Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.703
Filter
1.
Genet Res (Camb) ; 2024: 3005195, 2024.
Article in English | MEDLINE | ID: mdl-39377096

ABSTRACT

Backgrounds: Glioma stands as one of the most formidable brain tumor types, with patient outcomes remaining bleak even in the face of advancements in treatment modalities. FBXW4, a constituent of the F-box and WD repeat domain-containing protein family, is recognized for its participation in diverse cellular activities, including those related to tumor dynamics. Yet, the therapeutic relevance and specific role of FBXW4 in the context of glioma are not well defined. This study aims to elucidate the functional dynamics and significance of FBXW4 in glioma cases. Methods: This research undertook a comprehensive analysis of FBXW4's expression patterns and clinical relevance in glioma by harnessing data from the TCGA and GTEx databases. Results: The investigation revealed a distinct downregulation of FBXW4 in glioma tissues compared to normal brain counterparts, with a pronounced correlation between FBXW4 levels and disease severity. Intriguingly, FBXW4 expression inversely related to WHO tumor grades, with the most advanced grade IV gliomas exhibiting the lowest FBXW4 levels, whereas grade II tumors demonstrated the highest. Cases presenting with IDH1/2 mutations or 1p/19q codeletions were also associated with elevated FBXW4 levels. Furthermore, diminished FBXW4 expression aligned with an increased risk of mortality. Conclusions: The findings suggest that FBXW4 holds promise as a prognostic marker and a potential therapeutic avenue in glioma management. Nonetheless, future research is imperative to decode the intricate signaling pathways involving FBXW4 and to understand its broader clinical ramifications in glioma treatment paradigms.


Subject(s)
Brain Neoplasms , F-Box Proteins , Glioma , Humans , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Prognosis , F-Box Proteins/genetics , F-Box Proteins/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Disease Progression , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Male , Female , Mutation
2.
Cell Death Dis ; 15(10): 735, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384743

ABSTRACT

High-grade serous ovarian carcinoma (HGSOC) is one of the most lethal gynecological cancer. Genetic studies have revealed gene copy number alterations (CNAs) frequently occurred in HGSOC pathogenesis, however the function and mechanism of CNAs for microRNAs are still not fully understood. Here, we show the dependence on gene copy number amplification of MIR937 that enhances cell autophagy and dictates HGSOC proliferative activity. Data mining of TCGA database revealed MIR937 amplification is correlated with increased MIR937 expression and cell proliferation of HGSOC. Deletion of MIR937 in HGSOC cells led to impaired autophagy and retarded cell proliferation, and the extent for its inhibitory effects scaled with the degree of MIR937 copy loss. Rescue assay confirmed miR-937-5p, a mature product of MIR937, was sufficient to restore its oncogenic function. Mechanistically, MIR937 amplification raised the expression of miR-937-5p, enhanced its binding to 3' UTR of FBXO16 transcript, and thereby restricting FBXO16 degradative effects on ULK1. Our results demonstrate that MIR937 amplification augments cell autophagy and proliferation, and suggest an alternative strategy of MIR937/FBXO16/ULK1 targeting for HGSOC treatment.


Subject(s)
Autophagy-Related Protein-1 Homolog , Autophagy , Cell Proliferation , F-Box Proteins , Gene Expression Regulation, Neoplastic , MicroRNAs , Ovarian Neoplasms , Humans , MicroRNAs/metabolism , MicroRNAs/genetics , Autophagy/genetics , Female , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , F-Box Proteins/metabolism , F-Box Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Gene Amplification , Animals
3.
Mol Plant Pathol ; 25(9): e70006, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39267531

ABSTRACT

The pathogen Agrobacterium tumefaciens is known for causing crown gall tumours in plants. However, it has also been harnessed as a valuable tool for plant genetic transformation. Apart from the T-DNA, Agrobacterium also delivers at least five virulence proteins into the host plant cells, which are required for an efficient infection. One of these virulence proteins is VirD5. F-box proteins, encoded in the host plant genome or the Ti plasmid, and the ubiquitin/26S proteasome system (UPS) also play an important role in facilitating Agrobacterium infection. Our study identified two Arabidopsis F-box proteins, D5BF1 and D5BF2, that bind VirD5 and facilitate its degradation via the UPS. Additionally, we found that Agrobacterium partially suppresses the expression of D5BF1 and D5BF2. Lastly, stable transformation and tumorigenesis efficiency assays revealed that D5BF1 and D5BF2 negatively regulate the Agrobacterium infection process, showing that the plant F-box proteins and UPS play a role in defending against Agrobacterium infection.


