Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 786
Filtrer
1.
Gen Physiol Biophys ; 43(4): 301-312, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953570

RÉSUMÉ

Vascular endothelial growth factor A (VEGFA) is an important regulator for non-small cell lung cancer (NSCLC). Our study aimed to reveal its upstream pathway to provide new ideas for developing the therapeutic targets of NSCLC. The mRNA and protein levels of VEGFA, ubiquitin-specific peptidase 35 (USP35), and FUS were determined by quantitative real-time PCR and Western blot. Cell proliferation, apoptosis, invasion and angiogenesis were detected using CCK8 assay, EdU assay, flow cytometry, transwell assay and tube formation assay. The interaction between USP35 and VEGFA was assessed by Co-IP assay and ubiquitination assay. Animal experiments were performed to assess USP35 and VEGFA roles in vivo. VEGFA had elevated expression in NSCLC tissues and cells. Interferences of VEGFA inhibited NSCLC cell proliferation, invasion, angiogenesis, and increased apoptosis. USP35 could stabilize VEGFA protein level by deubiquitination, and USP35 knockdown suppressed NSCLC cell growth, invasion and angiogenesis via reducing VEGFA expression. FUS interacted with USP35 to promote its mRNA stability, thereby positively regulating VEGFA expression. Also, USP35 silencing could reduce NSCLC tumorigenesis by downregulating VEGFA. FUS-stabilized USP35 facilitated NSCLC cell growth, invasion and angiogenesis through deubiquitinating VEGFA, providing a novel idea for NSCLC treatment.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Tumeurs du poumon , Invasion tumorale , Néovascularisation pathologique , Protéine FUS de liaison à l'ARN , Ubiquitination , Facteur de croissance endothéliale vasculaire de type A , Humains , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Protéine FUS de liaison à l'ARN/métabolisme , Protéine FUS de liaison à l'ARN/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Prolifération cellulaire/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Invasion tumorale/génétique , Lignée cellulaire tumorale , Souris , Animaux , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Souris nude ,
2.
Allergol Immunopathol (Madr) ; 52(4): 60-67, 2024.
Article de Anglais | MEDLINE | ID: mdl-38970266

RÉSUMÉ

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is a familiar disease, and owns high morbidity and mortality, which critically damages the health of patients. Ubiquitin-specific peptidase 8 (USP8) is a pivotal protein to join in the regulation of some diseases. In a previous report, it was determined that USP8 expression is down-regulated in LPS-treated BEAS-2B cells, and USP8 restrains inflammatory response and accelerates cell viability. However, the regulatory roles of USP8 on ferroptosis in COPD are rarely reported, and the associated molecular mechanisms keep vague. OBJECTIVE: To investigate the regulatory functions of USP8 in COPD progression. MATERIAL AND METHODS: The lung functions were measured through the Buxco Fine Pointe Series Whole Body Plethysmography (WBP). The Fe level was tested through the Fe assay kit. The protein expressions were assessed through western blot. The levels of tumor necrosis -factor-α, interleukin 6, and interleukin 8 were evaluated through enzyme-linked immunosorbent serologic assay. Cell viability was tested through CCK-8 assay. RESULTS: In this work, it was discovered that overexpression of USP8 improved lung function in COPD mice. In addition, overexpression of USP8 repressed ferroptosis by regulating glutathione peroxidase 4 and acyl-CoA synthetase long-chain family 4 expressions in COPD mice. Overexpression of USP8 suppressed inflammation in COPD mice. Furthermore, overexpression of USP8 suppressed ferroptosis in COPD cell model. At last, it was verified that overexpression of USP8 accelerated ubiquitin aldehyde-binding protein 1 (OTUB1)/solute carrier family 7 member 11 (SLC7A11) pathway. CONCLUSION: This study manifested that overexpression of USP8 restrained inflammation and ferroptosis in COPD by regulating the OTUB1/SLC7A11 signaling pathway. This discovery hinted that USP8 could be a potential target for COPD treatment.


Sujet(s)
Système y+ de transport d'acides aminés , Ferroptose , Broncho-pneumopathie chronique obstructive , Transduction du signal , Ubiquitin thiolesterase , Ferroptose/physiologie , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/génétique , Animaux , Humains , Souris , Transduction du signal/immunologie , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Mâle , Inflammation/métabolisme , Inflammation/immunologie , Modèles animaux de maladie humaine , Souris de lignée C57BL , Complexes de tri endosomique requis pour le transport/métabolisme , Complexes de tri endosomique requis pour le transport/génétique , Lignée cellulaire , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Endopeptidases
3.
Clin Respir J ; 18(7): e13799, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38987867

