Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 107
Filtrer
1.
Article de Anglais | MEDLINE | ID: mdl-39073416

RÉSUMÉ

Glioblastoma (GBM) is the most aggressive form of brain cancer, characterized by rapid growth and invasion into surrounding brain tissue. Ubiquitin-specific protease 9X (USP9X) has emerged as a key regulator in various cancers, but its role in GBM pathogenesis remains unclear. Understanding the molecular mechanisms underlying USP9X modulation of GBM progression could unveil potential therapeutic targets for this deadly disease. The mRNA and protein levels were determined in GBM tissues and/or cells using quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting assays, respectively. Cell migration was evaluated through wound-healing assay, while cell proliferation was measured using colony formation and CCK-8 assays. Flow cytometry analysis was performed to quantify the CD206-positive macrophages to assess M2 polarization. Co-immunoprecipitation (Co-IP) assays were conducted to elucidate the association between USP9X and transformation/transcription domain-associated protein (TRRAP). Cycloheximide (CHX) treatment was used to determine the impact of USP9X on TRRAP protein stabilization. Furthermore, the effect of USP9X depletion on GBM cell malignancy was validated using a xenograft mouse model. We found that USP9X expression was elevated in GBM tissues and cells. Depletion of USP9X suppressed GBM cell migration, proliferation, and M2 macrophage polarization. Mechanistically, USP9X stabilized TRRAP through the deubiquitination pathway in GBM cells, and TRRAP mitigated the effects of USP9X silencing on GBM cell malignant phenotypes and M2 macrophage polarization. Moreover, silencing of USP9X inhibited tumor formation in vivo. Together, USP9X deubiquitinated TRRAP, thereby promoting glioblastoma cell proliferation, migration, and M2 macrophage polarization. These results highlight the potential of targeting the USP9X-TRRAP axis as a therapeutic strategy for GBM.

2.
BMC Gastroenterol ; 24(1): 239, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075342

RÉSUMÉ

BACKGROUND: MutT homolog 1 (MTH1) sanitizes oxidized dNTP pools to promote the survival of cancer cells and its expression is frequently upregulated in cancers. Polyubiquitination stabilizes MTH1 to facilitate the proliferation of melanoma cells, suggesting the ubiquitin system controls the stability and function of MTH1. However, whether ubiquitination regulates MTH1 in gastric cancers has not been well defined. This study aims to investigate the interaction between MTH1 and a deubiquitinase, USP9X, in regulating the proliferation, survival, migration, and invasion of gastric cancer cells. METHODS: The interaction between USP9X and MTH1 was evaluated by co-immunoprecipitation (co-IP) in HGC-27 gastric cancer cells. siRNAs were used to interfere with USP9X expression in gastric cancer cell lines HGC-27 and MKN-45. MTT assays were carried out to examine the proliferation, propidium iodide (PI) and 7-AAD staining assays were performed to assess the cell cycle, Annexin V/PI staining assays were conducted to examine the apoptosis, and transwell assays were used to determine the migration and invasion of control, USP9X-deficient, and USP9X-deficient plus MTH1-overexpressing HGC-27 and MKN-45 gastric cancer cells. RESULTS: Co-IP data show that USP9X interacts with and deubiquitinates MTH1. Overexpression of USP9X elevates MTH1 protein level by downregulating its ubiquitination, while knockdown of USP9X has the opposite effect on MTH1. USP9X deficiency in HGC-27 and MKN-45 cells causes decreased proliferation, cell cycle arrest, extra apoptosis, and defective migration and invasion, which could be rescued by excessive MTH1. CONCLUSION: USP9X interacts with and stabilizes MTH1 to promote the proliferation, survival, migration and invasion of gastric cancer cells.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Enzymes de réparation de l'ADN , Invasion tumorale , Phosphoric monoester hydrolases , Tumeurs de l'estomac , Ubiquitin thiolesterase , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Prolifération cellulaire/génétique , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Lignée cellulaire tumorale , Phosphoric monoester hydrolases/métabolisme , Phosphoric monoester hydrolases/génétique , Enzymes de réparation de l'ADN/métabolisme , Enzymes de réparation de l'ADN/génétique , Ubiquitination , Survie cellulaire , Apoptose , Petit ARN interférent
3.
Ren Fail ; 46(2): 2361089, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38874156

