Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell Rep Med ; 5(5): 101533, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38744278

ABSTRACT

Brain metastases (BrMs) are the leading cause of death in patients with solid cancers. BrMs exhibit a highly immunosuppressive milieu and poor response to immunotherapies; however, the underlying mechanism remains largely unclear. Here, we show that upregulation of HSP47 in tumor cells drives metastatic colonization and outgrowth in the brain by creating an immunosuppressive microenvironment. HSP47-mediated collagen deposition in the metastatic niche promotes microglial polarization to the M2 phenotype via the α2ß1 integrin/nuclear factor κB pathway, which upregulates the anti-inflammatory cytokines and represses CD8+ T cell anti-tumor responses. Depletion of microglia reverses HSP47-induced inactivation of CD8+ T cells and abolishes BrM. Col003, an inhibitor disrupting HSP47-collagen association restores an anti-tumor immunity and enhances the efficacy of anti-PD-L1 immunotherapy in BrM-bearing mice. Our study supports that HSP47 is a critical determinant of M2 microglial polarization and immunosuppression and that blocking the HSP47-collagen axis represents a promising therapeutic strategy against brain metastatic tumors.


Subject(s)
Brain Neoplasms , CD8-Positive T-Lymphocytes , Collagen , HSP47 Heat-Shock Proteins , Microglia , Animals , Microglia/metabolism , Microglia/drug effects , Microglia/immunology , Brain Neoplasms/secondary , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Collagen/metabolism , Mice , HSP47 Heat-Shock Proteins/metabolism , HSP47 Heat-Shock Proteins/genetics , Cell Line, Tumor , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Tumor Microenvironment/immunology , Mice, Inbred C57BL , Cell Polarity/drug effects , Female , NF-kappa B/metabolism
2.
Adv Sci (Weinh) ; 10(33): e2303561, 2023 11.
Article in English | MEDLINE | ID: mdl-37822160

ABSTRACT

Mesenchymal glioblastoma (GBM) is highly resistant to radio-and chemotherapy and correlates with worse survival outcomes in GBM patients; however, the underlying mechanism determining the mesenchymal phenotype remains largely unclear. Herein, it is revealed that FBXO7, a substrate-recognition component of the SCF complex implicated in the pathogenesis of Parkinson's disease, confers mesenchymal properties and chemoresistance in GBM by controlling Rbfox2-mediated alternative splicing. Specifically, FBXO7 ubiquitinates Rbfox2 Lys249 through K63-linked ubiquitin chains upon arginine dimethylation at Arg341 and Arg441 by PRMT5, leading to Rbfox2 stabilization. FBXO7 controls Rbfox2-mediated splicing of mesenchymal genes, including FoxM1, Mta1, and Postn. FBXO7-induced exon Va inclusion of FoxM1 promotes FoxM1 phosphorylation by MEK1 and nuclear translocation, thereby upregulates CD44, CD9, and ID1 levels, resulting in GBM stem cell self-renewal and mesenchymal transformation. Moreover, FBXO7 is stabilized by temozolomide, and FBXO7 depletion sensitizes tumor xenografts in mice to chemotherapy. The findings demonstrate that the FBXO7-Rbfox2 axis-mediated splicing contributes to mesenchymal transformation and tumorigenesis, and targeting FBXO7 represents a potential strategy for GBM treatment.


Subject(s)
F-Box Proteins , Glioblastoma , Animals , Humans , Mice , Alternative Splicing/genetics , Drug Resistance, Neoplasm/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , RNA Splicing , RNA Splicing Factors/genetics , Trans-Activators/genetics
3.
Proc Natl Acad Sci U S A ; 120(32): e2303400120, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37523556