RÉSUMÉ

BACKGROUND: Mitochondrial ribosomal protein L35 (MRPL35) has been reported to contribute to the growth of non-small cell lung cancer (NSCLC) cells. However, the functions and mechanisms of MRPL35 on glutamine metabolism in NSCLC remain unclear. METHODS: The detection of mRNA and protein of MRPL35, ubiquitin-specific protease 39 (USP39), and solute carrier family 7 member 5 (SLC7A5) was conducted using qRT-PCR and western blotting. Cell proliferation, apoptosis, and invasion were evaluated using the MTT assay, EdU assay, flow cytometry, and transwell assay, respectively. Glutamine metabolism was analyzed by detecting glutamine consumption, α-ketoglutarate level, and glutamate production. Cellular ubiquitination analyzed the deubiquitination effect of USP39 on MRPL35. An animal experiment was conducted for in vivo analysis. RESULTS: MRPL35 was highly expressed in NSCLC tissues and cell lines, and high MRPL35 expression predicted poor outcome in NSCLC patients. In vitro analyses suggested that MRPL35 knockdown suppressed NSCLC cell proliferation, invasion, and glutamine metabolism. Moreover, MRPL35 silencing hindered tumor growth in vivo. Mechanistically, USP39 stabilized MRPL35 expression by deubiquitination and then promoted NSCLC cell proliferation, invasion, and glutamine metabolism. In addition, MRPL35 positively affected SLC7A5 expression in NSCLC cells in vitro and in vivo. Moreover, the anticancer effects of MRPL35 silencing could be rescued by SLC7A5 overexpression in NSCLC cells. CONCLUSION: MRPL35 expression was stabilized by USP39-induced deubiquitination in NSCLC cells, and knockdown of MRPL35 suppressed NSCLC cell proliferation, invasion, and glutamine metabolism in vitro and impeded tumor growth in vivo by upregulating SLC7A5, providing a promising therapeutic target for NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Glutamine , Tumeurs du poumon , Invasion tumorale , Régulation positive , Humains , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Prolifération cellulaire/physiologie , Glutamine/métabolisme , Souris , Animaux , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Mâle , Apoptose , Femelle , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique
4.
J Obstet Gynaecol ; 44(1): 2361862, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38916982

RÉSUMÉ

BACKGROUND: Ovarian cancer stands as a highly aggressive malignancy. The core aim of this investigation is to uncover genes pivotal to the progression and prognosis of ovarian cancer, while delving deep into the intricate mechanisms that govern their impact. METHODS: The study entailed the retrieval of RNA-seq data and survival data from the XENA database. Outliers were meticulously excluded in accordance with TCGA guidelines and through principal components analysis. The R package 'deseq2' was harnessed to extract differentially expressed genes. WGCNA was employed to prioritise these genes, and Cox regression analysis and survival analysis based on disease-specific time were conducted to identify significant genes. Immunohistochemistry validation was undertaken to confirm the distinct expression of USP43. Furthermore, the influence of USP43 on the biological functions of ovarian cancer cells was explored using techniques such as RNA interference, western blotting, scratch assays, and matrigel invasion assays. The examination of immune infiltration was facilitated via CIBERSORT. RESULTS: The study unearthed 5195 differentially expressed genes between ovarian cancer and normal tissue, comprising 3416 up-regulated and 1779 down-regulated genes. WGCNA pinpointed 204 genes most intimately tied to tumorigenesis. The previously undisclosed gene USP43 exhibited heightened expression in tumour tissues and exhibited associations with overall survival and disease-specific survival. USP43 emerged as a driver of cell migration (43.27 ± 3.91% vs 19.69 ± 1.94%) and invasion ability (314 ± 32 vs 131 ± 12) through the mechanism of epithelial mesenchymal transition, potentially mediated by the KRAS pathway. USP43 was also identified as a booster of CD4+ T memory resting cell infiltration, while concurrently reducing M1 macrophages within cancer, thereby fostering a milieu with relatively immune suppressive traits. Interestingly, USP43 demonstrated connections with epigenetically regulated-mRNAsi, although not with mRNAsi. CONCLUSION: This study underscores the role of USP43 in facilitating tumour migration and invasion. It postulates USP43 as a novel therapeutic target for ovarian cancer treatment.


Ovarian cancer is the most deadly tumour among all gynecological tumours. Thus we tried to explore the relevant mechanism of ovarian cancer because its occurrence and development mechanism has not been fully elucidated. We used bioinformatics methods to perform differential gene analysis on ovarian cancer tissues and normal tissues, and used methods such as WGCNA and COX regression analysis to find the gene USP43 related to tumour development and prognosis. USP43 is a gene that has not been studied in ovarian cancer before. Through RNA interference technology, we found that it can promote the migration and invasion ability of ovarian cancer and promote epithelial-mesenchymal transition of ovarian cancer cells. In addition, this gene has also been proven to be related to tumour immunity and tumour stemness. These results indicate that USP43 can promote the tumorigenesis of ovarian cancer and can be used as a drug target.


Sujet(s)
Cystadénocarcinome séreux , Régulation de l'expression des gènes tumoraux , Tumeurs de l'ovaire , Ubiquitin-specific proteases , Femelle , Humains , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Ubiquitin-specific proteases/génétique , Ubiquitin-specific proteases/métabolisme , Cystadénocarcinome séreux/génétique , Cystadénocarcinome séreux/métabolisme , Cystadénocarcinome séreux/anatomopathologie , Lignée cellulaire tumorale , Pronostic , Mouvement cellulaire/génétique , Transition épithélio-mésenchymateuse/génétique , Analyse de survie , Pertinence clinique
5.
Int Immunopharmacol ; 136: 112346, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38850785