RÉSUMÉ

As a pattern recognition receptor, Toll-like receptor 4 (TLR4) is crucial for the development and progression of acute kidney injury (AKI). This study aims to explore whether the deubiquitinase Usp9x influences the TLR4/NF-B pathway to cause sepsis-induced acute kidney injury (S-AKI). The model of AKI was established in Sprague-Dawley rats using the cecal ligation and puncture (CLP) method, while renal tubular epithelial cell NRK-52E was stimulated with lipopolysaccharide (LPS) in vitro. All plasmids were transfected into NRK-52E cells according to the indicated group. The deubiquitinase of TLR4 was predicted by the online prediction software Ubibrowser. Subsequently, Western blot and Pearson correlation analysis identified Usp9x protein as a potential candidate. Co-IP analysis verified the interaction between TLR4 and Usp9x. Further research revealed that overexpression of Usp9x inhibited degradation of TLR4 protein by downregulating its ubiquitination modification levels. Both in vivo and in vitro experiments observed that interference with Usp9x effectively alleviated the inflammatory response and apoptosis of renal tubular epithelial cells (RTECs) induced by CLP or LPS, whereas overexpression of TLR4 reversed this situation. Transfection with sh-Usp9x in NRK-52E cells suppressed the expression of proteins associated with the TLR4/NF-κB pathway induced by LPS. Moreover, the overexpression of TLR4 reversed the effect of sh-Usp9x transfection. Therefore, the deubiquitinase Usp9x interacts with TLR4, leading to the upregulation of its expression through deubiquitination modification, and the activation of the TLR4/NF-κB signaling pathway, thereby promoting inflammation and apoptosis in renal tubular epithelial cells and contributing to sepsis-induced acute kidney injury.


Sujet(s)
Atteinte rénale aigüe , Apoptose , Cellules épithéliales , Inflammation , Tubules rénaux , Facteur de transcription NF-kappa B , Rat Sprague-Dawley , Sepsie , Transduction du signal , Récepteur de type Toll-4 , Ubiquitin thiolesterase , Animaux , Récepteur de type Toll-4/métabolisme , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/étiologie , Atteinte rénale aigüe/anatomopathologie , Sepsie/complications , Sepsie/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Rats , Cellules épithéliales/métabolisme , Tubules rénaux/anatomopathologie , Tubules rénaux/métabolisme , Tubules rénaux/cytologie , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Mâle , Inflammation/métabolisme , Modèles animaux de maladie humaine , Lignée cellulaire , Lipopolysaccharides , Ubiquitination
4.
J Biomed Sci ; 31(1): 55, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38802791

RÉSUMÉ

BACKGROUND: Radioresistance is a key clinical constraint on the efficacy of radiotherapy in lung cancer patients. REV1 DNA directed polymerase (REV1) plays an important role in repairing DNA damage and maintaining genomic stability. However, its role in the resistance to radiotherapy in lung cancer is not clear. This study aims to clarify the role of REV1 in lung cancer radioresistance, identify the intrinsic mechanisms involved, and provide a theoretical basis for the clinical translation of this new target for lung cancer treatment. METHODS: The effect of targeting REV1 on the radiosensitivity was verified by in vivo and in vitro experiments. RNA sequencing (RNA-seq) combined with nontargeted metabolomics analysis was used to explore the downstream targets of REV1. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to quantify the content of specific amino acids. The coimmunoprecipitation (co-IP) and GST pull-down assays were used to validate the interaction between proteins. A ubiquitination library screening system was constructed to investigate the regulatory proteins upstream of REV1. RESULTS: Targeting REV1 could enhance the radiosensitivity in vivo, while this effect was not obvious in vitro. RNA sequencing combined with nontargeted metabolomics revealed that the difference result was related to metabolism, and that the expression of glycine, serine, and threonine (Gly/Ser/Thr) metabolism signaling pathways was downregulated following REV1 knockdown. LC-MS/MS demonstrated that REV1 knockdown results in reduced levels of these three amino acids and that cystathionine γ-lyase (CTH) was the key to its function. REV1 enhances the interaction of CTH with the E3 ubiquitin ligase Rad18 and promotes ubiquitination degradation of CTH by Rad18. Screening of the ubiquitination compound library revealed that the ubiquitin-specific peptidase 9 X-linked (USP9X) is the upstream regulatory protein of REV1 by the ubiquitin-proteasome system, which remodels the intracellular Gly/Ser/Thr metabolism. CONCLUSION: USP9X mediates the deubiquitination of REV1, and aberrantly expressed REV1 acts as a scaffolding protein to assist Rad18 in interacting with CTH, promoting the ubiquitination and degradation of CTH and inducing remodeling of the Gly/Ser/Thr metabolism, which leads to radioresistance. A novel inhibitor of REV1, JH-RE-06, was shown to enhance lung cancer cell radiosensitivity, with good prospects for clinical translation.