ABSTRACT

Amplification of chromosome 7p11 (7p11) is the most common alteration in primary glioblastoma (GBM), resulting in gains of epidermal growth factor receptor (EGFR) copy number in 50 to 60% of GBM tumors. However, treatment strategies targeting EGFR have thus far failed in clinical trials, and the underlying mechanism remains largely unclear. We here demonstrate that EGFR amplification at the 7p11 locus frequently encompasses its neighboring genes and identifies SEC61G as a critical regulator facilitating GBM immune evasion and tumor growth. We found that SEC61G is always coamplified with EGFR and is highly expressed in GBM. As an essential subunit of the SEC61 translocon complex, SEC61G promotes translocation of newly translated immune checkpoint ligands (ICLs, including PD-L1, PVR, and PD-L2) into the endoplasmic reticulum and promotes their glycosylation, stabilization, and membrane presentation. Depletion of SEC61G promotes the infiltration and cytolytic activity of CD8+ T cells and thus inhibits GBM occurrence. Further, SEC61G inhibition augments the therapeutic efficiency of EGFR tyrosine kinase inhibitors in mice. Our study demonstrates a critical role of SEC61G in GBM immune evasion, which provides a compelling rationale for combination therapy of EGFR-amplified GBMs.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Mice , Glioblastoma/pathology , CD8-Positive T-Lymphocytes/metabolism , ErbB Receptors/metabolism , Cell Line, Tumor , Brain Neoplasms/pathology
4.
Radiat Oncol ; 18(1): 50, 2023 Mar 11.
Article in English | MEDLINE | ID: mdl-36906568

ABSTRACT

BACKGROUND: During the last decades, radiotherapy (RT) for non-small cell lung cancer (NSCLC) with brain metastases (BM) has been developed. However, the lack of predictive biomarkers for therapeutic responses has limited the precision treatment in NSCLC-BM. PATIENTS AND METHODS: In order to find the predictive biomarkers for RT, we investigated the influence of RT on the cell-free DNA (cfDNA) from cerebrospinal fluid (CSF) and the frequency of T cell subsets of NSCLC patients with BM. A total of 19 patients diagnosed as NSCLC with BM were enrolled. The CSF from 19 patients and matched plasma samples from 11 patients were collected before RT, during RT, and after RT. The cfDNA from CSF and plasma were extracted, and the cerebrospinal fluid tumor mutation burden (cTMB) was calculated after through next-generation sequencing. The frequency of T cell subsets in peripheral blood was using flow cytometry. RESULTS: The detection rate of cfDNA was higher in CSF compared to plasma in the matched samples. The mutation abundance of cfDNA in CSF was decreased after RT. However, no significant difference was observed in cTMB before and after RT. Although the median intracranial progression-free survival (iPFS) has not yet been reached in patients with decreased or undetectable cTMB, there was a trend that these patients possessed longer iPFS compared to those with stable or increased cTMB (HR 0.28, 95% CI 0.07-1.18, P = 0.067). The proportion of CD4+T cells in peripheral blood was decreased after RT. CONCLUSION: Our study indicates that cTMB can serve as a prognostic biomarker in NSCLC patients with BMs.


Subject(s)
Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Cell-Free Nucleic Acids , Lung Neoplasms , Humans , Biomarkers, Tumor/cerebrospinal fluid , Brain Neoplasms/diagnosis , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/pathology , Cell-Free Nucleic Acids/cerebrospinal fluid , Lung Neoplasms/pathology , Mutation , Prognosis
5.
Cell Death Discov ; 8(1): 445, 2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36333288

ABSTRACT

Overproduction of reactive oxygen species (ROS) and aberrant lipid metabolism are established hallmarks of cancer; however, the role of ROS in lipid synthesis during tumorigenesis is almost unknown. Herein, we show that ROS regulates lipid synthesis and thus controls colorectal tumorigenesis through a p53-dependent mechanism. In p53 wild-type colorectal cancer (CRC) cells, hydrogen peroxide (H2O2)-induced p53 expression represses the transcription of deubiquitinase USP22, which otherwise deubiquitinates and stabilizes Fatty Acid Synthase (FASN), and thus inhibits fatty acid synthesis. Whereas, in p53-deficient CRC cells, ROS-mediated inhibition of USP22 is relieved, leading to FASN stabilization, which thus promotes lipid synthesis and tumor growth. In human CRC specimens, USP22 expression is positively correlated with FASN expression. Our study demonstrates that ROS critically regulates lipid synthesis and tumorigenesis through the USP22-FASN axis in a p53-dependent manner, and targeting the USP22-FASN axis may represent a potential strategy for the treatment of colorectal cancer.