RÉSUMÉ

Myocardial infarction (MI) is an event of heart attack due to the formation of plaques in the interior walls of the arteries. This study is conducted to explore the role of ubiquitin-specific peptidase 47 (USP47) in cardiac function and inflammatory immunity. MI mouse models were established, followed by an appraisal of cardiac functions, infarct size, pathological changes, and USP47 and NLRP3 levels. MI cell models were established in HL-1 cells using anoxia. Levels of cardiac function-associated proteins, USP7, interferon regulatory factor 1 (IRF1), platelet factor-4 (CXCL4), pyroptotic factors, and neutrophil extracellular traps (NETs) were determined. The bindings of IRF1 to USP47 and the CXCL4 promoter and the ubiquitination of IRF1 were analyzed. USP47 was upregulated in myocardial tissues of MI mice. USP47 inhibition alleviated cardiac functions, and decreased infarct size, pro-inflammatory cytokines, NETs, NLRP3, and pyroptosis. The ubiquitination and expression levels of IRF1 were increased by silencing USP47, and IRF1 bound to the CXCL4 promoter to promote CXCL4. Overexpression of IRF1 or CXCL4 in vitro and injection of Nigericin in vivo reversed the effect of silencing USP47 on alleviating pyroptosis and cardiac functions. Collectively, USP47 stabilized IRF1 and promoted CXCL4, further promoting pyroptosis, impairing cardiac functions, and aggravating immune inflammation through NLRP3 pathways.


Sujet(s)
Inflammasomes , Souris de lignée C57BL , Infarctus du myocarde , Protéine-3 de la famille des NLR contenant un domaine pyrine , Transduction du signal , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Infarctus du myocarde/immunologie , Infarctus du myocarde/métabolisme , Souris , Inflammasomes/métabolisme , Mâle , Pyroptose , Facteur-1 de régulation d'interféron/métabolisme , Facteur-1 de régulation d'interféron/génétique , Modèles animaux de maladie humaine , Lignée cellulaire , Pièges extracellulaires/métabolisme , Pièges extracellulaires/immunologie , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Facteur-4 plaquettaire/métabolisme , Facteur-4 plaquettaire/génétique , Ubiquitination , Humains
6.
Nat Commun ; 15(1): 5441, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937512

RÉSUMÉ

Recent studies have shown the crucial role of podocyte injury in the development of diabetic kidney disease (DKD). Deubiquitinating modification of proteins is widely involved in the occurrence and development of diseases. Here, we explore the role and regulating mechanism of a deubiquitinating enzyme, OTUD5, in podocyte injury and DKD. RNA-seq analysis indicates a significantly decreased expression of OTUD5 in HG/PA-stimulated podocytes. Podocyte-specific Otud5 knockout exacerbates podocyte injury and DKD in both type 1 and type 2 diabetic mice. Furthermore, AVV9-mediated OTUD5 overexpression in podocytes shows a therapeutic effect against DKD. Mass spectrometry and co-immunoprecipitation experiments reveal an inflammation-regulating protein, TAK1, as the substrate of OTUD5 in podocytes. Mechanistically, OTUD5 deubiquitinates K63-linked TAK1 at the K158 site through its active site C224, which subsequently prevents the phosphorylation of TAK1 and reduces downstream inflammatory responses in podocytes. Our findings show an OTUD5-TAK1 axis in podocyte inflammation and injury and highlight the potential of OTUD5 as a promising therapeutic target for DKD.


Sujet(s)
Néphropathies diabétiques , Inflammation , MAP Kinase Kinase Kinases , Souris knockout , Podocytes , Ubiquitination , Podocytes/métabolisme , Podocytes/anatomopathologie , Animaux , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Néphropathies diabétiques/génétique , Souris , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique , Humains , Mâle , Souris de lignée C57BL , Phosphorylation , Diabète expérimental/métabolisme , Diabète expérimental/anatomopathologie , Diabète expérimental/complications , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Cellules HEK293 , Enzymes de désubiquitinylation/métabolisme , Enzymes de désubiquitinylation/génétique
7.
Mol Med ; 30(1): 97, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937697

RÉSUMÉ

BACKGROUND: Ubiquitin-specific protease 38 (USP38), belonging to the USP family, is recognized for its role in controlling protein degradation and diverse biological processes. Ventricular arrhythmias (VAs) following heart failure (HF) are closely linked to ventricular electrical remodeling, yet the specific mechanisms underlying VAs in HF remain inadequately explored. In this study, we examined the impact of USP38 on VAs in pressure overload-induced HF. METHODS: Cardiac-specific USP38 knockout mice, cardiac-specific USP38 transgenic mice and their matched control littermates developed HF induced by aortic banding (AB) surgery. After subjecting the mice to AB surgery for a duration of four weeks, comprehensive investigations were conducted, including pathological analysis and electrophysiological assessments, along with molecular analyses. RESULTS: We observed increased USP38 expression in the left ventricle of mice with HF. Electrocardiogram showed that the USP38 knockout shortened the QRS interval and QTc, while USP38 overexpression prolonged these parameters. USP38 knockout decreased the susceptibility of VAs by shortening action potential duration (APD) and prolonging effective refractory period (ERP). In addition, USP38 knockout increased ion channel and Cx43 expression in ventricle. On the contrary, the increased susceptibility of VAs and the decreased expression of ventricular ion channels and Cx43 were observed with USP38 overexpression. In both in vivo and in vitro experiments, USP38 knockout inhibited TBK1/AKT/CAMKII signaling, whereas USP38 overexpression activated this pathway. CONCLUSION: Our data indicates that USP38 increases susceptibility to VAs after HF through TBK1/AKT/CAMKII signaling pathway, Consequently, USP38 may emerge as a promising therapeutic target for managing VAs following HF.