Subject(s)
Agrobacterium tumefaciens , Arabidopsis Proteins , Arabidopsis , F-Box Proteins , Transformation, Genetic , Arabidopsis/microbiology , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Agrobacterium tumefaciens/genetics , Agrobacterium tumefaciens/pathogenicity , F-Box Proteins/metabolism , F-Box Proteins/genetics , Carcinogenesis/genetics , Plant Tumors/microbiology , Proteasome Endopeptidase Complex/metabolism , Gene Expression Regulation, Plant
4.
Int J Med Sci ; 21(11): 2158-2169, 2024.
Article in English | MEDLINE | ID: mdl-39239542

ABSTRACT

Pancreatic cancer (PC) is a challenging and heterogeneous disease with a high mortality rate. Despite advancements in treatment, the prognosis for PC patients remains poor, with a high chance of disease recurrence. Biomarkers are crucial for diagnosing cancer, predicting patient prognosis and selecting treatments. However, the current lack of effective biomarkers for PC could contribute to the insufficiency of existing treatments. These findings underscore the urgent need to develop novel strategies to fight this disease. This study utilized multiple comprehensive bioinformatic analyses to identify potential therapeutic target genes in PC, focusing on histone lysine demethylases (KDMs). We found that high expression levels of KDM family genes, particularly KDM1A, KDM5A and KDM5B, were associated with improved overall survival in the cohort. Furthermore, the infiltration of various immune cells, including B cells, neutrophils, CD8+ T cells, dendritic cells, and macrophages, was positively correlated with KDM1A, KDM5A, and KDM5B expression. Moreover, MetaCore pathway analysis revealed interesting connections between KDM1A and the cell cycle and proliferation, between KDM5A and DNA damage and double-strand break repair through homologous recombination, and between KDM5B and WNT/ß-catenin signaling. These findings suggest that KDM1A, KDM5A and KDM5B may serve as promising biomarkers and therapeutic targets for PC, a disease of high importance due to its aggressive nature and urgent need for novel biomarkers to improve diagnosis and treatment.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Jumonji Domain-Containing Histone Demethylases , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Prognosis , Computational Biology , F-Box Proteins/metabolism , F-Box Proteins/genetics , Histone Demethylases/metabolism , Histone Demethylases/genetics , Molecular Targeted Therapy/methods , Retinoblastoma-Binding Protein 2/metabolism , Retinoblastoma-Binding Protein 2/genetics , Wnt Signaling Pathway/genetics , Cell Proliferation/genetics , Nuclear Proteins , Repressor Proteins
5.
Arch Esp Urol ; 77(7): 796-804, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39238305

ABSTRACT

BACKGROUNDS: F-box protein 45 (FBXO45) has been implicated in the progression of several diseases. Whether FBXO45 is involved in the development of bladder cancer remains unclear. Thus, this study focused on the effect of FBXO45 on the malignant progression of bladder cancer cells. METHODS: FBXO45 small-interference fragment was transfected into RT4 and 5637 cells by liposome-mediated transfection, and the knockdown efficiency of FBXO45 was verified by Western blot assay. The growth rate between FBXO45 knockdown cell lines and control cell lines was compared by counting kit 8 and plate cloning experiments. The motility of bladder cancer cells was observed via the Transwell test and Wound healing test. The effects of FBXO45 silencing on apoptosis and cell division were confirmed by flow cytometry. Western blot assay was performed to determine the function of FBXO45 knockdown on key proteins of cell apoptosis and the ERK/Cyclin D1/CDK4 pathway. RESULTS: After FBXO45 knockdown, the proliferation of bladder cancer cells was blocked (p < 0.01), and the migration and invasion abilities were reduced (p < 0.01). FBXO45 knockdown reduced the number of S-phase cells (RT4, p < 0.01; 5637, p < 0.05) and enhanced the apoptotic rate (p < 0.01). FBXO45 knockdown decreased the levels of p-ERK1/2, CDK4 and Cyclin D1 (p < 0.01). CONCLUSIONS: This study revealed that FBXO45 plays a carcinogenic role in bladder cancer via the ERK/Cyclin D1/CDK4 pathway, which provides a reference for the clinical treatment of patients with bladder cancer.


