Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Publication year range
1.
Aging (Albany NY) ; 12(23): 24394-24423, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33260160

ABSTRACT

FBXW7 functions as an E3 ubiquitin ligase to mediate oncoprotein degradation via the ubiquitin-proteasome system in cancer cells, effectively inhibiting the growth and survival of tumor cells. However, little is known about the functions of FBXW7 in macrophages and the tumor immune microenvironment. In this study, we find that FBXW7 suppresses M2-like tumor-associated macrophage (TAM) polarization to limit tumor progression. We identified a significant increase in the proportion of M2-like TAMs and aggravated tumor growth in mice with myeloid FBXW7 deficiency by subcutaneous inoculation with Lewis lung carcinoma cells (LLCs). When stimulated with LLCs supernatant in vitro, FBXW7-knockout macrophages displayed increased M2 macrophage polarization and enhanced ability of supporting cancer cells growth. In mechanism, we confirmed that FBXW7 inhibited M2-like TAM polarization by mediating c-Myc degradation via the ubiquitin-proteasome system. These findings highlight the role of FBXW7 in M2-like TAM polarization and provide new insights into the potential targets for cancer immunotherapies.


Subject(s)
Carcinoma, Lewis Lung/enzymology , F-Box-WD Repeat-Containing Protein 7/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor-Associated Macrophages/enzymology , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Proliferation , Cells, Cultured , F-Box-WD Repeat-Containing Protein 7/deficiency , F-Box-WD Repeat-Containing Protein 7/genetics , Gene Expression Regulation, Neoplastic , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , Tumor Burden , Tumor Microenvironment , Tumor-Associated Macrophages/pathology , Ubiquitination
2.
Neoplasia ; 22(11): 644-658, 2020 11.
Article in English | MEDLINE | ID: mdl-33070870

ABSTRACT

Fbw7 is a tumor suppressor that regulates the degradation of oncogenic substrates such as c-Jun, c-Myc, Notch1 intracellular domain (ICD), and cyclin E by functioning as the substrate recognition protein in the Skp1-Cullin-F-box (SCF) ubiquitin ligase complex. Consequently, low expression or loss of FBXW7 in breast cancer has been hypothesized to result in the accumulation of oncogenic transcription factors that are master regulators of proliferation, apoptosis, and ultimately transformation. Despite this, the direct effect of Fbw7 loss on mammary gland morphology and tumorigenesis has not been examined. Here, we demonstrate that conditional deletion of Fbxw7 in murine mammary tissue initiates breast tumor development and also results in lactation and involution defects. Further, while Fbxw7 loss results in the overexpression of Notch1-ICD, c-Jun, cyclin E, and c-Myc, the downstream transcription factor pathways associated with c-Myc and cyclin E are the most dysregulated, including at the single-cell level. These pathways are dysregulated early after Fbxw7 loss, and their sustained loss results in tumorigenesis and reinforced c-Myc and cyclin E-E2F pathway disruption. We also find that loss of Fbxw7 is linked to the acquisition of Trp53 mutations, similar to the mutational spectrum observed in patients. Our results demonstrate that the loss of Fbxw7 promotes the acquisition of Trp53 mutations and that the two cooperate in breast tumor development. Targeting c-Myc, E2F, or p53 may therefore be a beneficial treatment strategy for FBXW7-altered breast cancer patients.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , E2F Transcription Factors/metabolism , F-Box-WD Repeat-Containing Protein 7/deficiency , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/genetics , Amino Acid Sequence , Animals , Breast Neoplasms , Cell Line, Tumor , Disease Models, Animal , Disease Susceptibility , F-Box-WD Repeat-Containing Protein 7/chemistry , F-Box-WD Repeat-Containing Protein 7/genetics , Female , Gene Expression Regulation, Neoplastic , Immunohistochemistry , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Transcription, Genetic
3.
Mol Oncol ; 12(6): 883-895, 2018 06.
Article in English | MEDLINE | ID: mdl-29633504

ABSTRACT

Gefitinib, an epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI), is an effective treatment for non-small-cell lung cancer (NSCLC) with EGFR activating mutations, but inevitably, the clinical efficacy is impeded by the emergence of acquired resistance. The tumor suppressor gene FBXW7 modulates chemosensitivity in various human cancers. However, its role in EGFR-TKI therapy in NSCLC has not been well studied. Here, we demonstrate that the mice with deficient Fbxw7 have greater susceptibility to urethane-induced lung tumor development. Through analysis of The Cancer Genome Atlas data, we show that deletion of FBXW7 occurs in 30.9% of lung adenocarcinomas and 63.5% of lung squamous cell carcinomas, which significantly leads to decrease in FBXW7 mRNA expression. The reduction in FBXW7 mRNA level is associated with poor overall survival in lung cancer patients. FBXW7 knockdown dramatically promotes epithelial-mesenchymal transition, migration, and invasion in NSCLC cells. Moreover, with silenced FBXW7, EGFR-TKI-sensitive cells become resistant to gefitinib, which is reversed by the mammalian target of rapamycin inhibitor, rapamycin. Furthermore, xenograft mouse model studies show that FBXW7 knockdown enhances tumorigenesis and resistance to gefitinib. Combination of gefitinib with rapamycin treatment suppresses tumor formation of gefitinib-resistant (GR) FBXW7-knockdown cells. In conclusion, our findings suggest that loss of FBXW7 promotes NSCLC progression as well as gefitinib resistance and combination of gefitinib and rapamycin may provide an effective therapy for GR NSCLC.