Sujet(s)
Défaillance cardiaque , Souris knockout , Ubiquitin-specific proteases , Remodelage ventriculaire , Animaux , Souris , Remodelage ventriculaire/génétique , Défaillance cardiaque/métabolisme , Défaillance cardiaque/étiologie , Défaillance cardiaque/génétique , Défaillance cardiaque/physiopathologie , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Modèles animaux de maladie humaine , Mâle , Troubles du rythme cardiaque/étiologie , Troubles du rythme cardiaque/métabolisme , Troubles du rythme cardiaque/génétique , Ventricules cardiaques/métabolisme , Ventricules cardiaques/physiopathologie , Souris transgéniques , Transduction du signal , Électrocardiographie
8.
Sci Rep ; 14(1): 13906, 2024 06 17.
Article de Anglais | MEDLINE | ID: mdl-38886545

RÉSUMÉ

Colon adenocarcinoma (COAD) is the second leading cause of cancer death, and there is still a lack of diagnostic biomarkers and therapeutic targets. In this study, bioinformatics analysis of the TCGA database was used to obtain RUNX1, a gene with prognostic value in COAD. RUNX1 plays an important role in many malignancies, and its molecular regulatory mechanisms in COAD remain to be fully understood. To explore the physiological role of RUNX1, we performed functional analyses, such as CCK-8, colony formation and migration assays. In addition, we investigated the underlying mechanisms using transcriptome sequencing and chromatin immunoprecipitation assays. RUNX1 is highly expressed in COAD patients and significantly correlates with survival. Silencing of RUNX1 significantly slowed down the proliferation and migratory capacity of COAD cells. Furthermore, we demonstrate that CDC20 and MCM2 may be target genes of RUNX1, and that RUNX1 may be physically linked to the deubiquitinating enzyme USP31, which mediates the upregulation of RUNX1 protein to promote transcriptional function. Our results may provide new insights into the mechanism of action of RUNX1 in COAD and reveal potential therapeutic targets for this disease.


Sujet(s)
Protéines Cdc20 , Sous-unité alpha 2 du facteur CBF , Régulation de l'expression des gènes tumoraux , Composant-2 du complexe de maintenance des minichromosomes , Ubiquitination , Humains , Sous-unité alpha 2 du facteur CBF/métabolisme , Sous-unité alpha 2 du facteur CBF/génétique , Protéines Cdc20/métabolisme , Protéines Cdc20/génétique , Composant-2 du complexe de maintenance des minichromosomes/métabolisme , Composant-2 du complexe de maintenance des minichromosomes/génétique , Lignée cellulaire tumorale , Tumeurs du côlon/génétique , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Prolifération cellulaire/génétique , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Évolution de la maladie , Mouvement cellulaire/génétique
9.
Nat Commun ; 15(1): 5266, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902237

RÉSUMÉ

Functionally characterizing the genetic alterations that drive pancreatic cancer is a prerequisite for precision medicine. Here, we perform somatic CRISPR/Cas9 mutagenesis screens to assess the transforming potential of 125 recurrently mutated pancreatic cancer genes, which revealed USP15 and SCAF1 as pancreatic tumor suppressors. Mechanistically, we find that USP15 functions in a haploinsufficient manner and that loss of USP15 or SCAF1 leads to reduced inflammatory TNFα, TGF-ß and IL6 responses and increased sensitivity to PARP inhibition and Gemcitabine. Furthermore, we find that loss of SCAF1 leads to the formation of a truncated, inactive USP15 isoform at the expense of full-length USP15, functionally coupling SCAF1 and USP15. Notably, USP15 and SCAF1 alterations are observed in 31% of pancreatic cancer patients. Our results highlight the utility of in vivo CRISPR screens to integrate human cancer genomics and mouse modeling for the discovery of cancer driver genes with potential prognostic and therapeutic implications.


Sujet(s)
Systèmes CRISPR-Cas , Tumeurs du pancréas , Animaux , Humains , Souris , Lignée cellulaire tumorale , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Désoxycytidine/usage thérapeutique , , Régulation de l'expression des gènes tumoraux , Mutation , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Ubiquitin-specific proteases/génétique , Ubiquitin-specific proteases/métabolisme
10.
Eur J Med Chem ; 275: 116568, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-38889606

RÉSUMÉ

USP1 has emerged as a novel and potential target for drug discovery in single therapeutic agents or combination with chemotherapy and molecular targeted therapy. In this study, based on the disclosed structure of ML323 and KSQ-4279, we designed and synthesized a series of pyrido[2,3-d]pyrimidin-7(8H)-one derivatives as potent USP1 inhibitors by cyclization strategy and the systematic structure-activity relationship exploration was conducted. The representative compounds 1k, 1m and 2d displayed excellent USP1/UAF inhibition and exhibited strong antiproliferation effect in NCI-H1299 cells. Further flow cytometry analysis revealed that they could arrest breast cancer cells MDA-MB-436 in the S phase. Inhibition mechanism study of compound 1m indicated these derivatives acted as reversible and noncompetitive USP1 inhibitors. Of note, the combination of compound 1m with PARP inhibitor olaparib generated enhanced cell killing in olaparib-resistant MDA-MB-436/OP cells, and compound 1m exhibited excellent oral pharmacokinetic properties in mice. Overall, our efforts may provide a reliable basis for the development of novel USP1 inhibitor as a single therapeutic agent and in combination with PARP inhibitors.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Conception de médicament , Tests de criblage d'agents antitumoraux , Pyrimidinones , Humains , Relation structure-activité , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Animaux , Pyrimidinones/pharmacologie , Pyrimidinones/composition chimique , Pyrimidinones/synthèse chimique , Structure moléculaire , Souris , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Antienzymes/pharmacologie , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Ubiquitin-specific proteases/antagonistes et inhibiteurs , Ubiquitin-specific proteases/métabolisme
11.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(3): 670-678, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-38926952

RÉSUMÉ

OBJECTIVE: To investigate the clinical significance, functional role and potential downstream mechanism of USP5 in acute myeloid leukemia (AML). METHODS: The expression of USP5 in AML and normal tissues and its correlation with patients' survival were analyzed based on TCGA database. USP5 was knocked down and overexpressed in Jurkat and HL-60 cells using lentivirus. USP5 mRNA and protein expression were detected by RT-qPCR and Western blot, respectively. Cell proliferation and growth were measured by CCK-8 and methylcellulose colony-forming assay. Flow cytometry was used to analyze cell cycle and apoptosis. RESULTS: USP5 was highly expression in AML compared with normal tissues. Up-regulation of USP5 was negatively correlated with the survival of AML patients. USP5 knockdown and overexpression inhibited and promoted the proliferation and colony growth of AML cells, respectively. Cell cycle arrest and apoptosis were induced in USP5 knockdown Jurkat and HL-60 cells. Furthermore, USP5 knockdown inhibited the phosphrylation of AKT, mTOR and 4EBP1. CONCLUSION: Overexpression of USP5 predicts poor survival of AML patients. Targeting USP5 suppresses AKT/mTOR/4EBP1 signaling and reduces the proliferation and growth of AML cells.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Apoptose , Prolifération cellulaire , Leucémie aigüe myéloïde , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Humains , Leucémie aigüe myéloïde/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Cellules HL-60 , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Cellules Jurkat , Ubiquitin-specific proteases/métabolisme , Pertinence clinique
12.
Int J Biol Sci ; 20(8): 2943-2964, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904030

RÉSUMÉ

Background: Shear stress-induced Dickkopf-1 (DKK1) secretion by endothelial cells (ECs) promotes EC dysfunction and accelerates atherosclerosis (AS). However, the paracrine role of endothelial DKK1 in modulating adjacent smooth muscle cells (SMCs) in atherosclerosis remains unclear. This study investigated the role of EC-secreted DKK1 in SMC-derived foam cell formation under shear stress, in vitro and in vivo. Methods: Parallel-plate co-culture flow system was used to explore the cellular communication between ECs and SMCs under shear stress in vitro. Endothelium-specific knockout of DKK1 (DKK1ECKO/APOE-/-) and endothelium-specific overexpression of DKK1 (DKK1ECTg) mice were constructed to investigate the role of endothelial DKK1 in atherosclerosis and SMC-derived foam cell formation in vivo. RNA sequencing (RNA-seq) was used to identify the downstream targets of DKK1. Reverse transcription quantitative polymerase chain reaction (RT-qPCR), western blot, coimmunoprecipitation (Co-IP) assays and chromatin immunoprecipitation (ChIP) experiments were conducted to explore the underlying regulatory mechanisms. Results: DKK1 is transcriptionally upregulated in ECs under conditions of low shear stress, but not in co-cultured SMCs. However, DKK1 protein in co-cultured SMCs is increased via uptake of low shear stress-induced endothelial DKK1, thereby promoting lipid uptake and foam cell formation in co-cultured SMCs via the post-translational upregulation of scavenger receptor-A (SR-A) verified in parallel-plate co-culture flow system, DKK1ECKO and DKK1ECTg mice. RNA sequencing revealed that DKK1-induced SR-A upregulation in SMCs is dependent on Ubiquitin-specific Protease 53 (USP53), which bound to SR-A via its USP domain and cysteine at position 41, exerting deubiquitination to maintain the stability of the SR-A protein by removing the K48 ubiquitin chain and preventing proteasomal pathway degradation, thereby mediating the effect of DKK1 on lipid uptake in SMCs. Moreover, DKK1 regulates the transcription of USP53 by facilitating the binding of transcription factor CREB to the USP53 promoter. SMC-specific overexpression of USP53 via adeno-associated virus serotype 2 vectors in DKK1ECKO/APOE-/- mice reversed the alleviation of atherosclerotic plaque burden, SR-A expression and lipid accumulation in SMCs within plaques resulting from DKK1 deficiency. Conclusions: Our findings demonstrate that, endothelial DKK1, induced by pathological low shear stress, acts as an intercellular mediator, promoted the foam cell formation of SMCs. These results suggest that targeted intervention with endothelial DKK1 may confer beneficial effects on atherosclerosis.