Subject(s)
Cyclin D1 , Cyclin-Dependent Kinase 4 , Disease Progression , F-Box Proteins , Gene Knockdown Techniques , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Humans , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin D1/metabolism , Cyclin D1/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , MAP Kinase Signaling System , Tumor Cells, Cultured , Cell Line, Tumor , Cell Proliferation
6.
Sci Rep ; 14(1): 22574, 2024 09 29.
Article in English | MEDLINE | ID: mdl-39343799

ABSTRACT

Emerging evidence have demonstrated that F-box only protein 2 (FBXO2) is intimately associated with malignant tumor development and occurrence. However, neither the functions nor the molecular mechanisms underlying FBXO2 have been determined in the papillary thyroid carcinoma (PTC). The quantitative real-time PCR (qRT-PCR), western blotting and immunohistochemistry were carried out to detect the FBXO2 expression in PTC tissues. CCK-8 assay, EdU assay and flow cytometry were used to assess cell proliferation, cell cycle and apoptosis. The trans-well assay was conducted to determine the cell invasiveness. The effect of FBXO2 on PTC cell proliferation in vivo was observed through a subcutaneous tumor formation experiment in nude mice. Immunoprecipitation were conducted to detect the interaction between FBXO2 and p53. The ubiquitination assays were conducted to assess the regulation of p53 ubiquitination by FBXO2. FBXO2 was overexpressed in both PTC tissues and cell lines. FBXO2 expression positively correlated with PTC tumor size, lymphatic metastasis, and extramembranous invasion. Furthermore, silencing FBXO2 inhibited PTC cell proliferation and promoted apoptosis. The overexpression of FBXO2 significantly promotes PTC cell proliferation. Mechanistic studies revealed that FBXO2 could directly bind to p53 and promote its ubiquitination degradation. Knockdown of p53 partially reversed the progression arrest induced by FBXO2 Knockdown in PTC cells. FBXO2 knockdown inhibited PTC cell proliferation and promoted apoptosis by targeting p53 for ubiquitination and degradation. This process represents a research foundation for its diagnostic and therapeutic applications.


Subject(s)
Apoptosis , Cell Proliferation , F-Box Proteins , Thyroid Cancer, Papillary , Thyroid Neoplasms , Tumor Suppressor Protein p53 , Ubiquitination , Adult , Animals , Female , Humans , Male , Mice , Middle Aged , Apoptosis/genetics , Cell Line, Tumor , Disease Progression , F-Box Proteins/metabolism , F-Box Proteins/genetics , Gene Expression Regulation, Neoplastic , Mice, Nude , Signal Transduction , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics
7.
EMBO Rep ; 25(10): 4206-4225, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39266770

ABSTRACT

SKP1-CUL1-F-box protein (SCF) ubiquitin ligases are versatile protein complexes that mediate the ubiquitination of protein substrates. The direct substrate recognition relies on a large family of F-box-domain-containing subunits. One of these substrate receptors is FBXO38, which is encoded by a gene found mutated in families with early-onset distal motor neuronopathy. SCFFBXO38 ubiquitin ligase controls the stability of ZXDB, a nuclear factor associated with the centromeric chromatin protein CENP-B. Loss of FBXO38 in mice results in growth retardation and defects in spermatogenesis characterized by deregulation of the Sertoli cell transcription program and compromised centromere integrity. Moreover, it was reported that SCFFBXO38 mediates the degradation of PD-1, a key immune-checkpoint inhibitor in T cells. Here, we have re-addressed the link between SCFFBXO38 and PD-1 proteolysis. Our data do not support the notion that SCFFBXO38 directly or indirectly controls the abundance and stability of PD-1 in T cells.