Sujet(s)
Tumeurs du poumon , Nucleotidyltransferases , Radiotolérance , Ubiquitin-protein ligases , Ubiquitination , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/radiothérapie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Lignée cellulaire tumorale , Souris , Animaux , DNA-directed DNA polymerase
5.
Biochim Biophys Acta Rev Cancer ; 1879(3): 189099, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38582329

RÉSUMÉ

Protein ubiquitination, one of the most significant post-translational modifications, plays an important role in controlling the proteins activity in diverse cellular processes. The reversible process of protein ubiquitination, known as deubiquitination, has emerged as a critical mechanism for maintaining cellular homeostasis. The deubiquitinases (DUBs), which participate in deubiquitination process are increasingly recognized as potential candidates for drug discovery. Among these DUBs, ubiquitin-specific protease 9× (USP9X), a highly conserved member of the USP family, exhibits versatile functions in various cellular processes, including the regulation of cell cycle, protein endocytosis, apoptosis, cell polarity, immunological microenvironment, and stem cell characteristics. The dysregulation and abnormal activities of USP9X are influenced by intricate cellular signaling pathway crosstalk and upstream non-coding RNAs. The complex expression patterns and controversial clinical significance of USP9X in cancers suggest its potential as a prognostic biomarker. Furthermore, USP9X inhibitors has shown promising antitumor activity and holds the potential to overcome therapeutic resistance in preclinical models. However, a comprehensive summary of the role and molecular functions of USP9X in cancer progression is currently lacking. In this review, we provide a comprehensive delineation of USP9X participation in numerous critical cellular processes, complicated signaling pathways within the tumor microenvironment, and its potential translational applications to combat therapeutic resistance. By systematically summarizing the updated molecular mechanisms of USP9X in cancer biology, this review aims to contribute to the advancement of cancer therapeutics and provide essential insights for specialists and clinicians in the development of improved cancer treatment strategies.


Sujet(s)
Tumeurs , Transduction du signal , Ubiquitin thiolesterase , Ubiquitination , Humains , Ubiquitin thiolesterase/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Tumeurs/génétique , Animaux , Thérapie moléculaire ciblée , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Microenvironnement tumoral
6.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-1039530

RÉSUMÉ

【Objective】 To explore the expression of USP9X in platelets and its effect on platelet function. 【Methods】 The expression of USP9X in human and mouse was evaluated by PCR and Western blot. Platelets from young and old mice were separated and prepared, and the expression of USP9X was detected. USP9X inhibitos were used to assess the regulation of USP9X in platelet function, including aggregation, ATP release and spreading. Platelet lysates were collected in different time points to evaluate the change of phosphorylation of Akt in USP9X inhibitors treated platelets. 【Results】 Both human and mouse platelets expressed USP9X. Compared to the young mice, the old mice showed significantly enhanced expression of USP9X(P<0.05). To assess the effect of USP9X on platelet function, USP9X inhibitor was used to pre-incubate platelets for 30 min and platelet function were examined later. Results showed that USP9X inhibitor significantly decreased platelet activation including aggregation, ATP release and spreading(P<0.05). Compared to the control group, the inhibitor treated group showed a significant decrease in the spreading area after 45 minutes. The Western blot results showed a significant decrease in Akt phosphorylation levels of platelets in the USP9X inhibitor treated group. 【Conclusion】 Both human and mouse platelet express USP9X, and inhibition of USP9X decreased platelet function including aggregation, ATP release and spreading. USP9X can also influence the phosphorylation of Akt. The inhibitor of USP9X may become a potential therapeutic target for thrombosis intervention.

7.
Genes (Basel) ; 14(10)2023 10 17.
Article de Anglais | MEDLINE | ID: mdl-37895297

RÉSUMÉ

Axenfeld-Rieger anomaly (ARA) is a specific ocular disorder that is frequently associated with other systemic abnormalities. PITX2 and FOXC1 variants explain the majority of individuals with Axenfeld-Rieger syndrome (ARS) but leave ~30% unsolved. Here, we present pathogenic/likely pathogenic variants in nine families with ARA/ARS or similar phenotypes affecting five different genes/regions. USP9X and JAG1 explained three families each. USP9X was recently linked with syndromic cognitive impairment that includes hearing loss, dental defects, ventriculomegaly, Dandy-Walker malformation, skeletal anomalies (hip dysplasia), and other features showing a significant overlap with FOXC1-ARS. Anterior segment anomalies are not currently associated with USP9X, yet our cases demonstrate ARA, congenital glaucoma, corneal neovascularization, and cataracts. The identification of JAG1 variants, linked with Alagille syndrome, in three separate families with a clinical diagnosis of ARA/ARS highlights the overlapping features and high variability of these two phenotypes. Finally, intragenic variants in CDK13, BCOR, and an X chromosome deletion encompassing HCCS and AMELX (linked with ocular and dental anomalies, correspondingly) were identified in three additional cases with ARS. Accurate diagnosis has important implications for clinical management. We suggest that broad testing such as exome sequencing be applied as a second-tier test for individuals with ARS with normal results for PITX2/FOXC1 sequencing and copy number analysis, with attention to the described genes/regions.