6.
Sci Adv ; 8(40): eabn2571, 2022 10 07.
Article in English | MEDLINE | ID: mdl-36197973

ABSTRACT

Histone 2A (H2A) monoubiquitination is a fundamental epigenetics mechanism of gene expression, which plays a critical role in regulating cell fate. However, it is unknown if H2A ubiquitination is involved in EGFR-driven tumorigenesis. In the current study, we have characterized a previously unidentified oncogenic lncRNA (lncEPAT) that mediates the integration of the dysregulated EGFR pathway with H2A deubiquitination in tumorigenesis. LncEPAT was induced by the EGFR pathway, and high-level lncEPAT expression positively correlated with the glioma grade and predicted poor survival of glioma patients. Mass spectrometry analyses revealed that lncEPAT specifically interacted with deubiquitinase USP16. LncEPAT inhibited USP16's recruitment to chromatin, thereby blocking USP16-mediated H2A deubiquitination and repressing target gene expression, including CDKN1A and CLUSTERIN. Depletion of lncEPAT promoted USP16-induced cell cycle arrest and cellular senescence, and then repressed GBM cell tumorigenesis. Thus, the EGFR-lncEPAT-ubH2A coupling represents a previously unidentified mechanism for epigenetic gene regulation and senescence resistance during GBM tumorigenesis.


Subject(s)
Glioblastoma , RNA, Long Noncoding , Carcinogenesis/genetics , Chromatin , Clusterin/metabolism , ErbB Receptors/genetics , Glioblastoma/genetics , Histones/metabolism , Humans , Ubiquitin Thiolesterase/genetics
7.
Front Immunol ; 13: 961933, 2022.
Article in English | MEDLINE | ID: mdl-35990696

ABSTRACT

Background: Pyroptosis is a critical type of programmed cell death that is strongly associated with the regulation of tumor and immune cell functions. However, the role of pyroptosis in tumor progression and remodeling of the tumor microenvironment in gliomas has not been extensively studied. Thus, in this study, we aimed to establish a comprehensive pyroptosis-related signature and uncover its potential clinical application in gliomas. Methods: The TCGA glioma cohort was obtained and divided into training and internal validation cohorts, while the CGGA glioma cohort was used as an external validation cohort. Unsupervised consensus clustering was performed to identify pyroptosis-related expression patterns. A Cox regression analysis was performed to establish a pyroptosis-related risk signature. Real-time quantitative PCR was performed to analyze the expression of signature genes in glioma tissues. Immune infiltration was analyzed and validated by immunohistochemical staining. The expression patterns of signature genes in different cell types were analyzed using single-cell RNA sequencing data. Finally, therapeutic responses to chemotherapy, immunotherapy, and potential small-molecule inhibitors were investigated. Results: Patients with glioma were stratified into clusters 1 and 2 based on the expression patterns of pyroptosis-related genes. Cluster 2 showed a longer overall (P<0.001) and progression-free survival time (P<0.001) than Cluster 1. CD8+ T cell enrichment was observed in Cluster 1. A pyroptosis-related risk signature (PRRS) was then established. The high PRRS group showed a significantly poorer prognosis than the low PRRS group in the training cohort (P<0.001), with validation in the internal and external validation cohorts. Immunohistochemical staining demonstrated that CD8+ T cells were enriched in high PRRS glioma tissues. PRRS genes also showed cell-specific expression in tumor and immune cells. Moreover, the high PRRS risk group showed higher temozolomide sensitivity and increased response to anti-PD1 treatment in a glioblastoma immunotherapy cohort. Finally, Bcl-2 inhibitors were screened as candidates for adjunct immunotherapy of gliomas. Conclusion: The pyroptosis-related signature established in this study can be used to reliably predict clinical outcomes and immunotherapy responses in glioma patients. The correlation between the pyroptosis signature and the tumor immune microenvironment may be used to further guide the sensitization of glioma patients to immunotherapy.


Subject(s)
Brain Neoplasms , Glioma , Porcine Reproductive and Respiratory Syndrome , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Glioma/genetics , Glioma/metabolism , Glioma/therapy , Humans , Immunotherapy , Prognosis , Pyroptosis/genetics , Swine , Tumor Microenvironment/genetics
8.
Sci Adv ; 7(8)2021 02.
Article in English | MEDLINE | ID: mdl-33597240

ABSTRACT

Translation is a crucial process in cancer development and progression. Many oncogenic signaling pathways target the translation initiation stage to satisfy the increased anabolic demands of cancer cells. Using quantitative profiling of initiating ribosomes, we found that ribosomal pausing at the start codon serves as a "brake" to restrain the translational output. In response to oncogenic RAS signaling, the initiation pausing relaxes and contributes to the increased translational flux. Intriguingly, messenger RNA (mRNA) m6A modification in the vicinity of start codons influences the behavior of initiating ribosomes. Under oncogenic RAS signaling, the reduced mRNA methylation leads to relaxed initiation pausing, thereby promoting malignant transformation and tumor growth. Restored initiation pausing by inhibiting m6A demethylases suppresses RAS-mediated oncogenic translation and subsequent tumorigenesis. Our findings unveil a paradigm of translational control that is co-opted by RAS mutant cancer cells to drive malignant phenotypes.


Subject(s)
Carcinogenesis , Ribosomes , Carcinogenesis/genetics , Carcinogenesis/metabolism , Codon, Initiator/metabolism , Humans , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism
9.
ACS Omega ; 5(40): 25810-25818, 2020 Oct 13.
Article in English | MEDLINE | ID: mdl-33073105

ABSTRACT

Kinetics and thermodynamics of esterification by a cation-exchange resin in an intensified fixed-bed reactor was studied systematically. The resin type, catalyst loading, volume flow rate, initial molar ratio, temperature, and catalyst reusability were studied and optimized. The nonideality of the reaction system was corrected by the UNIFAC group contribution method. The Δr H 0, Δr S 0, and Δr G 0 of the reaction were acquired by two methods. The pseudo-homogeneous (PH) model and the Langmuir-Hinshelwood-Hougen-Watson (LHHW) model were adopted to simulate the kinetic process. The result indicated that the LHHW model was better suited to simulate the kinetic process than the PH model.

10.
Cancer Res ; 80(5): 1049-1063, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31888886

ABSTRACT

Wnt/ß-catenin signaling activates the transcription of target genes to regulate stem cells and cancer development. However, the contribution of epigenetic regulation to this process is unknown. Here, we report that Wnt activation stabilizes the epigenetic regulator KDM4C that promotes tumorigenesis and survival of human glioblastoma cells by epigenetically activating the transcription of Wnt target genes. KDM4C protein expression was upregulated in human glioblastomas, and its expression directly correlated with Wnt activity and Wnt target gene expression. KDM4C was essential for Wnt-induced gene expression and tumorigenesis of glioblastoma cells. In the absence of Wnt3a, protein kinase R phosphorylated KDM4C at Ser918, inducing KDM4C ubiquitination and degradation. Wnt3a stabilized KDM4C through inhibition of GSK3-dependent protein kinase R activity. Stabilized KDM4C accumulated in the nucleus and bound to and demethylated TCF4-associated histone H3K9 by interacting with ß-catenin, promoting HP1γ removal and transcriptional activation. These findings reveal that Wnt-KDM4C-ß-catenin signaling represents a novel mechanism for the transcription of Wnt target genes and regulation of tumorigenesis, with important clinical implications. SIGNIFICANCE: These findings identify the Wnt-KDM4C-ß-catenin signaling axis as a critical mechanism for glioma tumorigenesis that may serve as a new therapeutic target in glioblastoma.


Subject(s)
Brain Neoplasms/genetics , Carcinogenesis/genetics , Epigenesis, Genetic , Glioblastoma/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Wnt Proteins/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA Demethylation , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Histones/genetics , Histones/metabolism , Humans , Protein Stability , Transcription Factor 4/genetics , Transcription, Genetic , Ubiquitination/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
11.
ACS Omega ; 4(21): 19462-19468, 2019 Nov 19.
Article in English | MEDLINE | ID: mdl-31763570

ABSTRACT

Esterification kinetics on acetic acid with isopropyl alcohol was studied in an intensified fixed bed reactor at 333-353 K with Amberlyst 36 Wet. The effects of volume flow rate, molar ratio of reactants, catalyst loading, and operating temperature were investigated and optimized. The method of UNIFAC was applied to calculate the activity coefficient of each component for correcting the nonideality of the solution. Reaction enthalpy, entropy, and Gibbs free energy were calculated in different cases. The pseudohomogeneous model, Eley-Rideal model, and Langmuir-Hinshelwood-Hougen-Watson model were used to establish kinetic equations of the reaction conducted in the IFBR. It was proved that the LHHW model can accurately describe the esterification kinetics in the intensified fixed bed reactor.

12.
Cancer Res ; 79(1): 72-85, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30425057

ABSTRACT

Aberrant activation of ß-catenin signaling is a critical driver for tumorigenesis, but the mechanism underlying this activation is not completely understood. In this study, we demonstrate a critical role of ß-catenin signaling in stabilization of enhancer of zeste homolog 2 (EZH2) and control of EZH2-mediated gene repression in oncogenesis. ß-Catenin/TCF4 activated the transcription of the deubiquitinase USP1, which then interacted with and deubiquitinated EZH2 directly. USP1-mediated stabilization of EZH2 promoted its recruitment to the promoters of CDKN1B, RUNX3, and HOXA5, resulting in enhanced enrichment of histone H3K27me3 and repression of target gene expression. In human glioma specimens, expression levels of nuclear ß-catenin, USP1, and EZH2 correlated with one another. Depletion of ß-catenin/USP1/EZH2 repressed glioma cell proliferation in vitro and tumor formation in vivo. Our findings indicate that a ß-catenin-USP1-EZH2 axis orchestrates the interplay between dysregulated ß-catenin signaling and EZH2-mediated gene epigenetic silencing during glioma tumorigenesis. SIGNIFICANCE: These findings identify the ß-catenin-USP1-EZH2 signaling axis as a critical mechanism for glioma tumorigenesis that may serve as a new therapeutic target in glioblastoma.


Subject(s)
Carcinogenesis/pathology , Enhancer of Zeste Homolog 2 Protein/chemistry , Gene Expression Regulation, Neoplastic , Glioma/pathology , Ubiquitin-Specific Proteases/metabolism , beta Catenin/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Glioma/genetics , Glioma/metabolism , Humans , Mice , Mice, Nude , Prognosis , Protein Stability , Survival Rate , Tumor Cells, Cultured , Ubiquitin-Specific Proteases/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics
13.
Curr Pharm Des ; 23(29): 4290-4302, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-28699529

ABSTRACT

Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor in adults with intensive heterogeneity and one of the most lethal human cancers. Protein kinases control diverse cellular processes by coordinating different signaling pathways. Protein kinases are frequently dysregulated in human cancers, which contributes to tumor initiation and development. Thus, protein kinases are a growing drug target class for cancers including glioblastoma. This review focuses on the most important protein kinases and kinase-mediated signaling cascades in glioblastoma, and discusses the functional mechanism of these kinases in glioblastoma tumorigenesis. Moreover, this review has summarized the most recent preclinical and clinical advances of agents targeting protein kinases in the treatment of glioblastoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Humans , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology
14.
EMBO Rep ; 18(8): 1318-1330, 2017 08.
Article in English | MEDLINE | ID: mdl-28623188

ABSTRACT

Aberrant activation of the Hedgehog (Hh) signaling pathway drives the tumorigenesis of multiple cancers. In this study, we screened a panel of deubiquitinases that may regulate the Hh pathway. We find that deubiquitinase USP48 activates Gli-dependent transcription by stabilizing Gli1 protein. Mechanistically, USP48 interacts with Gli1 and cleaves its ubiquitin off directly. In glioblastoma cells, knockdown of USP48 inhibits cell proliferation and the expression of Gli1's downstream targets, which leads to repressed glioblastoma tumorigenesis. Importantly, USP48's effect on cell proliferation and tumorigenesis depends to some extent on Gli1. In addition, we find that the Sonic Hedgehog (SHH) pathway induces USP48 expression through Gli1-mediated transcriptional activation, which forms thus a positive feedback loop to regulate Hh signaling. In human glioblastoma specimens, the expression levels of USP48 and Gli1 proteins are clinically relevant, and high expression of USP48 correlates with glioma malignancy. In summary, our study reveals that the USP48-Gli1 regulatory axis is critical for glioma cell proliferation and glioblastoma tumorigenesis.


Subject(s)
Carcinogenesis , Glioblastoma/metabolism , Ubiquitin-Specific Proteases/metabolism , Zinc Finger Protein GLI1/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/physiopathology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Signal Transduction , Transcription Factors/metabolism , Ubiquitin-Specific Proteases/genetics , Zinc Finger Protein GLI1/genetics
15.
Cancer Cell ; 31(4): 591-606.e6, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28344040

ABSTRACT

The dynamic and reversible N6-methyladenosine (m6A) RNA modification installed and erased by N6-methyltransferases and demethylases regulates gene expression and cell fate. We show that the m6A demethylase ALKBH5 is highly expressed in glioblastoma stem-like cells (GSCs). Silencing ALKBH5 suppresses the proliferation of patient-derived GSCs. Integrated transcriptome and m6A-seq analyses revealed altered expression of certain ALKBH5 target genes, including the transcription factor FOXM1. ALKBH5 demethylates FOXM1 nascent transcripts, leading to enhanced FOXM1 expression. Furthermore, a long non-coding RNA antisense to FOXM1 (FOXM1-AS) promotes the interaction of ALKBH5 with FOXM1 nascent transcripts. Depleting ALKBH5 and FOXM1-AS disrupted GSC tumorigenesis through the FOXM1 axis. Our work uncovers a critical function for ALKBH5 and provides insight into critical roles of m6A methylation in glioblastoma.


Subject(s)
AlkB Homolog 5, RNA Demethylase/metabolism , Brain Neoplasms/pathology , Forkhead Box Protein M1/metabolism , Glioblastoma/pathology , 3' Untranslated Regions , AlkB Homolog 5, RNA Demethylase/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/genetics , Glioblastoma/mortality , Male , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Xenograft Model Antitumor Assays
16.
Nat Cell Biol ; 18(9): 954-966, 2016 09.
Article in English | MEDLINE | ID: mdl-27501329

ABSTRACT

Emerging evidence has shown that GSK3ß plays oncogenic roles in multiple tumour types; however, the underlying mechanisms remain largely unknown. Here, we show that nuclear GSK3ß is responsible for the accumulation of the histone demethylase KDM1A and critically regulates histone H3K4 methylation during tumorigenesis. GSK3ß phosphorylates KDM1A Ser683 upon priming phosphorylation of KDM1A Ser687 by CK1α. Phosphorylation of KDM1A induces its binding with and deubiquitylation by USP22, leading to KDM1A stabilization. GSK3ß- and USP22-dependent KDM1A stabilization is required for the demethylation of histone H3K4, thereby repressing BMP2, CDKN1A and GATA6 transcription, which results in cancer stem cell self-renewal and glioblastoma tumorigenesis. In human glioblastoma specimens, KDM1A levels are correlated with nuclear GSK3ß and USP22 levels. Furthermore, a GSK3 inhibitor, tideglusib, sensitizes tumour xenografts to chemotherapy in mice via KDM1A downregulation and improves survival. Our findings demonstrate that nuclear GSK3ß- and USP22-mediated KDM1A stabilization is essential for glioblastoma tumorigenesis.


Subject(s)
Carcinogenesis/metabolism , Endopeptidases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Histone Demethylases/metabolism , Thiolester Hydrolases/metabolism , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Glioblastoma/metabolism , Histones/metabolism , Humans , Mice, Nude , Phosphorylation/drug effects , Thiadiazoles/pharmacology , Ubiquitin Thiolesterase , Ubiquitination
17.
Oncotarget ; 7(27): 42513-42526, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27285761

ABSTRACT

A hallmark of aberrant activation of the Wnt/ß-catenin signaling pathway has been observed in most colorectal cancers (CRC), but little is known about the role of non-coding RNAs regulated by this pathway. Here, we found that miR-150 was the most significantly upregulated microRNA responsive to elevated of Wnt/ß-catenin signaling activity in both HCT116 and HEK293T cells. Mechanistically, the ß-catenin/LEF1 complex binds to the conserved TCF/LEF1-binding element in the miR-150 promoter and thereby transactivates its expression. Enforced expression of miR-150 in HCT116 cell line transformed cells into a spindle shape with higher migration and invasion activity. miR-150 markedly suppressed the CREB signaling pathway by targeting its core transcription factors CREB1 and EP300. Knockdown of CREB1 or EP300 and knockout of CREB1 by CRISPR/Cas9 phenocopied the epithelial-mesenchymal transition (EMT) observed in HCT116 cells in response to miR-150 overexpression. In summary, our data indicate that miR-150 is a novel Wnt effector that may significantly enhance EMT of CRC cells by targeting the CREB signaling pathway.


Subject(s)
Epithelial-Mesenchymal Transition , MicroRNAs/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cell Movement , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Progression , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Transcriptional Activation , Transfection
18.
Cancer Res ; 76(14): 4293-304, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27246830

ABSTRACT

Malignant glioma is an often fatal type of cancer. Aberrant activation of STAT3 leads to glioma tumorigenesis. STAT3-induced transcription of protein-coding genes has been extensively studied; however, little is known about STAT3-regulated miRNA gene transcription in glioma tumorigenesis. In this study, we found that abnormal activation or decreased expression of STAT3 promotes or inhibits the expression of miR-182-5p, respectively. Bioinformatics analyses determined that tumor suppressor protocadherin-8 (PCDH8) is a candidate target gene of miR-182-5p. miR-182-5p negatively regulated PCDH8 expression by directly targeting its 3'-untranslated region. PCDH8 knockdown induced the proliferative and invasive capacities of glioma cells. Silencing of PCDH8 or miR-182-5p mimics could reverse the inhibitory effect of WP1066, a STAT3 inhibitor, or STAT3 knockdown in vitro and in vivo on glioma progression. Clinically, expression levels of PCDH8 were inversely correlated with those of p-STAT3 or miR-182-5p in glioblastoma tissues. These findings reveal that the STAT3/miR-182-5p/PCDH8 axis has a critical role in glioma tumorigenesis and that targeting the axis may provide a new therapeutic approach for human glioma. Cancer Res; 76(14); 4293-304. ©2016 AACR.


Subject(s)
Brain Neoplasms/etiology , Cadherins/physiology , Glioma/etiology , MicroRNAs/physiology , STAT3 Transcription Factor/physiology , Animals , Brain Neoplasms/pathology , Cadherins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Computational Biology , Glioma/pathology , Humans , Mice , MicroRNAs/analysis , Neoplasm Invasiveness , Protocadherins
19.
EMBO J ; 35(6): 668-84, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26912724

ABSTRACT

A key step of Wnt signaling activation is the recruitment of ß-catenin to the Wnt target-gene promoter in the nucleus, but its mechanisms are largely unknown. Here, we identified FoxM1 as a novel target of Wnt signaling, which is essential for ß-catenin/TCF4 transactivation. GSK3 phosphorylates FoxM1 on serine 474 which induces FoxM1 ubiquitination mediated by FBXW7. Wnt signaling activation inhibits FoxM1 phosphorylation by GSK3-Axin complex and leads to interaction between FoxM1 and deubiquitinating enzyme USP5, thereby deubiquitination and stabilization of FoxM1. FoxM1 accumulation in the nucleus promotes recruitment of ß-catenin to Wnt target-gene promoter and activates the Wnt signaling pathway by protecting the ß-catenin/TCF4 complex from ICAT inhibition. Subsequently, the USP5-FoxM1 axis abolishes the inhibitory effect of ICAT and is required for Wnt-mediated tumor cell proliferation. Therefore, Wnt-induced deubiquitination of FoxM1 represents a novel and critical mechanism for controlling canonical Wnt signaling and cell proliferation.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Wnt Proteins/metabolism , beta Catenin/metabolism , Cell Line , Endopeptidases/metabolism , Forkhead Box Protein M1 , Humans , Transcriptional Activation , Ubiquitination , Wnt Signaling Pathway
20.
PLoS One ; 10(9): e0137386, 2015.
Article in English | MEDLINE | ID: mdl-26372013

ABSTRACT

Bursaphelenchus xylophilus is the pathogen of pine wilt disease. Bursaphelenchus mucronatus is similar to B. xylophilus in morphology. Both species share a common niche, but they are quite different in pathogenicity. Presently, the role of bacteria in pine wilt disease development has been widely speculated. The diversity of bacteria associated with B. xylophilus and B. mucronatus with different virulence remains unclear. In this study, virulence of four B. xylophilus and four B. mucronatus strains were evaluated by inoculating Pinus thunbergii. High-throughput sequencing targeted 16S rDNA of different virulence nematode strains was carried out. The associated bacterial community structures of the eight strains were analyzed. The results showed that 634,051 high-quality sequences were obtained from the eight nematode strains. The number of OTUs of bacteria associated with B. mucronatus was generally greater than those of B. xylophilus. The richness of the community of bacteria associated with high virulent B. xylophilus ZL1 and AmA3 was higher than moderately virulent B. xylophilus AA3, HE2, and all B. mucronatus strains. While the diversity of bacteria associated with B. mucronatus was higher than B. xylophilus. Stenotrophomonas, Pseudomonadaceae_Unclassified or Rhizobiaceae_Unclassified were predominant in the nematode strains with different virulence. Oxalobacteraceae and Achromobacter were found more abundant in the low virulent B. xylophilus and non-virulent B. mucronatus strains.


Subject(s)
Bacteria/classification , Biodiversity , High-Throughput Nucleotide Sequencing , Pinus , Sequence Analysis, DNA , Tylenchida/genetics , Tylenchida/pathogenicity , Animals , Species Specificity , Tylenchida/microbiology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...