Subject(s)
F-Box Proteins , Programmed Cell Death 1 Receptor , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Animals , Humans , Mice , F-Box Proteins/metabolism , F-Box Proteins/genetics , T-Lymphocytes/metabolism , Proteolysis , Ubiquitination , SKP Cullin F-Box Protein Ligases/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Male
9.
J Med Virol ; 96(9): e29891, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39223933

ABSTRACT

The ubiquitin-proteasome system is frequently employed to degrade viral proteins, thereby inhibiting viral replication and pathogenicity. Through an analysis of the degradation kinetics of all the SARS-CoV-2 proteins, our study revealed rapid degradation of several proteins, particularly NSP5. Additionally, we identified FBXO22, an E3 ubiquitin ligase, as the primary regulator of NSP5 ubiquitination. Moreover, we validated the interaction between FBXO22 and NSP5, demonstrating that FBXO22-mediated ubiquitination of NSP5 facilitated its recognition by the proteasome, leading to subsequent degradation. Specifically, FBXO22 catalyzed the formation of K48-linked polyubiquitin chains on NSP5 at lysine residues 5 and 90. Knockdown of FBXO22 resulted in decreased NSP5 ubiquitination levels, increased stability, and enhanced ability to evade the host innate immune response. Notably, the protein level of FBXO22 were negatively correlated with SARS-CoV-2 load, highlighting its importance in inhibiting viral replication. This study elucidates the molecular mechanism by which FBXO22 mediates the degradation of NSP5 and underscores its critical role in limiting viral replication. The identification of FBXO22 as a regulator of NSP5 stability provides new insights and potential avenues for targeting NSP5 in antiviral strategies.


Subject(s)
Proteasome Endopeptidase Complex , SARS-CoV-2 , Ubiquitination , Virus Replication , Humans , Proteasome Endopeptidase Complex/metabolism , SARS-CoV-2/physiology , SARS-CoV-2/metabolism , COVID-19/virology , COVID-19/metabolism , F-Box Proteins/metabolism , F-Box Proteins/genetics , HEK293 Cells , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Proteolysis , Coronavirus Papain-Like Proteases/metabolism , Receptors, Cytoplasmic and Nuclear
10.
Commun Biol ; 7(1): 1099, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39244596

ABSTRACT

Centromere pairing is crucial for synapsis in meiosis. This study delves into the Skp1-Cullin1-F-box protein (SCF) E3 ubiquitin ligase complex, specifically focusing on F-box protein 47 (FBXO47), in mouse meiosis. Here, we revealed that FBXO47 is localized at the centromere and it regulates centromere pairing cooperatively with SKP1 to ensure proper synapsis in pachynema. The absence of FBXO47 causes defective centromeres, resulting in incomplete centromere pairing, which leads to corruption of SC at centromeric ends and along chromosome axes, triggering premature dissociation of chromosomes and pachytene arrest. FBXO47 deficient pachytene spermatocytes exhibited drastically reduced SKP1 expression at centromeres and chromosomes. Additionally, FBXO47 stabilizes SKP1 by down-regulating its ubiquitination in HEK293T cells. In essence, we propose that FBXO47 collaborates with SKP1 to facilitate centromeric SCF formation in spermatocytes. In summary, we posit that the centromeric SCF E3 ligase complex regulates centromere pairing for pachynema progression in mice.


Subject(s)
Centromere , Chromosome Pairing , F-Box Proteins , Spermatocytes , Animals , Male , Centromere/metabolism , Centromere/genetics , Mice , Spermatocytes/metabolism , F-Box Proteins/metabolism , F-Box Proteins/genetics , Humans , HEK293 Cells , SKP Cullin F-Box Protein Ligases/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Meiosis , Mice, Knockout , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Mice, Inbred C57BL
11.
Front Biosci (Landmark Ed) ; 29(8): 296, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39206900