Sujet(s)
Malformations oculaires , Facteurs de transcription , Humains , Facteurs de transcription/génétique , Protéines à homéodomaine/génétique , Pôle antérieur du bulbe oculaire/malformations , Malformations oculaires/diagnostic , Malformations oculaires/génétique , Malformations oculaires/anatomopathologie , Ubiquitin thiolesterase
8.
J Biol Chem ; 299(8): 105055, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37454738

RÉSUMÉ

Post-translational modifications including protein ubiquitination regulate a plethora of cellular processes in distinct manners. RNA N6-methyladenosine is the most abundant post-transcriptional modification on mammalian mRNAs and plays important roles in various physiological and pathological conditions including hematologic malignancies. We previously determined that the RNA N6-methyladenosine eraser ALKBH5 is necessary for the maintenance of acute myeloid leukemia (AML) stem cell function, but the post-translational modifications involved in ALKBH5 regulation remain elusive. Here, we show that deubiquitinase ubiquitin-specific peptidase 9X (USP9X) stabilizes ALKBH5 and promotes AML cell survival. Through the use of mass spectrometry as an unbiased approach, we identify USP9X and confirm that it directly binds to ALKBH5. USP9X stabilizes ALKBH5 by removing the K48-linked polyubiquitin chain at K57. Using human myeloid leukemia cells and a murine AML model, we find that genetic knockdown or pharmaceutical inhibition of USP9X inhibits leukemia cell proliferation, induces apoptosis, and delays AML development. Ectopic expression of ALKBH5 partially mediates the function of USP9X in AML. Overall, this study uncovers deubiquitinase USP9X as a key for stabilizing ALKBH5 expression and reveals the important role of USP9X in AML, which provides a promising therapeutic strategy for AML treatment in the clinic.


Sujet(s)
AlkB Homolog 5, RNA demethylase , Leucémie aigüe myéloïde , Ubiquitin thiolesterase , Animaux , Humains , Souris , AlkB Homolog 5, RNA demethylase/génétique , Lignée cellulaire tumorale , Survie cellulaire , Leucémie aigüe myéloïde/génétique , ARN , Ubiquitin thiolesterase/génétique , Ubiquitination
9.
Mol Carcinog ; 62(10): 1487-1503, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37314216

RÉSUMÉ

Cell division cycle 123 (CDC123) has been implicated in a variety of human diseases. However, it remains unclear whether CDC123 plays a role in tumorigenesis and how its abundance is regulated. In this study, we found that CDC123 was highly expressed in breast cancer cells, and its high expression was positively correlated with a poor prognosis. Knowndown of CDC123 impaired the proliferation of breast cancer cells. Mechanistically, we identified a deubiquitinase, ubiquitin-specific peptidase 9, X-linked (USP9X), that could physically interact with and deubiquitinate K48-linked ubiquitinated CDC123 at the K308 site. Therefore, the expression of CDC123 was positively correlated with USP9X in breast cancer cells. In addition, we found that deletion of either USP9X or CDC123 led to altered expression of cell cycle-related genes and resulted in the accumulation of cells population in the G0/G1 phase, thereby suppressing cell proliferation. Treatment with the deubiquitinase inhibitor of USP9X, WP1130 (Degrasyn, a small molecule compound that USP9X deubiquitinase inhibitor), also led to the accumulation of breast cancer cells in the G0/G1 phase, but this effect could be rescued by overexpression of CDC123. Furthermore, our study revealed that the USP9X/CDC123 axis promotes the occurrence and development of breast cancer through regulating the cell cycle, and suggests that it may be a potential target for breast cancer intervention. In conclusion, our study demonstrates that USP9X is a key regulator of CDC123, providing a novel pathway for the maintenance of CDC123 abundance in cells, and supports USP9X/CDC123 as a potential target for breast cancer intervention through regulating the cell cycle.


Sujet(s)
Tumeurs du sein , Transformation cellulaire néoplasique , Femelle , Humains , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Cycle cellulaire , Lignée cellulaire tumorale , Enzymes de désubiquitinylation , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme
10.
Int J Cancer ; 153(6): 1300-1312, 2023 09 15.
Article de Anglais | MEDLINE | ID: mdl-37260183

RÉSUMÉ

Mammalian target of rapamycin (mTOR) is a central regulator of mammalian metabolism and physiology. Aberrant hyperactivation of the mTOR pathway promotes tumor growth and metastasis, and can also promote tumor resistance to chemotherapy and cancer drugs; this makes mTOR an attractive cancer therapeutic target. mTOR inhibitors have been approved to treat cancer; however, the mechanisms underlying drug sensitivity remain poorly understood. Here, whole exome sequencing of three chromophobe renal cell carcinoma (chRCC) patients with exceptional mTOR inhibitor sensitivity revealed that all three patients shared somatic mutations in the deubiquitinase gene USP9X. The clonal characteristics of the mutations, which were amassed by studying multiple patients' primary and metastatic samples from various years, together with the low USP9X mutation rate in unselected chRCC series, reinforced a causal link between USP9X and mTOR inhibitor sensitivity. Rapamycin treatment of USP9X-depleted HeLa and renal cancer 786-O cells, along with the pharmacological inhibition of USP9X, confirmed that this protein plays a role in patients' sensitivity to mTOR inhibitors. USP9X was not found to exert a direct effect on mTORC1, but subsequent ubiquitylome analyses identified p62 as a direct USP9X target. Increased p62 ubiquitination and the augmented rapamycin effect upon bortezomib treatment, together with the results of p62 and LC3 immunofluorescence assays, suggested that dysregulated autophagy in USP9X-depleted cells can have a synergistic effect with mTOR inhibitors. In summary, we show that USP9X constitutes a potential novel marker of sensitivity to mTOR inhibitors in chRCC patients, and represents a clinical strategy for increasing the sensitivity to these drugs.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Humains , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Enzymes de désubiquitinylation , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/génétique , Inhibiteurs de mTOR , Sirolimus/pharmacologie , Sérine-thréonine kinases TOR/génétique , Sérine-thréonine kinases TOR/métabolisme , Ubiquitin thiolesterase/génétique
11.
Cancers (Basel) ; 15(9)2023 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-37173959

