMT1) and Nedd4 were assessed after ICH. Furthermore, the impact of Nedd4 overexpression was evaluated through analyses of hematoma area, ferroptosis, and neurobehavioral function. The mechanism underlying Nedd4-mediated degradation of DMT1 was elecidated using immunoprecipitation (IP) after ICH. RESULTS: Upon ICH, the level of DMT1 in the brain increased, but decreased when Nedd4 was overexpressed using Lentivirus, suggesting a negative correlation between Nedd4 and DMT1. Additionally, the degradation of DMT1 was inhibited after ICH. Furthermore, it was found that Nedd4 can interact with and ubiquitinate DMT1 at lysine residues 6, 69, and 277, facilitating the degradation of DMT1. Functional analysis indicated that overexpression of Nedd4 can alleviate ferroptosis and promote recovery following ICH. CONCLUSION: The results demonstrated that ferroptosis occurs via the Nedd4/DMT1 pathway during ICH, suggesting it potential as a valuable target to inhibit ferroptosis for the treatment of ICH.


Sujet(s)
Transporteurs de cations , Hémorragie cérébrale , Ferroptose , Ubiquitine protéine ligases NEDD4 , Animaux , Mâle , Souris , Encéphale/métabolisme , Hémorragie cérébrale/métabolisme , Hémorragie cérébrale/anatomopathologie , Ferroptose/génétique , Souris de lignée C57BL , Ubiquitination , Ubiquitine protéine ligases NEDD4/métabolisme , Transporteurs de cations/métabolisme
10.
Virology ; 595: 110056, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38552409

RÉSUMÉ

The Peste des petits ruminant virus (PPRV) is a member of the Paramyxoviridae family and is classified into the genus Measles virus. PPRV predominantly infects small ruminants, leading to mortality rates of nearly 100%, which have caused significant economic losses in developing countries. Host proteins are important in virus replication, but the PPRV nucleocapsid (N) protein-host interacting partners for regulating PPRV replication remain unclear. The present study confirmed the interaction between PPRV-N and the host protein vimentin by co-immunoprecipitation and co-localization experiments. Overexpression of vimentin suppressed PPRV replication, whereas vimentin knockdown had the opposite effect. Mechanistically, N was subjected to degradation via the ubiquitin/proteasome pathway, where vimentin recruits the E3 ubiquitin ligase NEDD4L to fulfill N-ubiquitination, resulting in the degradation of the N protein. These findings suggest that the host protein vimentin and E3 ubiquitin ligase NEDD4L have an anti-PPRV effect.


Sujet(s)
Protéines nucléocapside , Virus de la peste des petits ruminants , Vimentine , Réplication virale , Protéines nucléocapside/métabolisme , Protéines nucléocapside/génétique , Vimentine/métabolisme , Vimentine/génétique , Animaux , Virus de la peste des petits ruminants/physiologie , Virus de la peste des petits ruminants/génétique , Virus de la peste des petits ruminants/métabolisme , Humains , Ubiquitination , Interactions hôte-pathogène , Cellules HEK293 , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Lignée cellulaire , Peste des petits ruminants/virologie , Peste des petits ruminants/métabolisme , Liaison aux protéines
11.
Mol Carcinog ; 63(5): 803-816, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38411267

RÉSUMÉ

Ovarian cancer is a major cause of death among cancer patients. Recent research has shown that the transmembrane emp24 domain (TMED) protein family plays a role in the progression of various types of cancer. In this study, we investigated the expression of TMED3 in ovarian cancer tumors compared to nontumor tissues using immunohistochemical staining. We found that TMED3 was overexpressed in ovarian cancer tumors, and its high expression was associated with poor disease-free and overall survival. To understand the functional implications of TMED3 overexpression in ovarian cancer, we conducted experiments to knockdown TMED3 using short hairpin RNA (shRNA). We observed that TMED3 knockdown resulted in reduced cell viability and migration, as well as increased cell apoptosis. Additionally, in subcutaneous xenograft models in BALB-c nude mice, TMED3 knockdown inhibited tumor growth. Further investigation revealed that SMAD family member 2 (SMAD2) was a downstream target of TMED3, driving ovarian cancer progression. TMED3 stabilized SMAD2 by inhibiting the E3 ligase NEDD4-mediated ubiquitination of SMAD2. To confirm the importance of SMAD2 in TMED3-mediated ovarian cancer, we performed functional rescue experiments and found that SMAD2 played a critical role in this process. Moreover, we discovered that the PI3K-AKT pathway was involved in the promoting effects of TMED3 overexpression on ovarian cancer cells. Overall, our study identifies TMED3 as a prognostic indicator and tumor promoter in ovarian cancer. Its function is likely mediated through the regulation of the SMAD2 and PI3K-AKT signaling pathway. These findings contribute to our understanding of the molecular mechanisms underlying ovarian cancer progression and provide potential targets for therapeutic intervention.


Sujet(s)
Tumeurs de l'ovaire , Protéines du transport vésiculaire , Animaux , Femelle , Humains , Souris , Lignée cellulaire tumorale , Prolifération cellulaire , Souris nude , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Petit ARN interférent/métabolisme , Protéine Smad2/génétique , Protéine Smad2/métabolisme , Protéine Smad2/pharmacologie , Ubiquitination , Protéines du transport vésiculaire/génétique , Protéines du transport vésiculaire/métabolisme , Ubiquitine protéine ligases NEDD4/métabolisme
12.
J Biol Chem ; 300(3): 105715, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38309503

RÉSUMÉ

NEDD4L is a HECT-type E3 ligase that catalyzes the addition of ubiquitin to intracellular substrates such as the cardiac voltage-gated sodium channel, NaV1.5. The intramolecular interactions of NEDD4L regulate its enzymatic activity which is essential for proteostasis. For NaV1.5, this process is critical as alterations in Na+ current is involved in cardiac diseases including arrhythmias and heart failure. In this study, we perform extensive biochemical and functional analyses that implicate the C2 domain and the first WW-linker (1,2-linker) in the autoregulatory mechanism of NEDD4L. Through in vitro and electrophysiological experiments, the NEDD4L 1,2-linker was determined to be important in substrate ubiquitination of NaV1.5. We establish the preferred sites of ubiquitination of NEDD4L to be in the second WW-linker (2,3-linker). Interestingly, NEDD4L ubiquitinates the cytoplasmic linker between the first and second transmembrane domains of the channel (DI-DII) of NaV1.5. Moreover, we design a genetically encoded modulator of Nav1.5 that achieves Na+ current reduction using the NEDD4L HECT domain as cargo of a NaV1.5-binding nanobody. These investigations elucidate the mechanisms regulating the NEDD4 family and furnish a new molecular framework for understanding NaV1.5 ubiquitination.


Sujet(s)
Complexes de tri endosomique requis pour le transport , Canal sodique voltage-dépendant NAV1.5 , Ubiquitine protéine ligases NEDD4 , Ubiquitination , Complexes de tri endosomique requis pour le transport/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine/métabolisme , Humains , Canal sodique voltage-dépendant NAV1.5/métabolisme , Cellules HEK293
13.
Acta Pharmacol Sin ; 45(4): 831-843, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38052867

RÉSUMÉ

Chronic rhinosinusitis with nasal polyp (CRSwNP) is a refractory inflammatory disease with epithelial-mesenchymal transition (EMT) as one of the key features. Since ubiquitin modification has been shown to regulate the EMT process in other diseases, targeting ubiquitin ligases may be a potential strategy for the treatment of CRSwNP. In this study we investigated whether certain E3 ubiquitin ligases could regulate the EMT process in CRSwNP, and whether these regulations could be the potential drug targets as well as the underlying mechanisms. After screening the potential drug target by bioinformatic analyses, the expression levels of three potential E3 ubiquitin ligases were compared among the control, eosinophilic nasal polyp (ENP) and non-eosinophilic nasal polyp (NENP) group in clinical samples, and the significant decrement of the expression level of NEDD4L was found. Then, IP-MS, bioinformatics and immunohistochemistry studies suggested that low NEDD4L expression may be associated with the EMT process. In human nasal epithelial cells (hNECs) and human nasal epithelial cell line RPMI 2650, knockdown of NEDD4L promoted EMT, while upregulating NEDD4L reversed this effect, suggesting that NEDD4L inhibited EMT in nasal epithelial cells. IP-MS and Co-IP studies revealed that NEDD4L mediated the degradation of DDR1. We demonstrated that NEDD4L inhibited the ß-catenin/HIF-1α positive feedback loop either directly (degrading ß-catenin and HIF-1α) or indirectly (mediating DDR1 degradation). These results were confirmed in a murine NP model in vivo. This study for the first time reveals the regulatory role of ubiquitin in the EMT process of nasal epithelial cells, and identifies a novel drug target NEDD4L, which has promising efficacy against both ENP and NENP by suppressing ß-catenin/HIF-1α positive feedback loop.


Sujet(s)
Transition épithélio-mésenchymateuse , Thérapie moléculaire ciblée , Polypes du nez , Ubiquitine protéine ligases NEDD4 , , Animaux , Humains , Souris , bêta-Caténine/métabolisme , Maladie chronique , Rétroaction , Polypes du nez/traitement médicamenteux , Polypes du nez/enzymologie , /traitement médicamenteux , /enzymologie , Ubiquitines/métabolisme , Ubiquitine protéine ligases NEDD4/antagonistes et inhibiteurs , Ubiquitine protéine ligases NEDD4/métabolisme
14.
Physiology (Bethesda) ; 39(1): 18-29, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-37962894

RÉSUMÉ

The Nedd4 family of E3 ubiquitin ligases, consisting of a C2-WW(n)-HECT domain architecture, includes the closely related Nedd4/Nedd4-1 and Nedd4L/Nedd4-2, which play critical roles in human physiology and pathophysiology.This review focuses on the regulation of enzymatic activity of these Nedd4 proteins, as well as on their roles in regulating stability and function of membrane and other signaling proteins, such as ion channels, ion transporters, and growth factor receptors. The diseases caused by impairment of such regulation are discussed, as well as opportunities and challenges for targeting these enzymes for therapy.


Sujet(s)
Complexes de tri endosomique requis pour le transport , Ubiquitine , Humains , Complexes de tri endosomique requis pour le transport/génétique , Complexes de tri endosomique requis pour le transport/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitination
15.
J Biol Chem ; 300(1): 105593, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38145746

RÉSUMÉ

Neural precursor cell expressed developmentally downregulated 4 (NEDD4), an E3 ubiquitin ligase, is commonly upregulated in human hepatocellular carcinoma (HCC) and functions as an oncogenic factor in the progression of HCC, but the molecular mechanism needs be further explored. In this study, we found that NEDD4 could facilitate the proliferation of HCC cells, which was associated with regulating the ERK signaling. Further investigation showed that protocadherin 17 (PCDH17) was a potential substrate of NEDD4, and restoration of PCDH17 could block the facilitation of ERK signaling and HCC cells proliferation induced by NEDD4 overexpression. Whereafter, we confirmed that NEDD4 interacted with PCDH17 and promoted the Lys33-linked polyubiquitination and degradation of it via the proteasome pathway. Finally, NEDD4 protein level was found to be inversely correlated with that of PCDH17 in human HCC tissues. In conclusion, these results suggest that NEDD4 acts as an E3 ubiquitin ligase for PCDH17 ubiquitination and degradation thereby promoting the proliferation of HCC cells through regulating the ERK signaling, which may provide novel evidence for NEDD4 to be a promising therapeutic target for HCC.


Sujet(s)
Cadhérines , Carcinome hépatocellulaire , Tumeurs du foie , Ubiquitine protéine ligases NEDD4 , Humains , Carcinome hépatocellulaire/anatomopathologie , Prolifération cellulaire , Tumeurs du foie/anatomopathologie , Ubiquitine protéine ligases NEDD4/génétique , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitination , Cadhérines/métabolisme
16.
Cell Biol Int ; 48(3): 325-333, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38108119

RÉSUMÉ

Deoxyribonuclease 1-like 3 (DNASE1L3) has been shown to play nonnegligible roles in several types of carcinomas. Nevertheless, the biological function, clinical relevance, and influence of DNASE1L3 in colorectal cancer (CRC) remain obscure. Immunohistochemistry was adopted to examine DNASE1L3 and CDKN1A expression in CRC tissue, and the clinical significance of DNASE1L3 was assessed. Cell counting kit-8, colony formation, and transwell assays were employed for assessing tumor proliferation and migration. The mechanisms underlying the impact of DNASE1L3 were explored via western blot analysis, co-immunoprecipitation, and ubiquitination assay. It was observed that DNASE1L3 was downregulated in CRC tissues and was tightly associated with patient prognosis. DNASE1L3 impaired CRC cell proliferation and migration through elevating CDKN1A via suppressing CDKN1A ubiquitination. Meanwhile, DNASE1L3 was positively related to CDKN1A. In mechanism, DNASE1L3 and CDKN1A interacted with the E3 ubiquitin ligase NEDD4. Moreover, DNASE1L3 was competitively bound to NEDD4, thus repressing NEDD4-mediated CDKN1A ubiquitination and degradation. These discoveries implied the potential mechanisms of DNASE1L3 during tumorigenesis, suggesting that DNASE1L3 may serve as a new potential therapeutic agent for CRC.


Sujet(s)
Tumeurs colorectales , Ubiquitin-protein ligases , Humains , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Désoxyribonucléases/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitination
17.
Immunol Lett ; 264: 36-45, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37940007

RÉSUMÉ

BACKGROUND: Intracerebral hemorrhage (ICH) is a serious medical problem, and promising strategy is limited. Macrophage initiated brain inflammatory injury following ICH, but the molecular mechanism had not been well identified. E3 ligase Nedd4L is implicated in the pathogenesis of the inflammatory immune response. METHODS: In the present study, we detected the levels of Nedd4L in macrophages following ICH. Furthermore, Macrophage M1 polarization, pro-inflammatory cytokine production, BBB disruption, brain water content and neurological function were examined in ICH mice. RESULTS: Here, we demonstrated that E3 ligase Nedd4L levels of macrophage increased following ICH, promoted M1 polarization inflammation by TRAF3. Nedd4L promoted BBB disruption, as well as neurological deficits. Inhibition of Nedd4L significantly attenuated M1 polarization in vivo. Inhibition of Nedd4L decreased TRAF3 and TBK1 levels, and subsequent phosphorylation of p38 and NF-κB p65 subunit following ICH. CONCLUSIONS: Our data demonstrated that Nedd4L was involved in the pathogenesis of ICH, which promoted inflammatory responses and exacerbated brain damage by TRAF3 following ICH.


Sujet(s)
Encéphale , Hémorragie cérébrale , Ubiquitine protéine ligases NEDD4 , Facteur-3 associé aux récepteurs de TNF , Animaux , Souris , Encéphale/immunologie , Encéphale/anatomopathologie , Hémorragie cérébrale/immunologie , Hémorragie cérébrale/anatomopathologie , Macrophages/enzymologie , Macrophages/immunologie , Transduction du signal/physiologie , Facteur-3 associé aux récepteurs de TNF/métabolisme , Ubiquitine protéine ligases NEDD4/métabolisme
18.
J Nutr Biochem ; 120: 109413, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37423323

RÉSUMÉ

The ubiquitin-proteasomal pathway regulates the functional expression of many membrane transporters in a variety of cellular systems. Nothing is currently known about the role of ubiquitin E3 ligase, neural precursor cell-expressed developmentally down-regulated gene 4 (Nedd4-1) and the proteasomal degradation pathway in regulating human vitamin C transporter-2 (hSVCT2) in neuronal cells. hSVCT2 mediates the uptake of ascorbic acid (AA) and is the predominantly expressed vitamin C transporter isoform in neuronal systems. Therefore, we addressed this knowledge gap in our study. Analysis of mRNA revealed markedly higher expression of Nedd4-1 in neuronal samples than that of Nedd4-2. Interestingly, Nedd4-1 expression in the hippocampus was higher in patients with Alzheimer's disease (AD) and age-dependently increased in the J20 mouse model of AD. The interaction of Nedd4-1 and hSVCT2 was confirmed by coimmunoprecipitation and colocalization. While the coexpression of Nedd4-1 with hSVCT2 displayed a significant decrease in AA uptake, siRNA-mediated knockdown of Nedd4-1 expression up-regulated the AA uptake. Further, we mutated a classical Nedd4 protein interacting motif ("PPXY") within the hSVCT2 polypeptide and observed markedly decreased AA uptake due to the intracellular localization of the mutated hSVCT2. Also, we determined the role of the proteasomal degradation pathway in hSVCT2 functional expression in SH-SY5Y cells and the results indicated that the proteasomal inhibitor (MG132) significantly up-regulated the AA uptake and hSVCT2 protein expression level. Taken together, our findings show that the regulation of hSVCT2 functional expression is at least partly mediated by the Nedd4-1 dependent ubiquitination and proteasomal pathways.


Sujet(s)
Neuroblastome , Transporteurs de vitamine C couplés au sodium , Animaux , Humains , Souris , Acide ascorbique/pharmacologie , Acide ascorbique/métabolisme , Complexes de tri endosomique requis pour le transport/génétique , Complexes de tri endosomique requis pour le transport/métabolisme , Cellules épithéliales/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Ubiquitine protéine ligases NEDD4/métabolisme , Transporteurs de vitamine C couplés au sodium/génétique , Transporteurs de vitamine C couplés au sodium/métabolisme , Ubiquitine/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitination
19.
Cell Biol Int ; 47(10): 1688-1701, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37415495

RÉSUMÉ

Neural precursor cell-expressed developmentally downregulated gene 4 (NEDD4) is an E3 ubiquitin ligase that recognizes substrates via protein-protein interactions and takes part in tumor development. This study aims to clarify NEDD4's functions in diffuse large B-cell lymphoma (DLBCL) and its downstream mechanisms. Collection of 53 DLBCL tissues and adjacent normal lymphoid tissues, and detection of NEDD4 and Forkhead box protein A1 (FOXA1) in the tissues were conducted. The selection of DLBCL cells was for FARAGE, and test of cells' advancement was after transfection. Analysis of NEDD4 and FOXA1's link, and test of Wnt/ß-catenin pathway were implemented. In vivo tumor xenograft experiments were put into effect. Detection of the pathological conditions of tumor tissues and the positive Ki67 in the family was implemented. It came out NEDD4 was reduced in DLBCL tissues and cell lines, and FOXA1 was elevated; Enhancing NEDD4 or repressing FOXA1 refrained DLBCL cells' advancement; NEDD4 could combine with FOXA1 and trigger its ubiquitination and degradation; NEDD4 inactivates the Wnt/ß-catenin pathway by motivating FOXA1 ubiquitination; NEDD4 enhancement refrained DLBCL growth in vivo. In conclusion, the E3 ubiquitin ligase NEDD4 accelerates FOXA1 ubiquitination but refrains DLBCL cell proliferation via the Wnt/ß-Catenin pathway.


Sujet(s)
Lymphome B diffus à grandes cellules , Ubiquitin-protein ligases , Humains , bêta-Caténine/métabolisme , Prolifération cellulaire , Complexes de tri endosomique requis pour le transport/génétique , Complexes de tri endosomique requis pour le transport/métabolisme , Facteur nucléaire hépatocytaire HNF-3 alpha/génétique , Facteur nucléaire hépatocytaire HNF-3 alpha/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitination
20.
Biochem Pharmacol ; 214: 115641, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37307883

RÉSUMÉ

Accumulating evidence has demonstrated that NEDD4 E3 ubiquitin ligase family plays a pivotal oncogenic role in a variety of malignancies via mediating ubiquitin dependent degradation processes. Moreover, aberrant expression of NEDD4 E3 ubiquitin ligases is often indicative of cancer progression and correlated with poor prognosis. In this review, we are going to address association of expression of NEDD4 E3 ubiquitin ligases with cancers, the signaling pathways and the molecular mechanisms by which the NEDD4 E3 ubiquitin ligases regulate oncogenesis and progression, and the therapies targeting the NEDD4 E3 ubiquitin ligases. This review provides the systematic and comprehensive summary of the latest research status of E3 ubiquitin ligases in the NEDD4 subfamily, and proposes that NEDD4 family E3 ubiquitin ligases are promising anti-cancer drug targets, aiming to provide research direction for clinical targeting of NEDD4 E3 ubiquitin ligase therapy.


Sujet(s)
Complexes de tri endosomique requis pour le transport , Tumeurs , Humains , Ubiquitination , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitin-protein ligases/métabolisme , Tumeurs/traitement médicamenteux , Ubiquitine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...