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is a malignant form of lung cancer, and its prognosis could be improved by identifying key therapeutic targets. Thus, this study investigates the potential role of F-box Only Protein 33 (FBXO33) in NSCLC. METHODS: The expression levels of FBXO33 in NSCLC were determined using University of Alabama at Birmingham Cancer Data Analysis Portal (UALCAN) prediction, and its correlation with overall survival (OS) was analyzed via Kaplan-Meier survival analysis. These results were validated through quantitative polymerase chain reaction (qPCR), western blot (WB), and immunofluorescence (IF). We modulated FBXO33 expression by overexpression or knockdown and analyzed its effects on cell growth, proliferation, migration, invasion, and stemness characteristics in NSCLC cell lines. Additionally, the interaction between FBXO33 and Myelocytomatosis (Myc) and its impact on Myc ubiquitination were examined. An in vivo NSCLC xenograft model was used to corroborate the in vivo experimental results. RESULTS: The study found an inverse correlation between FBXO33 expression in NSCLC and OS. Lower FBXO33 expression enhanced the growth, proliferation, migration, invasion, and stemness characteristics of NSCLC cell lines. FBXO33 interacted with Myc to promote its ubiquitination and subsequent degradation, which suppressed NSCLC development. CONCLUSION: FBXO33 is expressed at low levels in NSCLC and correlates with lower OS. Overexpression of FBXO33 promotes Myc ubiquitination and degradation and inhibits tumor cell proliferation, migration and stemness characteristics, thereby impeding NSCLC progression.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Proliferation , F-Box Proteins , Lung Neoplasms , Neoplastic Stem Cells , Proto-Oncogene Proteins c-myc , Ubiquitination , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , F-Box Proteins/metabolism , F-Box Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Cell Proliferation/genetics , Cell Movement/genetics , Mice, Nude , Mice , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Gene Expression Regulation, Neoplastic , Female , Proteolysis , Male
12.
Mol Cell ; 84(16): 3115-3127.e11, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39116872

ABSTRACT

Proteasome is essential for cell survival, and proteasome inhibition induces proteasomal gene transcription via the activated endoplasmic-reticulum-associated transcription factor nuclear factor erythroid 2-like 1 (Nrf1/NFE2L1). Nrf1 activation requires proteolytic cleavage by DDI2 and N-glycan removal by NGLY1. We previously showed that Nrf1 ubiquitination by SKP1-CUL1-F-box (SCF)FBS2/FBXO6, an N-glycan-recognizing E3 ubiquitin ligase, impairs its activation, although the molecular mechanism remained elusive. Here, we show that SCFFBS2 cooperates with the RING-between-RING (RBR)-type E3 ligase ARIH1 to ubiquitinate Nrf1 through oxyester bonds in human cells. Endo-ß-N-acetylglucosaminidase (ENGASE) generates asparagine-linked N-acetyl glucosamine (N-GlcNAc) residues from N-glycans, and N-GlcNAc residues on Nrf1 served as acceptor sites for SCFFBS2-ARIH1-mediated ubiquitination. We reconstituted the polyubiquitination of N-GlcNAc and serine/threonine residues on glycopeptides and found that the RBR-specific E2 enzyme UBE2L3 is required for the assembly of atypical ubiquitin chains on Nrf1. The atypical ubiquitin chains inhibited DDI2-mediated activation. The present results identify an unconventional ubiquitination pathway that inhibits Nrf1 activation.


Subject(s)
Nuclear Respiratory Factor 1 , Ubiquitination , Humans , HEK293 Cells , Nuclear Respiratory Factor 1/metabolism , Nuclear Respiratory Factor 1/genetics , NF-E2-Related Factor 1/metabolism , NF-E2-Related Factor 1/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Acetylglucosamine/metabolism , HeLa Cells , Proteasome Endopeptidase Complex/metabolism , F-Box Proteins/metabolism , F-Box Proteins/genetics
13.
Gene ; 930: 148835, 2024 Dec 20.
Article in English | MEDLINE | ID: mdl-39127414

ABSTRACT

BACKGROUND: Gliomas are the most common primary malignant tumours of the central nervous system, and neddylation may be a potential target for the treatment of gliomas. Our study analysed neddylation's potential role in gliomas of different pathological types and its correlation with immunotherapy. METHODS: Genes required for model construction were sourced from existing literature, and their expression data were extracted from the TCGA and CGGA databases. LASSO regression was employed to identify genes associated with the prognosis of glioma patients in TCGA and to establish a clinical prognostic model. Biological changes in glioma cell lines following intervention with hub genes were evaluated using the CCK-8 assay and transwell assay. The genes implicated in the model construction were validated across various cell lines using Western blot. We conducted analyses to examine correlations between model scores and clinical data, tumor microenvironments, and immune checkpoints. Furthermore, we investigated potential differences in molecular functions and mechanisms among different groups. RESULTS: We identified 249 genes from the Reactome database and analysed their expression profiles in the TCGA and CGGA databases. After using LASSO-Cox, four genes (BRCA1, BIRC5, FBXL16 and KLHL25, p < 0.05) with significant correlations were identified. We selected FBXL16 for validation in in vitro experiments. Following FBXL16 overexpression, the proliferation, migration, and invasion abilities of glioma cell lines all showed a decrease. Then, we constructed the NEDD Index for gliomas. The nomogram indicated that this model could serve as an independent prognostic marker. Analysis of the tumour microenvironment and immune checkpoints revealed that the NEDD index was also correlated with immune cell infiltration and the expression levels of various immune checkpoints. CONCLUSION: The NEDD index can serve as a practical tool for predicting the prognosis of glioma patients, and it is correlated with immune cell infiltration and the expression levels of immune checkpoints.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Gene Expression Regulation, Neoplastic , Glioma , Humans , Glioma/genetics , Glioma/immunology , Glioma/pathology , Prognosis , Cell Line, Tumor , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , NEDD8 Protein/genetics , NEDD8 Protein/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Cell Proliferation/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , Female , Gene Expression Profiling/methods , Databases, Genetic , Cell Movement/genetics , Male
14.
Am J Physiol Heart Circ Physiol ; 327(4): H937-H946, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39150394