RÉSUMÉ

BACKGROUND: Radiotherapy constitutes an important therapeutic option for prostate cancer. However, prostate cancer cells often acquire resistance during cancer progression, limiting the cytotoxic effects of radiotherapy. Among factors regulating sensitivity to radiotherapy are members of the Bcl-2 protein family, known to regulate apoptosis at the mitochondrial level. Here, we analyzed the role of anti-apoptotic Mcl-1 and USP9x, a deubiquitinase stabilizing Mcl-1 protein levels, in prostate cancer progression and response to radiotherapy. METHODS: Changes in Mcl-1 and USP9x levels during prostate cancer progression were determined by immunohistochemistry. Neutralization of Mcl-1 and USP9x was achieved by siRNA-mediated knockdown. We analyzed Mcl-1 stability after translational inhibition by cycloheximide. Cell death was determined by flow cytometry using an exclusion assay of mitochondrial membrane potential-sensitive dye. Changes in the clonogenic potential were examined by colony formation assay. RESULTS: Protein levels of Mcl-1 and USP9x increased during prostate cancer progression, and high protein levels correlated with advanced prostate cancer stages. The stability of Mcl-1 reflected Mcl-1 protein levels in LNCaP and PC3 prostate cancer cells. Moreover, radiotherapy itself affected Mcl-1 protein turnover in prostate cancer cells. Particularly in LNCaP cells, the knockdown of USP9x expression reduced Mcl-1 protein levels and increased sensitivity to radiotherapy. CONCLUSION: Posttranslational regulation of protein stability was often responsible for high protein levels of Mcl-1. Moreover, we demonstrated that deubiquitinase USP9x as a factor regulating Mcl-1 levels in prostate cancer cells, thus limiting cytotoxic response to radiotherapy.

12.
Mol Syndromol ; 14(2): 158-163, 2023 Apr.
Article de Anglais | MEDLINE | ID: mdl-37064340

RÉSUMÉ

Introduction: The X-chromosomal USP9X gene encodes a deubiquitylating enzyme involved in protein turnover and TGF-ß signaling during fetal and neuronal development. USP9X variants in females are primarily associated with complete loss-of-function (LOF) alleles, leading to neurodevelopmental delay and intellectual disability, as well as a wide range of congenital anomalies. In contrast, USP9X missense variants in males often result in partial rather than complete LOF, specifically affecting neuronal migration and development. USP9X variants in males are associated with intellectual disability, behavioral disorders, global developmental delay, speech delay, and structural CNS defects. Facial dysmorphisms are found in almost all patients. Case Presentation: We report the case of an Italian boy presenting dysmorphism, intellectual disability, structural brain anomalies, and congenital heart disease. Using next-generation sequencing analysis, we identified a hemizygous de novo variant in the USP9X gene (c.5470A>G, p.Met1824Val) that was never reported in the literature. Conclusion: We provide an overview of the available literature on USP9X variants in males, in order to further expand the genotypic and phenotypic landscape of male-restricted X-linked mental retardation syndrome. Our findings confirm the involvement of USP9X variants in neuronal development and corroborate the possible association between the novel USP9X variant and congenital heart malformation.

13.
J Cancer ; 14(4): 591-599, 2023.
Article de Anglais | MEDLINE | ID: mdl-37057289

RÉSUMÉ

An increasing number of studies have shown that USP9X is closely related to cancer. However, its role in carcinogenesis and progression of laryngeal cancer has not yet been investigated. In this study, we found that USP9X was upregulated in laryngeal cancer tissues. The expression of USP9X was significantly correlated with degree of laryngeal cancer differentiation and lymphatic metastasis. USP9X knockdown led to a decrease in the ability of proliferation, migration, and invasion of FaDu cells. The proportion of FaDu apoptotic cells increased by interfering with the endogenous expression of USP9X. We speculated that inhibiting USP9X might induce apoptosis in FaDu cells by downregulating Mcl-1 and upregulating Bax protein expression. Our findings for the first time suggest the expression level and trend of USP9X in laryngeal cancer tissue and USP9X may plays an important role in promoting the occurrence and progression of laryngeal cancer. USP9X may be a potential target for intervention in treatment of laryngeal cancer.