Sujet(s)
Athérosclérose , Cellules spumeuses , Protéines et peptides de signalisation intercellulaire , Myocytes du muscle lisse , Animaux , Athérosclérose/métabolisme , Souris , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéines et peptides de signalisation intercellulaire/génétique , Cellules spumeuses/métabolisme , Myocytes du muscle lisse/métabolisme , Cellules endothéliales/métabolisme , Humains , Ubiquitination , Mâle , Techniques de coculture , Souris knockout , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Souris de lignée C57BL
13.
Technol Cancer Res Treat ; 23: 15330338241250298, 2024.
Article de Anglais | MEDLINE | ID: mdl-38706215

RÉSUMÉ

Objective: Ubiquitin-specific peptidase 39 (USP39) plays a carcinogenic role in many cancers, but little research has been conducted examining whether it is involved in head and neck squamous cell carcinoma (HNSCC). Therefore, this study explored the functional role of USP39 in HNSCC. Method: Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to identify differentially expressed proteins (DEPs) between the HNSCC tumor and adjacent healthy tissues. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were used to assess the functional enrichment of DEPs. Immunohistochemistry was used to detect protein expression. The viability and migration of two HNSCC cell lines, namely CAL27 and SCC25, were detected using the cell counting kit-8 assay and a wound healing assay, respectively. Quantitative real-time PCR was used to detect the expression level of signal transducer and activator of transcription 1 (STAT1) mRNA. Results: LC-MS/MS results identified 590 DEPs between HNSCC and adjacent tissues collected from 4 patients. Through GO and KEGG pathway analyses, 34 different proteins were found to be enriched in the spliceosome pathway. The expression levels of USP39 and STAT1 were significantly higher in HNSCC tumor tissue than in adjacent healthy tissue as assessed by LC-MS/MS analysis, and the increased expression of USP39 and STAT1 protein was confirmed by immunohistochemistry in clinical samples collected from 7 additional patients with HNSCC. Knockdown of USP39 or STAT1 inhibited the viability and migration of CAL27 and SCC25 cells. In addition, USP39 knockdown inhibited the expression of STAT1 mRNA in these cells. Conclusion: Our findings indicated that USP39 knockdown may inhibit HNSCC viability and migration by suppressing STAT1 expression. The results of this study suggest that USP39 may be a potential new target for HNSCC clinical therapy or a new biomarker for HNSCC.


Sujet(s)
Mouvement cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs de la tête et du cou , Facteur de transcription STAT-1 , Carcinome épidermoïde de la tête et du cou , Ubiquitin-specific proteases , Humains , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique , Mouvement cellulaire/génétique , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme , Lignée cellulaire tumorale , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/métabolisme , Survie cellulaire/génétique , Spectrométrie de masse en tandem , Prolifération cellulaire , Chromatographie en phase liquide , Femelle , Mâle , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Protéomique/méthodes
14.
BMC Plant Biol ; 24(1): 404, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38750451

RÉSUMÉ

BACKGROUND: Ubiquitin-specific proteases (UBPs) are a large family of deubiquitinating enzymes (DUBs). They are widespread in plants and are critical for plant growth, development, and response to external stresses. However, there are few studies on the functional characteristics of the UBP gene family in the important staple crop, maize (Zea mays L.). RESULTS: In this study, we performed a bioinformatic analysis of the entire maize genome and identified 45 UBP genes. Phylogenetic analysis indicated that 45 ZmUBP genes can be divided into 15 subfamilies. Analysis of evolutionary patterns and divergence levels indicated that ZmUBP genes were present before the isolation of dicotyledons, were highly conserved and subjected to purifying selection during evolution. Most ZmUBP genes exhibited different expression levels in different tissues and developmental stages. Based on transcriptome data and promoter element analysis, we selected eight ZmUBP genes whose promoters contained a large number of plant hormones and stress response elements and were up-regulated under different abiotic stresses for RT-qPCR analysis, results showed that these genes responded to abiotic stresses and phytohormones to varying degrees, indicating that they play important roles in plant growth and stress response. CONCLUSIONS: In this study, the structure, location and evolutionary relationship of maize UBP gene family members were analyzed for the first time, and the ZmUBP genes that may be involved in stress response and plant growth were identified by combining promoter element analysis, transcriptome data and RT-qPCR analysis. This study informs research on the involvement of maize deubiquitination in stress response.


Sujet(s)
Régulation de l'expression des gènes végétaux , Génome végétal , Famille multigénique , Phylogenèse , Ubiquitin-specific proteases , Zea mays , Zea mays/génétique , Zea mays/enzymologie , Ubiquitin-specific proteases/génétique , Ubiquitin-specific proteases/métabolisme , Protéines végétales/génétique , Protéines végétales/métabolisme , Stress physiologique/génétique , Gènes de plante , Analyse de profil d'expression de gènes , Régions promotrices (génétique)/génétique
15.
Mol Cancer ; 23(1): 88, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702734

RÉSUMÉ

Drug resistance represents a significant obstacle in cancer treatment, underscoring the need for the discovery of novel therapeutic targets. Ubiquitin-specific proteases (USPs), a subclass of deubiquitinating enzymes, play a pivotal role in protein deubiquitination. As scientific research advances, USPs have been recognized as key regulators of drug resistance across a spectrum of treatment modalities, including chemotherapy, targeted therapy, immunotherapy, and radiotherapy. This comprehensive review examines the complex relationship between USPs and drug resistance mechanisms, focusing on specific treatment strategies and highlighting the influence of USPs on DNA damage repair, apoptosis, characteristics of cancer stem cells, immune evasion, and other crucial biological functions. Additionally, the review highlights the potential clinical significance of USP inhibitors as a means to counter drug resistance in cancer treatment. By inhibiting particular USP, cancer cells can become more susceptible to a variety of anti-cancer drugs. The integration of USP inhibitors with current anti-cancer therapies offers a promising strategy to circumvent drug resistance. Therefore, this review emphasizes the importance of USPs as viable therapeutic targets and offers insight into fruitful directions for future research and drug development. Targeting USPs presents an effective method to combat drug resistance across various cancer types, leading to enhanced treatment strategies and better patient outcomes.


Sujet(s)
Antinéoplasiques , Résistance aux médicaments antinéoplasiques , Tumeurs , Ubiquitin-specific proteases , Humains , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Ubiquitin-specific proteases/antagonistes et inhibiteurs , Ubiquitin-specific proteases/métabolisme , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Animaux , Thérapie moléculaire ciblée , Réparation de l'ADN , Apoptose/effets des médicaments et des substances chimiques
16.
Int Immunopharmacol ; 135: 112274, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38772301

RÉSUMÉ

OBJECTIVE: The current treatment and mechanism of Sjogren's syndrome (SS) are unclear. The purpose of the present study was to potential molecular mechanisms of SS. METHODS: Immunohistochemical and immunofluorescence techniques reveal the targets and therapeutic approaches of SS. RESULTS: We found through molecular biology techniques such as immunoblotting and immunoprecipitation that USP5 is a novel regulator of NLRP3 involvement in the pathological process of SS. USP5 was significantly downregulated in submandibular gland tissue of SS. Meanwhile, it was found that USP5 is a negative regulator of NLRP3 via ubiquitination NLRP3. In addition, SalvianolicacidB (SaB), a natural USP5 agonist, can alleviate ss by regulating the USP5/NLRP3 signaling pathway. CONCLUSION: Therefore, this study provides a new mechanism for SS and also provides new therapeutic targets for treating SS.


Sujet(s)
Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Syndrome de Gougerot-Sjögren , Syndrome de Gougerot-Sjögren/métabolisme , Syndrome de Gougerot-Sjögren/immunologie , Syndrome de Gougerot-Sjögren/anatomopathologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Animaux , Humains , Inflammasomes/métabolisme , Femelle , Glande submandibulaire/anatomopathologie , Glande submandibulaire/métabolisme , Ubiquitination , Transduction du signal , Souris , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Souris de lignée C57BL , Mâle
17.
Cell Signal ; 120: 111225, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38735506

RÉSUMÉ

Ubiquitin-specific proteases (USPs) have been proved to play important roles in the progression of diabetic retinopathy. In this study, we explored the role of USP5 and its possible mechanisms in diabetic retinopathy development. Cell proliferation, apoptosis, inflammation and oxidative stress were determined using CCK-8 assay, EdU staining assay, flow cytometry, and ELISA, respectively. The mRNA and protein expression of ROBO4 and USP5 were measured through RT-qPCR and western blot, respectively. Co-IP and deubiquitination assay were conducted to evaluate the interaction between ROBO4 and USP5. The results showed that high glucose (HG) stimulation significantly led to HRPE cell damage as described by suppressing proliferation, and promoting oxidative stress, inflammation and apoptosis. ROBO4 was markedly increased in diabetic retinopathy plasma samples and HG-triggered HRPE cells. Depletion of ROBO4 could alleviate HG-caused HRPE cell damage. USP5 was also significantly elevated in diabetic retinopathy plasma samples and HG-triggered HRPE cells. USP5 overexpression aggravated HG-induced HRPE cell damage. USP5 stabilized ROBO4 through deubiquitination. Moreover, USP5 knockdown decreased ROBO4 expression to mitigate HG-triggered cell damage in HRPE cells. USP5 stabilized ROBO4 via deubiquitination to repress cell proliferation, and facilitate inflammation, cell apoptosis and oxidative stress in HG-treated HRPE cells, thereby promoting the development of diabetic retinopathy.


Sujet(s)
Rétinopathie diabétique , Récepteurs de surface cellulaire , Ubiquitination , Rétinopathie diabétique/métabolisme , Rétinopathie diabétique/anatomopathologie , Humains , Récepteurs de surface cellulaire/métabolisme , Apoptose , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Prolifération cellulaire , Stress oxydatif , Animaux , Glucose/métabolisme , Glucose/pharmacologie , Lignée cellulaire ,
18.
Oncogene ; 43(26): 2025-2037, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38744954

RÉSUMÉ

Lung adenocarcinoma (LUAD) is one of the most prevalent types of cancer. Ubiquitination is crucial in modulating cell proliferation and aerobic glycolysis in cancer. The frequency of TP53 mutations in LUAD is approximately 50%. Currently, therapeutic targets for wild-type (WT) p53-expressing LUAD are limited. In the present study, we systemically explored the expression of ubiquitin-specific protease genes using public datasets. Then, we focused on ubiquitin-specific protease 54 (USP54), and explored its prognostic significance in LUAD patients using public datasets, analyses, and an independent cohort from our center. We found that the expression of USP54 was lower in LUAD tissues compared with that in the paracancerous tissues. Low USP54 expression levels were linked to a malignant phenotype and worse survival in patients with LUAD. The results of functional experiments revealed that up-regulation of USP54 suppressed LUAD cell proliferation in vivo and in vitro. USP54 directly interacted with p53 protein and the levels of ubiquitinated p53 were inversely related to USP54 levels, consistent with a role of USP54 in deubiquitinating p53 in p53-WT LUAD cells. Moreover, up-regulation of the USP54 expression inhibited aerobic glycolysis in LUAD cells. Importantly, we confirmed that USP54 inhibited aerobic glycolysis and the growth of tumor cells by a p53-mediated decrease in glucose transporter 1 (GLUT1) expression in p53-WT LUAD cells. Altogether, we determined a novel mechanism of survival in the p53-WT LUAD cells to endure the malnourished tumor microenvironment and provided insights into the role of USP54 in the adaptation of p53-WT LUAD cells to metabolic stress.