ABSTRACT

Influenza A virus (IAV) infection while primarily affecting the lungs, is often associated with cardiovascular complications. However, the mechanisms underlying this association are not fully understood. Here, we investigated the potential role of FBXL19, a member of the Skp1-Cullin-1-F-box family of E3 ubiquitin ligase, in IAV-induced cardiac inflammation. We demonstrated that FBXL19 overexpression in endothelial cells (ECs) reduced viral titers and IAV matrix protein 1 (M1) levels while increasing antiviral gene expression, including interferon (IFN)-α, -ß, and -γ and RANTES (regulated on activation normal T cell expressed and secreted) in the cardiac tissue of IAV-infected mice. Moreover, EC-specific overexpression of FBXL19 attenuated the IAV infection-reduced interferon regulatory factor 3 (IRF3) level without altering its mRNA level and suppressed cardiac inflammation. Furthermore, IAV infection triggered cellular senescence programs in the heart as indicated by the upregulation of p16 and p21 mRNA levels and the downregulation of lamin-B1 levels, which were partially reversed by FBXL19 overexpression in ECs. Our findings indicate that EC-specific overexpression of FBXL19 protects against IAV-induced cardiac damage by enhancing interferon-mediated antiviral signaling, reducing cardiac inflammation, and suppressing cellular senescence programs.NEW & NOTEWORTHY Our study reveals a novel facet of IAV infection, demonstrating that it can trigger cellular senescence within the heart. Intriguingly, upregulation of endothelial FBXL19 promotes host innate immunity, reduces cardiac senescence, and diminishes inflammation. These findings highlight the therapeutic potential of targeting FBXL19 to mitigate IAV-induced cardiovascular complications.


Subject(s)
Cellular Senescence , Endothelial Cells , Interferon Regulatory Factor-3 , Orthomyxoviridae Infections , Animals , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Endothelial Cells/metabolism , Endothelial Cells/immunology , Endothelial Cells/virology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Mice, Inbred C57BL , Mice , F-Box Proteins/metabolism , F-Box Proteins/genetics , Humans , Influenza A virus/pathogenicity , Myocardium/metabolism , Myocardium/immunology , Myocardium/pathology , Disease Models, Animal , Signal Transduction , Interferons/metabolism , Interferons/genetics , Male , Chemokine CCL5
15.
Elife ; 132024 Aug 20.
Article in English | MEDLINE | ID: mdl-39163107

ABSTRACT

Ribonucleoprotein (RNP) granules are membraneless electron-dense structures rich in RNAs and proteins, and involved in various cellular processes. Two RNP granules in male germ cells, intermitochondrial cement and the chromatoid body (CB), are associated with PIWI-interacting RNAs (piRNAs) and are required for transposon silencing and spermatogenesis. Other RNP granules in male germ cells, the reticulated body and CB remnants, are also essential for spermiogenesis. In this study, we disrupted FBXO24, a testis-enriched F-box protein, in mice and found numerous membraneless electron-dense granules accumulated in sperm flagella. Fbxo24 knockout (KO) mice exhibited malformed flagellar structures, impaired sperm motility, and male infertility, likely due to the accumulation of abnormal granules. The amount and localization of known RNP granule-related proteins were not disrupted in Fbxo24 KO mice, suggesting that the accumulated granules were distinct from known RNP granules. Further studies revealed that RNAs and two importins, IPO5 and KPNB1, abnormally accumulated in Fbxo24 KO spermatozoa and that FBXO24 could ubiquitinate IPO5. In addition, IPO5 and KPNB1 were recruited to stress granules, RNP complexes, when cells were treated with oxidative stress or a proteasome inhibitor. These results suggest that FBXO24 is involved in the degradation of IPO5, disruption of which may lead to the accumulation of abnormal RNP granules in sperm flagella.


Subject(s)
F-Box Proteins , Infertility, Male , Mice, Knockout , Sperm Tail , Male , Animals , Infertility, Male/genetics , Infertility, Male/metabolism , Mice , Sperm Tail/metabolism , F-Box Proteins/metabolism , F-Box Proteins/genetics , Cytoplasmic Granules/metabolism , Spermatozoa/metabolism
16.
Proc Natl Acad Sci U S A ; 121(35): e2408322121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39163340

ABSTRACT

The circadian clock is an endogenous oscillator, and its importance lies in its ability to impart rhythmicity on downstream biological processes, or outputs. Our knowledge of output regulation, however, is often limited to an understanding of transcriptional connections between the clock and outputs. For instance, the clock is linked to plant growth through the gating of photoreceptors via rhythmic transcription of the nodal growth regulators, PHYTOCHROME-INTERACTING FACTORs (PIFs), but the clock's role in PIF protein stability is less clear. Here, we identified a clock-regulated, F-box type E3 ubiquitin ligase, CLOCK-REGULATED F-BOX WITH A LONG HYPOCOTYL 1 (CFH1), that specifically interacts with and degrades PIF3 during the daytime. Additionally, genetic evidence indicates that CFH1 functions primarily in monochromatic red light, yet CFH1 confers PIF3 degradation independent of the prominent red-light photoreceptor phytochrome B (phyB). This work reveals a clock-mediated growth regulation mechanism in which circadian expression of CFH1 promotes sustained, daytime PIF3 degradation in parallel with phyB signaling.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Basic Helix-Loop-Helix Transcription Factors , Circadian Clocks , Phytochrome B , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Arabidopsis/metabolism , Arabidopsis/genetics , Circadian Clocks/physiology , Circadian Clocks/genetics , Phytochrome B/metabolism , Phytochrome B/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Plant , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Circadian Rhythm/physiology , F-Box Proteins/metabolism , F-Box Proteins/genetics , Light
17.
FEBS Lett ; 598(18): 2259-2268, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39171510

ABSTRACT

The cytosolic peptide:N-glycanase (PNGase) is involved in the quality control of N-glycoproteins via the endoplasmic reticulum-associated degradation (ERAD) pathway. Mutations in the gene encoding cytosolic PNGase (NGLY1 in humans) cause NGLY1 deficiency. Recent findings indicate that the F-box protein FBS2 of the SCFFBS2 ubiquitin ligase complex can be a promising drug target for NGLY1 deficiency. Here, we determined the crystal structure of bovine FBS2 complexed with the adaptor protein SKP1 and a sugar ligand, Man3GlcNAc2, which corresponds to the core pentasaccharide of N-glycan. Our crystallographic data together with NMR data revealed the structural basis of disparate sugar-binding specificities in homologous FBS proteins and identified a potential druggable pocket for in silico docking studies. Our results provide a potential basis for the development of selective inhibitors against FBS2 in NGLY1 deficiency.


Subject(s)
Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase , Animals , Cattle , Humans , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , F-Box Proteins/metabolism , F-Box Proteins/chemistry , F-Box Proteins/genetics , Models, Molecular , Molecular Docking Simulation , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/genetics , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/chemistry , Protein Binding
18.
J Cell Mol Med ; 28(16): e70021, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39153212