14.
Cell Rep Med ; 4(4): 101007, 2023 04 18.
Article de Anglais | MEDLINE | ID: mdl-37030295

RÉSUMÉ

Pancreatic ductal adenocarcinomas (PDACs) frequently harbor KRAS mutations. Although MEK inhibitors represent a plausible therapeutic option, most PDACs are innately resistant to these agents. Here, we identify a critical adaptive response that mediates resistance. Specifically, we show that MEK inhibitors upregulate the anti-apoptotic protein Mcl-1 by triggering an association with its deubiquitinase, USP9X, resulting in acute Mcl-1 stabilization and protection from apoptosis. Notably, these findings contrast the canonical positive regulation of Mcl-1 by RAS/ERK. We further show that Mcl-1 inhibitors and cyclin-dependent kinase (CDK) inhibitors, which suppress Mcl-1 transcription, prevent this protective response and induce tumor regression when combined with MEK inhibitors. Finally, we identify USP9X as an additional potential therapeutic target. Together, these studies (1) demonstrate that USP9X regulates a critical mechanism of resistance in PDAC, (2) reveal an unexpected mechanism of Mcl-1 regulation in response to RAS pathway suppression, and (3) provide multiple distinct promising therapeutic strategies for this deadly malignancy.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Humains , Protéine Mcl-1/génétique , Protéine Mcl-1/métabolisme , Lignée cellulaire tumorale , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/génétique , Mitogen-Activated Protein Kinase Kinases , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme
15.
Clin Exp Hypertens ; 45(1): 2186319, 2023 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-36890708

RÉSUMÉ

Endothelial pyroptosis is a pathological mechanism of atherosclerosis (AS). Circular RNAs (circRNAs) are vital in AS progression by regulating endothelial cell functions. The study aimed to explore whether circ-USP9× regulated pyroptosis of endothelial cell to involve in AS development and the molecular mechanism. Pyroptosis was determined using lactate dehydrogenase (LDH) assay, enzyme linked immunosorbent assay (ELISA), flow cytometry, propidium iodide (PI) staining assay, and western blot. The mechanism of circ-USP9× was determined using RNA pull-down and RNA binding protein immunoprecipitation (RIP) assays. Results showed that circ-USP9× was upregulated in AS and oxidized low-density lipoprotein (ox-LDL)-treated human umbilical vein endothelial cells (HUVECs). Knockdown of circ-USP9× suppressed ox-LDL induced pyroptosis of HUVECs. Mechanically, circ-USP9× could bind to EIF4A3 in the cytoplasm. Moreover, EIF4A3 was bound to GSDMD and further affects GSDMD stability. Overexpression of EIF4A3 rescued cell pyroptosis induced by circ-USP9× depletion. In short, circ-USP9× interacted with EIF4A3 to enhance GSDMD stability, thus further promoting ox-LDL-induced pyroptosis of HUVECs. These findings suggested that circ-USP9× participates in AS progression and may be a potential therapeutic target for AS.


Sujet(s)
Athérosclérose , microARN , Humains , Apoptose , Athérosclérose/génétique , Prolifération cellulaire , DEAD-box RNA helicases , Test ELISA , Facteur-4A d'initiation eucaryote , Cellules endothéliales de la veine ombilicale humaine , L-Lactate dehydrogenase , Lipoprotéines LDL/pharmacologie , Protéines de liaison aux phosphates/génétique , Perforines , Pyroptose
16.
J Virol ; 97(3): e0176322, 2023 03 30.
Article de Anglais | MEDLINE | ID: mdl-36995092

RÉSUMÉ

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi sarcoma (KS), the plasmablastic form of multicentric Castleman's disease, and primary effusion lymphoma. In sub-Saharan Africa, KS is the most common HIV-related malignancy and one of the most common childhood cancers. Immunosuppressed patients, including HIV-infected patients, are more prone to KSHV-associated disease. KSHV encodes a viral protein kinase (vPK) that is expressed from ORF36. KSHV vPK contributes to the optimal production of infectious viral progeny and upregulation of protein synthesis. To elucidate the interactions of vPK with cellular proteins in KSHV-infected cells, we used a bottom-up proteomics approach and identified host protein ubiquitin-specific peptidase 9X-linked (USP9X) as a potential interactor of vPK. Subsequently, we validated this interaction using a co-immunoprecipitation assay. We report that both the ubiquitin-like and the catalytic domains of USP9X are important for association with vPK. To uncover the biological relevance of the USP9X/vPK interaction, we investigated whether the knockdown of USP9X would modulate viral reactivation. Our data suggest that depletion of USP9X inhibits both viral reactivation and the production of infectious virions. Understanding how USP9X influences the reactivation of KSHV will provide insights into how cellular deubiquitinases regulate viral kinase activity and how viruses co-opt cellular deubiquitinases to propagate infection. Hence, characterizing the roles of USP9X and vPK during KSHV infection constitutes a first step toward identifying a potentially critical interaction that could be targeted by future therapeutics. IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi sarcoma (KS), the plasmablastic form of multicentric Castleman's disease, and primary effusion lymphoma. In sub-Saharan Africa, KS is the most common HIV-related malignancy. KSHV encodes a viral protein kinase (vPK) that aids viral replication. To elucidate the interactions of vPK with cellular proteins in KSHV-infected cells, we used an affinity purification approach and identified host protein ubiquitin-specific peptidase 9X-linked (USP9X) as a potential interactor of vPK. Depletion of USP9X inhibits both viral reactivation and the production of infectious virions. Overall, our data suggest a proviral role for USP9X.


Sujet(s)
Herpèsvirus humain de type 8 , Sarcome de Kaposi , Ubiquitin thiolesterase , Enfant , Humains , Enzymes de désubiquitinylation , Herpèsvirus humain de type 8/physiologie , Infections à VIH/complications , Lymphome primitif des séreuses , Protein kinases/génétique , Protein kinases/métabolisme , Sarcome de Kaposi/métabolisme , Sarcome de Kaposi/anatomopathologie , Sarcome de Kaposi/virologie , Ubiquitin thiolesterase/génétique , Protéines virales/génétique
17.
Am J Med Genet A ; 191(5): 1350-1354, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36680497

RÉSUMÉ

The ubiquitin-specific protease USP9X has been found to play a role in multiple aspects of neural development including processes of neuronal migrations. In males, hemizygous partial loss of function variants in USP9X lead to a clinical phenotype primarily characterized by intellectual disability, hypotonia, speech and language impairment, behavioral disturbances accompanied by additional clinical features with variable expressivity. Structural brain abnormalities are reported in all cases where neuro-imaging was performed. The most common radiological features described include hypoplasia/agenesis of the corpus callosum, widened ventricles, white matter disturbances, and cerebellar hypoplasia. Here we report a child harboring a missense variant in USP9X presenting with the classical neurodevelopmental phenotype and a previously unreported radiological picture of periventricular heterotopia. This case expands the phenotypic landscape of this emergent condition and supports the critical role of USP9X in neuronal migration processes.


Sujet(s)
Déficience intellectuelle , Hétérotopie nodulaire périventriculaire , Humains , Enfant , Mâle , Hétérotopie nodulaire périventriculaire/imagerie diagnostique , Hétérotopie nodulaire périventriculaire/génétique , Déficience intellectuelle/diagnostic , Déficience intellectuelle/génétique , Déficience intellectuelle/complications , Mutation faux-sens , Incapacités de développement/génétique , Radiographie , Ubiquitin thiolesterase/génétique
18.
Cell Biol Int ; 47(2): 394-405, 2023 Feb.
Article de Anglais | MEDLINE | ID: mdl-36525374

RÉSUMÉ

Alveolar epithelial cells (AECs) function as a vital defense barrier avoiding the invasion of exogenous agents and preserving the functional and structural integrity of lung tissues, while damage/breakdown of this airway epithelial barrier is frequently associated with the pathogenesis of acute lung injury (ALI). NOD-like receptor family, pyrindomain-containing 3 (NLRP3) inflammasome activation-associated pyroptosis is involved in the development of ALI. Yet, how the activity of NLRP3 inflammasome is regulated in the context of ALI remains unknown. Herein we hypothesized that USP9X, an important deubiquitinase, participates in modulating the activation of NLRP3 inflammasome, thereby affecting the phenotypes in a lipopolysaccharide (LPS)-stimulated AEC model. Human pulmonary AECs were subjected to LPS/adenosine triphosphate (ATP) treatment to induce NLRP3 inflammasome activation and cell pyroptosis. Knockdown and overexpression of USP9X were applied to validate the function of USP9X. Inhibitors of proteinase and protein synthesis, as well as approach of co-immunoprecipitation coupled with Western blot, were utilized to explore the molecular mechanism. LPS/ATP challenge resulted in pronouncedly increased pyroptosis of AECs, activation of NLRP3 inflammasome and release of interleukin (IL)-1ß and IL-18 cytokines, while downregulation of USP9X could reverse these alterations. USP9X was found to have marked impact on NLRP3 protein instead of mRNA level. Furthermore, increased ubiquitination of NLRP3 was observed upon downregulating USP9X. Additionally, the inhibitory effect of USP9X downregulation was reversed by NLRP3 overexpression, while the promoting impact of USP9X overexpression was dampened by NLRP3 inhibitor in terms of cell pyroptosis and cytokine secretion. USP9X modulated the activity of NLRP3 inflammasome and pyroptosis of AECs via its deubiquitination function.