Sujet(s)
Adénocarcinome pulmonaire , Transporteur de glucose de type 1 , Glycolyse , Tumeurs du poumon , Protéine p53 suppresseur de tumeur , Animaux , Femelle , Humains , Mâle , Souris , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Transporteur de glucose de type 1/métabolisme , Transporteur de glucose de type 1/génétique , Glycolyse/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Souris nude , Pronostic , Protéolyse , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Ubiquitination
19.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167132, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38565386

RÉSUMÉ

The Epstein-Barr virus (EBV) is implicated in several cancers, including EBV-associated gastric cancer (EBVaGC). This study focuses on EBV-encoded BALF1 (BamH1 A fragment leftward reading frame 1), a key apoptosis regulator in EBV-related cancers, whose specific impact on EBVaGC was previously unknown. Our findings indicate that BALF1 overexpression in gastric cancer cells significantly enhances their proliferation, migration, and resistance to chemotherapy-induced apoptosis, confirming BALF1's oncogenic potential. A novel discovery is that BALF1 undergoes degradation via the ubiquitin-proteasome pathway. Through analysis of 69 deubiquitinating enzymes (DUBs), ovarian tumor protease (OTU) domain-containing protein 1 (OTUD1) emerged as a vital regulator for maintaining BALF1 protein stability. Furthermore, BALF1 was found to play a role in regulating the stability of the B-cell lymphoma-2 (Bcl-2) protein, increasing its levels through deubiquitination. This mechanism reveals BALF1's multifaceted oncogenic role in gastric cancer, as it contributes both directly and indirectly to cancer progression, particularly by stabilizing Bcl-2, known for its anti-apoptotic characteristics. These insights significantly deepen our understanding of EBV's involvement in the pathogenesis of gastric cancer. The elucidation of OTUD1's role in BALF1 regulation and its influence on Bcl-2 stabilization provide new avenues for therapeutic intervention in EBVaGC, bridging the gap between viral oncogenesis and cellular protein regulation and offering a more holistic view of gastric cancer development under the influence of EBV.


Sujet(s)
Apoptose , Protéines proto-oncogènes c-bcl-2 , Tumeurs de l'estomac , Ubiquitination , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/virologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique , Lignée cellulaire tumorale , Herpèsvirus humain de type 4/métabolisme , Herpèsvirus humain de type 4/génétique , Protéines virales/métabolisme , Protéines virales/génétique , Prolifération cellulaire , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/métabolisme , Infections à virus Epstein-Barr/anatomopathologie , Infections à virus Epstein-Barr/génétique , Stabilité protéique , Mouvement cellulaire , Animaux , Enzymes de désubiquitinylation/métabolisme , Enzymes de désubiquitinylation/génétique , Protéines virales régulatrices ou accessoires
20.
Cancer Lett ; 593: 216875, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38643837

RÉSUMÉ

Mesenchymal glioma stem cells (MES GSCs) are a subpopulation of cells in glioblastoma (GBM) that contribute to a worse prognosis owing to their highly aggressive nature and resistance to radiation therapy. Here, OCT4 is characterized as a critical factor in sustaining the stemness phenotype of MES GSC. We find that OCT4 is expressed intensively in MES GSC and is intimately associated with poor prognosis, moreover, OCT4 depletion leads to diminished invasive capacity and impairment of the stem phenotype in MES GSC. Subsequently, we demonstrated that USP5 is a deubiquitinating enzyme which directly interacts with OCT4 and preserves OCT4 stability through its deubiquitination. USP5 was additionally proven to be aberrantly over-expressed in MES GSCs, and its depletion resulted in a noticeable diminution of OCT4 and consequently a reduced self-renewal and tumorigenic capacity of MES GSCs, which can be substantially restored by ectopic expression of OCT4. In addition, we detected the dominant molecule that regulates USP5 transcription, E2F1, with dual luciferase reporter gene analysis. In combination, targeting the E2F1-USP5-OCT4 axis is a potentially emerging strategy for the therapy of GBM.


Sujet(s)
Tumeurs du cerveau , Facteur de transcription E2F1 , Cellules souches tumorales , Facteur de transcription Oct-3 , Ubiquitin-specific proteases , Humains , Facteur de transcription Oct-3/génétique , Facteur de transcription Oct-3/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Animaux , Facteur de transcription E2F1/métabolisme , Facteur de transcription E2F1/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Ubiquitin-specific proteases/génétique , Ubiquitin-specific proteases/métabolisme , Gliome/anatomopathologie , Gliome/génétique , Gliome/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Souris , Stabilité protéique , Glioblastome/anatomopathologie , Glioblastome/génétique , Glioblastome/métabolisme , Ubiquitination
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...