Subject(s)
Carcinogenesis/pathology , Drug Resistance, Neoplasm/genetics , F-Box-WD Repeat-Containing Protein 7/genetics , Gefitinib/pharmacology , Gene Deletion , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Disease Susceptibility , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , F-Box-WD Repeat-Containing Protein 7/deficiency , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Prognosis , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
4.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 46(2): 111-117, 2017 03 25.
Article in Chinese | MEDLINE | ID: mdl-28752701

ABSTRACT

Objective: To investigate the effects of myeloid-specific deficiency of FBXW7 on lung metastasis of murine B16F10 melanoma and its mechanisms. Methods: Mice carrying the floxed allele of FBXW7 and lysozyme M-Cre were used for generation of mice with myeloid cell-specific deletion of FBXW7. Mouse genotypes were examined by genomic DNA PCR. B16F10 cells in PBS were injected into the tail vein of Lysm-FBXW7f/f and Lysm+FBXW7 f/f mice. After 14 d, the mice were sacrificed, and the lungs were removed and weighed. B16F10 tumor colonies in the lungs were counted. The myeloid cells were analyzed by flow cytometry. Results: Myeloid-specific deficiency of FBXW7 mice were generated successfully, as FBXW7 expressions in peritoneal macrophages and bone marrow-derived macrophages (BMDMs) of Lysm+FBXW7f/f mice were knockdown. Flow cytometry results showed the deletion of FBXW7 in myeloid lineages did not affect the development of myeloid immune cell subsets. Metastasis was reduced in Lysm+FBXW7 f/f mice compared with control mice. The number of tumor colonies was 165±42, 122±12 respectively. The proportion of metastasis-associated macrophages (MAM) in the lungs of Lysm+FBXW7 f/f mice was reduced [(23.15±7.59)% vs (13.13±2.26)%], while the proportion of resident macrophages was increased [(5.426±0.42)% vs (10.42±1.90)%]. The proportion of myeloid-derived suppressor cells in the lung showed no difference between Lysm-FBXW7f/f and Lysm+FBXW7 f/f mice. Conclusion: Myeloid-specific deficiency of FBXW7 can inhibit lung metastasis of B16F10 melanoma in mice, and the mechanism may be associated with regulation of MAM in the metastatic tumor lesions.


Subject(s)
F-Box-WD Repeat-Containing Protein 7 , Lung Neoplasms/secondary , Macrophages , Melanoma , Animals , F-Box-WD Repeat-Containing Protein 7/deficiency , F-Box-WD Repeat-Containing Protein 7/genetics , Gene Deletion , Gene Expression Regulation, Neoplastic , Lung Neoplasms/enzymology , Macrophages/enzymology , Melanoma/enzymology , Melanoma/genetics , Mice , Myeloid Cells/enzymology
5.
Nat Commun ; 8: 14654, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28287082

ABSTRACT

Viruses can escape from host recognition by degradation of RIG-I or interference with the RIG-I signalling to establish persistent infections. However, the mechanisms by which host cells stabilize RIG-I protein for avoiding its degradation are largely unknown. We report here that, upon virus infection, the E3 ubiquitin ligase FBXW7 translocates from the nucleus into the cytoplasm and stabilizes RIG-I. FBXW7 interacts with SHP2 and mediates the degradation and ubiquitination of SHP2, thus disrupting the SHP2/c-Cbl complex, which mediates RIG-I degradation. When infected with VSV or influenza A virus, FBXW7 conditional knockout mice (Lysm+FBXW7f/f) show impaired antiviral immunity. FBXW7-deficient macrophages have decreased RIG-I protein levels and type-I interferon signalling. Furthermore, PBMCs from RSV-infected children have reduced FBXW7 mRNA levels. Our results identify FBXW7 as an important interacting partner for RIG-I. These findings provide insights into the function of FBXW7 in antiviral immunity and its related clinical significance.


Subject(s)
DEAD Box Protein 58/genetics , F-Box-WD Repeat-Containing Protein 7/genetics , Host-Pathogen Interactions , Influenza A virus/immunology , Macrophages/immunology , Respiratory Syncytial Viruses/immunology , Vesiculovirus/immunology , Active Transport, Cell Nucleus , Animals , Child , DEAD Box Protein 58/immunology , F-Box-WD Repeat-Containing Protein 7/deficiency , F-Box-WD Repeat-Containing Protein 7/immunology , Gene Expression Regulation , HEK293 Cells , Humans , Influenza A virus/pathogenicity , Interferon Type I/genetics , Interferon Type I/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Protein Stability , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Proteolysis , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/immunology , RAW 264.7 Cells , Respiratory Syncytial Viruses/pathogenicity , Ubiquitination , Vesiculovirus/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...