Sujet(s)
Lésion pulmonaire aigüe , Protéine-3 de la famille des NLR contenant un domaine pyrine , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Inflammasomes/métabolisme , Lipopolysaccharides/pharmacologie , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/métabolisme , Adénosine triphosphate , Ubiquitin thiolesterase
19.
Cancer Lett ; 552: 215984, 2023 01 01.
Article de Anglais | MEDLINE | ID: mdl-36330954

RÉSUMÉ

The neomorphic transcription factor EWS-FLI1 is a key driver of Ewing sarcoma. Ablation of EWS-FLI1 may present a promising therapeutic strategy for this malignancy. Here we found that the deubiquitinase, ubiquitin specific peptidase 9 X-linked (USP9X) stabilizes EWS-FLI1 protein expression in Ewing sarcoma. We show that USP9X binds the ETS domain of EWS-FLI1 in Ewing sarcoma cells and deubiquitinates EWS-FLI1 and that USP9X and EWS-FLI1 protein expression is correlated in clinical Ewing sarcoma specimens. We found that treatment of Ewing sarcoma cells with the USP9X inhibitor WP1130 mediates rapid EWS-FLI1 degradation in vitro and in vivo which coincides with reduced growth of Ewing sarcoma cells and tumors. Our results suggest that USP9X might be a potential therapeutic target to mediate EWS-FLI1 depletion in Ewing sarcoma.


Sujet(s)
Sarcome d'Ewing , Humains , Sarcome d'Ewing/traitement médicamenteux , Sarcome d'Ewing/génétique , Sarcome d'Ewing/anatomopathologie , Lignée cellulaire tumorale , Protéine EWS de liaison à l'ARN/génétique , Protéine EWS de liaison à l'ARN/métabolisme , Protéine proto-oncogène c-fli-1/génétique , Protéine proto-oncogène c-fli-1/métabolisme , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Transformation cellulaire néoplasique , Régulation de l'expression des gènes tumoraux , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme
20.
Tumor ; (12): 935-946, 2023.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-1030343

RÉSUMÉ

Objective:To explore whether the deubiquitinating enzyme deubiquitin ligase 9X(USP9X)affects the proliferation of nasopharyngeal carcinoma(NPC)cells via regulating forkhead box Ml(FOXM1)mediated glycolysis. Methods:Western blotting and real-time fluorescence quantitative PCR,respectively,were used to detect the protein and mRNA expression levels of USP9X in NPC tissue and normal nasal mucosa tissue.NPC cells(CNE2 and HNE2)were infected with lentivirus carrying specific shRNA targeting USP9X gene(shUSP9X)or full-length USP9X gene(Flag-USP9X)for USP9X overexpression.CCK-8 assay was used to evaluate the effect of USP9X silencing or overexpression on the proliferation of NPC cells.Cellular energy metabolism assay and extracellular acidification rate(ECAR)assay were performed to investigate the impact of USP9X alteration on the glycolysis of NPC cells.The interaction between USP9X and FOXM1 was analyzed using ubibrowser_v3/database,co-immunoprecipitation,and ZDOCK software,and the changes in FOXM1 protein ubiquitination levels upon USP9X silencing was examined.Real-time fluorescence quantitative PCR and Western blotting,respectively,were used to detect the effect of USP9X alteration on the expression of FOXM1 mRNA and protein in NPC cells.Finally,FOXM1 expression was restored in USP9X silencing CNE2 cells by expression of recombinant plasmid Flag-FOXM1 carrying full-length FOXM1 gene through lentiviral infection.CCK-8 assay was used to evaluate the effect of FOXM1 upregulation on the proliferation of CNE2 cells.Cellular energy metabolism assay and ECAR assay were used to investigate the effect of FOXM1 upregulation on the glycolysis of CNE2 cells. Results:Compared with normal nasal mucosa tissue,the expression levels of USP9X mRNA and protein were significantly increased in NPC tissues(P<0.05).After USP9X downregulation,the proliferation and glycolysis of CNE2 cells was significantly inhibited as indicated by the results of CCK-8 assay,cellular energy metabolism assay and ECAR assay(P<0.05).In contrast,the proliferation and glycolysis of HNE2 cells was significantly enhanced after USP9X upregulation(P<0.05).The interaction between USP9X and FOXM1 was confirmed by ubibrowser_v3/database,co-immunoprecipitation,and ZDOCK software analysis.Silencing USP9X could significantly increase FOXM1 ubiquitination in CNE2 cells.Overexpressing FOXM1 in low-USP9X CNE2 cells restored the proliferation and glycolysis activity of NPC cells(P<0.05). Conclusion:USP9X can enhance NPC cell glycolysis by inhibiting FOXM1 ubiquitination and subsequent degradation,thereby promoting NCP cell proliferation.USP9X may be a potential novel therapeutic target for the treatment of NPC.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE