Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Pharmacol Sin ; 45(4): 831-843, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38052867

ABSTRACT

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.


Subject(s)
Epithelial-Mesenchymal Transition , Molecular Targeted Therapy , Nasal Polyps , Nedd4 Ubiquitin Protein Ligases , Rhinosinusitis , Animals , Humans , Mice , beta Catenin/metabolism , Chronic Disease , Feedback , Nasal Polyps/drug therapy , Nasal Polyps/enzymology , Rhinosinusitis/drug therapy , Rhinosinusitis/enzymology , Ubiquitins/metabolism , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Nedd4 Ubiquitin Protein Ligases/metabolism
2.
Cell Chem Biol ; 28(1): 14-25.e9, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33176158

ABSTRACT

The benzdiimidazole NAB2 rescues α-synuclein-associated trafficking defects associated with early onset Parkinson's disease in a Nedd4-dependent manner. Despite identification of E3 ubiquitin ligase Nedd4 as a putative target of NAB2, its molecular mechanism of action has not been elucidated. As such, the effect of NAB2 on Nedd4 activity and specificity was interrogated through biochemical, biophysical, and proteomic analyses. NAB2 was found to bind Nedd4 (KDapp = 42 nM), but this binding is side chain mediated and does not alter its conformation or ubiquitination kinetics in vitro. Nedd4 co-localizes with trafficking organelles, and NAB2 exposure did not alter its co-localization. Ubiquitin enrichment coupled proteomics revealed that NAB2 stimulates ubiquitination of trafficking-associated proteins, most likely through modulating the substrate specificity of Nedd4, providing a putative protein network involved in the NAB2 mechanism and revealing trafficking scaffold protein TFG as a Nedd4 substrate.


Subject(s)
Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Parkinson Disease/drug therapy , Small Molecule Libraries/pharmacology , Cell Line, Tumor , Female , Humans , Nedd4 Ubiquitin Protein Ligases/isolation & purification , Nedd4 Ubiquitin Protein Ligases/metabolism , Parkinson Disease/metabolism , Signal Transduction/drug effects
3.
Exp Cell Res ; 390(2): 111958, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32173470

ABSTRACT

The sodium-coupled bicarbonate cotransporter 1 (NBCe1) plays an essential role in the maintenance of acid-base homeostasis in the human body. However, little research has been done regarding the modification of NBCe1. Nedd4-2 is one of the most important ubiquitin E3 ligases in the kidney where it is responsible for mediating the ubiquitylation level of many important ion channel proteins; therefore, influencing their expression and membrane localization. In this study, we performed experiments based on a prediction from bioinformatics analysis that NBCe1 might be a Nedd4-2 target protein. The results of co-immunoprecipitation and glutathione S-transferase pull-down assays showed that Nedd4-2 interacted with NBCe1. An in vitro ubiquitination assay further demonstrated that Nedd4-2 is indeed the NBCe1 ubiquitin E3 ligase. The overexpression of Nedd4-2 decreased NBCe1 expression, while MG132 rescued the changes. Nedd4-2 overexpression also altered the subcellular distribution of NBCe1. Furthermore, the kidney specific Nedd4-2-knockout mice certified the alteration of NBCe1. In addition, we speculate that neddylation activates Nedd4-2. A co-immunoprecipitation analysis indicated that Nedd4-2 interacted with Nedd8. In vitro neddylation experiments further demonstrated that Nedd4-2 underwent neddylation modification. The overexpression of Nedd8 led to decreased NBCe1 expression, while Nedd4-2 inhibition rescued the changes. These findings demonstrate that Nedd4-2 acts as the ubiquitin E3 ligase of NBCe1, mediating the degradation and altering the subcellular distribution of NBCe1, and that the neddylation modification downregulated NBCe1 expression by upregulating Nedd4-2 activity.


Subject(s)
Epithelial Cells/metabolism , NEDD8 Protein/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Protein Processing, Post-Translational , Sodium-Bicarbonate Symporters/metabolism , Animals , Cell Line , Epithelial Cells/cytology , Epithelial Cells/drug effects , Kidney/cytology , Kidney/metabolism , Leupeptins/pharmacology , Mice , Mice, Transgenic , NEDD8 Protein/metabolism , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Nedd4 Ubiquitin Protein Ligases/genetics , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sodium-Bicarbonate Symporters/genetics , Ubiquitination
4.
FEBS J ; 287(3): 529-545, 2020 02.
Article in English | MEDLINE | ID: mdl-31423749

ABSTRACT

The E3 ubiquitin ligase neural precursor cell-expressed developmentally down-regulated protein 4 (NEDD4) plays a crucial role in governing a number of signaling pathways, including insulin and autophagy signaling. However, the molecular mechanism by which NEDD4 gene is transcriptionally regulated has not been fully elucidated. Here, we reported that NEDD4 mRNA and protein levels were increased by peroxisome proliferator-activated receptor-γ (PPARγ) in HepG2 hepatocytes. PPARγ antagonist GW9662 abolished thiazolidinedione (TZD)-induced NEDD4 expression. ChIP and luciferase reporter assays showed that PPARγ directly bound to the potential PPAR-responsive elements (PPREs) within the promoter region of the human NEDD4 gene. In addition, TZDs increased Akt phosphorylation and glucose uptake, which were abrogated through NEDD4 depletion. Furthermore, we showed that NEDD4-mediated autophagy induction and Akt phosphorylation were suppressed by oleic acid and high glucose treatment, activation of PPARγ successfully prevented this suppression. In conclusion, these results suggest that PPARγ plays a novel role in linking glucose metabolism and protein homeostasis through NEDD4-mediated effects on the autophagy machinery.


Subject(s)
Autophagy , Insulin Secretion , Nedd4 Ubiquitin Protein Ligases/genetics , PPAR gamma/metabolism , 3T3 Cells , Anilides/pharmacology , Animals , Glucose/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Mice , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Nedd4 Ubiquitin Protein Ligases/metabolism , PPAR gamma/antagonists & inhibitors , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Response Elements
5.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1477-1489, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30826466

ABSTRACT

BACKGROUND: Maternal immune activation (MIA) is an independent risk factor for psychiatric disorders including depression spectrum in the offsprings, but the molecular mechanism is unclear. Recent studies show that interferon-stimulated gene-15 (ISG15) is involved in inflammation and neuronal dendrite development; here we studied the role of ISG15 in MIA-induced depression and the underlying mechanisms. METHODS: By vena caudalis injecting polyinosinic: polycytidylic acid (poly I:C) into the pregnant rats to mimic MIA, we used AAV or lentivirus to introduce or silence ISG15 expression. Synaptic plasticity was detected by confocal microscope and Golgi staining. Cognitive performances of the offspring were measured by Open field, Forced swimming and Sucrose preference test. RESULTS: We found that MIA induced depression-like behaviors with dendrite impairments in the offspring with ISG15 level increased in the offsprings' brain. Overexpressing ISG15 in the prefrontal cortex of neonatal cubs (P0) could mimic dendritic pathology and depressive like behaviors, while downregulating ISG15 rescued these abnormalities in the offsprings. Further studies demonstrated that MIA-induced upregulation of inflammatory cytokines promoted ISG15 expression in the offspring' brain which suppressed Rap2A ubiquitination via NEDD4 and thus induced Rap2A accumulation, while upregulating NEDD4 abolished ISG15-induced dendrite impairments. CONCLUSIONS: These data reveal that MIA impedes offsprings' dendrite development and causes depressive like behaviors by upregulating ISG15 and suppressing NEDD4/Rap2A signaling. The current findings suggest that inhibiting ISG15 may be a promising intervention of MIA-induced psychiatric disorders in the offsprings.


Subject(s)
Cytokines/genetics , Dendrites/metabolism , GTP-Binding Proteins/genetics , Nedd4 Ubiquitin Protein Ligases/genetics , Prenatal Exposure Delayed Effects/genetics , Ubiquitins/genetics , Animals , Behavior Rating Scale , Cytokines/antagonists & inhibitors , Cytokines/immunology , Dendrites/immunology , Dendrites/pathology , Depression , Disease Models, Animal , Female , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/immunology , Gene Expression Regulation , Immunity, Innate/drug effects , Inflammation , Injections, Intravenous , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Nedd4 Ubiquitin Protein Ligases/immunology , Neurogenesis/drug effects , Neurogenesis/genetics , Neurogenesis/immunology , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neuronal Plasticity/immunology , Poly I-C/administration & dosage , Prefrontal Cortex/immunology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Ubiquitins/antagonists & inhibitors , Ubiquitins/immunology
6.
Mol Cell Biol ; 38(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29760280

ABSTRACT

CD133, a widely known marker of cancer stem cells, was recently found in extracellular vesicles. However, the mechanisms underlying CD133 translocation to the extracellular space remain largely unknown. Here we report that CD133 is monoubiquitinated. Ubiquitination occurs primarily on complex glycosylated CD133. The lysine 848 residue at the intracellular carboxyl terminus is one of the sites for CD133 ubiquitination. The K848R mutation does not affect CD133 degradation by the lysosomal pathway but significantly reduces CD133 secretion by inhibiting the interaction between CD133 and tumor susceptibility gene 101 (Tsg101). Furthermore, knockdown of the E3 ubiquitin protein ligase Nedd4 largely impairs CD133 ubiquitination and vesicle secretion. Importantly, CD133-containing vesicles are taken up by recipient cells, consequently promoting cell migration. The K848R mutation reduces cell migration induced by CD133. Taken together, our findings show that monoubiquitination contributes to CD133 vesicle secretion and promotes recipient cell migration. These findings provide a clue to the mechanisms of CD133 secretion and cancer stem cell microenvironment interactional effects.


Subject(s)
AC133 Antigen/metabolism , Neoplastic Stem Cells/metabolism , AC133 Antigen/chemistry , AC133 Antigen/genetics , Amino Acid Substitution , Cell Line , Cell Movement/genetics , Cell Movement/physiology , DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Extracellular Vesicles/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Lysine/metabolism , Lysosomes/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Neoplastic Stem Cells/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/metabolism , Ubiquitination
7.
Cell Cycle ; 17(6): 728-738, 2018.
Article in English | MEDLINE | ID: mdl-29480061

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related death worldwide. In China, the situation is even worse as cancer incidence and mortality continue to increase rapidly. Although tremendous progress has been made toward HCC treatments, the benefits for liver cancer patients are still limited. Therefore, it is necessary to identify and develop novel therapeutic methods. Neuronally expressed developmentally downregulated 4 (NEDD4), an E3 ubiquitin ligase, plays a critical role in the development and progression of various types of human cancers. In our study, NEDD4 acts as an oncoprotein in both QGY7703 and SMMC7721 liver cancer cell lines. We found that depletion of NEDD4 by siRNA transfection led to inhibition of cell growth, invasion and migration, and promotion of apoptosis. In contrast, overexpression of NEDD4 via plasmid transfection resulted in facilitated cell proliferation, invasion and migration, and decreased apoptosis. Importantly, we observed that tumor suppressor LATS1, also a core component of Hippo pathway, was negatively regulated by NEDD4 in liver cancer cells. Our findings suggested that NEDD4 may be involved in the HCC progression via regulating LATS1 associated signaling pathway. Therefore, targeting NEDD4-LATS1 signaling could be a potential therapeutic option for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Liver Neoplasms/physiopathology , Nedd4 Ubiquitin Protein Ligases/metabolism , Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Nedd4 Ubiquitin Protein Ligases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism
8.
Cell Cycle ; 16(16): 1509-1514, 2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28745938

ABSTRACT

The neural precursor cell expressed developmentally downregulated protein 4 (NEDD4) plays a pivotal oncogenic role in various types of human cancers. However, the function of NEDD4 in bladder cancer has not been fully investigated. In the present study, we aim to explore whether NEDD4 governs cell proliferation, apoptosis, migration, and invasion in bladder cancer cells. Our results showed that downregulation of NEDD4 suppressed cell proliferation in bladder cancer cells. Moreover, we found that inhibition of NEDD4 significantly induced cell apoptosis. Furthermore, downregulation of NEDD4 retarded cell migration and invasion. Notably, overexpression of NEDD4 enhanced cell growth and inhibited apoptosis. Consistently, upregulation of NEDD4 promoted cell migration and invasion in bladder cancer cells. Mechanically, our Western blotting results revealed that downregulation of NEDD4 activated PTEN and inhibited Notch-1 expression, whereas upregulation of NEDD4 reduced PTEN level and increased Notch-1 level in bladder cancer cells. Our findings indicated that NEDD4 exerts its oncogenic function partly due to regulation of PTEN and Notch-1 in bladder cancer cells. These results further revealed that targeting NEDD4 could be a useful approach for the treatment of bladder cancer.


Subject(s)
Apoptosis , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Urinary Bladder Neoplasms/pathology , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Humans , Nedd4 Ubiquitin Protein Ligases/metabolism , Neoplasm Invasiveness , PTEN Phosphohydrolase/metabolism , RNA, Small Interfering/metabolism , Receptors, Notch/metabolism , Urinary Bladder Neoplasms/genetics
9.
Sci Rep ; 7: 45375, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28349961

ABSTRACT

Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus that causes the most prevalent viral encephalitis in Asia. Since JEV is a neurotropic virus, it is important to identify key molecules that mediate JEV infection in neuronal cells and to investigate their underlying mechanisms. In this study, the critical role of Nedd4, an E3 ubiquitin ligase that is highly expressed in the central nervous system, was examined in JEV propagation. In SK-N-SH neuroblastoma cells, Nedd4 was up-regulated in response to JEV infection. Moreover, down-regulation of Nedd4 resulted in a significant decrease in JEV replication without alterations in virus attachment and internalization or in JEV pseudotyped virus infection, suggesting that Nedd4 participates in the replication but not in the entry stage of JEV infection. Further functional analysis showed that Nedd4 attenuated JEV-induced autophagy, which negatively regulates virus replication during infection. These results suggest that Nedd4 facilitates the replication of JEV by suppressing virus-induced autophagy. Taken together, our results indicate that Nedd4 plays a crucial role in JEV infection of neuronal cells, which provides a potential target for the development of novel treatment to combat JEV infection.


Subject(s)
Autophagy , Encephalitis Virus, Japanese/physiology , Nedd4 Ubiquitin Protein Ligases/metabolism , Beclin-1/antagonists & inhibitors , Beclin-1/genetics , Beclin-1/metabolism , Cell Line, Tumor , Central Nervous System/metabolism , Humans , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Nedd4 Ubiquitin Protein Ligases/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation , Virus Attachment , Virus Internalization , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...