ABSTRACT

Accumulating evidence has demonstrated that F-box protein 22 (FBXO22) participates in tumour development and progression in various types of human malignancies. However, the functions and detailed molecular mechanisms of FBXO22 in osteosarcoma tumorigenesis and progression remain elusive. In this study, we aimed to determine the effects of FBXO22 on the cell proliferation, migration and invasion of osteosarcoma cells using cell counting kit-8 and Matrigel Transwell approaches. Moreover, we explored the molecular mechanisms by which FBXO22 mediated oncogenesis and progression in osteosarcoma via Western blotting, immunoprecipitation and ubiquitination. We found that FBXO22 depletion inhibited the proliferation, migration and invasion of osteosarcoma cells, whereas FBXO22 overexpression increased the proliferation and motility of osteosarcoma cells. Mechanistically, FBXO22 promoted the ubiquitination and degradation of FoxO1 in osteosarcoma cells. FBXO22 depletion reduced cell proliferation and motility via regulation of FoxO1. Taken together, our findings provide new insight into FBXO22-induced osteosarcoma tumorigenesis. The inhibition of FBXO22 could be a promising strategy for the treatment of osteosarcoma.


Subject(s)
Cell Movement , Cell Proliferation , F-Box Proteins , Forkhead Box Protein O1 , Gene Expression Regulation, Neoplastic , Osteosarcoma , Ubiquitination , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/genetics , Humans , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , F-Box Proteins/metabolism , F-Box Proteins/genetics , Cell Movement/genetics , Cell Line, Tumor , Proteolysis , Disease Progression , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Neoplasm Invasiveness , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Receptors, Cytoplasmic and Nuclear
19.
Sci Rep ; 14(1): 18697, 2024 08 12.
Article in English | MEDLINE | ID: mdl-39134694

ABSTRACT

Neuroblastoma, the deadliest solid tumor in children, exhibits alarming mortality rates, particularly among high-risk cases. To enhance survival rates, a more precise risk stratification for patients is imperative. Utilizing proteomic data from 34 cases with or without N-Myc amplification, we identified 28 differentially expressed ubiquitination-related proteins (URGs). From these, a prognostic signature comprising 6 URGs was constructed. A nomogram incorporating clinical-pathological parameters yielded impressive AUC values of 0.88, 0.93, and 0.95 at 1, 3, and 5 years, respectively. Functional experiments targeting the E3 ubiquitin ligase FBXO42, a component of the prognostic signature, revealed its TP53-dependent promotion of neuroblastoma cell proliferation. In conclusion, our ubiquitination-related prognostic model robustly predicts patient outcomes, guiding clinical decisions. Additionally, the newfound pro-proliferative role of FBXO42 offers a novel foundation for understanding the molecular mechanisms of neuroblastoma.


Subject(s)
Cell Proliferation , Neuroblastoma , Humans , Neuroblastoma/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Prognosis , Male , Ubiquitination , Cell Line, Tumor , Female , Proteomics/methods , Gene Expression Regulation, Neoplastic , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , F-Box Proteins/metabolism , F-Box Proteins/genetics , Child, Preschool , Infant , Child
20.
PLoS Biol ; 22(8): e3002739, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39137238

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) poses a significant threat due to its tendency to evade early detection, frequent metastasis, and the subsequent challenges in devising effective treatments. Processes that govern epithelial-mesenchymal transition (EMT) in PDAC hold promise for advancing novel therapeutic strategies. SAMD1 (SAM domain-containing protein 1) is a CpG island-binding protein that plays a pivotal role in the repression of its target genes. Here, we revealed that SAMD1 acts as a repressor of genes associated with EMT. Upon deletion of SAMD1 in PDAC cells, we observed significantly increased migration rates. SAMD1 exerts its effects by binding to specific genomic targets, including CDH2, encoding N-cadherin, which emerged as a driver of enhanced migration upon SAMD1 knockout. Furthermore, we discovered the FBXO11-containing E3 ubiquitin ligase complex as an interactor and negative regulator of SAMD1, which inhibits SAMD1 chromatin-binding genome-wide. High FBXO11 expression in PDAC is associated with poor prognosis and increased expression of EMT-related genes, underlining an antagonistic relationship between SAMD1 and FBXO11. In summary, our findings provide insights into the regulation of EMT-related genes in PDAC, shedding light on the intricate role of SAMD1 and its interplay with FBXO11 in this cancer type.


Subject(s)
Carcinoma, Pancreatic Ductal , Epithelial-Mesenchymal Transition , F-Box Proteins , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms , Receptors, LDL , Animals , Humans , Cadherins/metabolism , Cadherins/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , F-Box Proteins/metabolism , F-Box Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Receptors, LDL/genetics , Receptors, LDL/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL