Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Mol Cell ; 79(6): 1008-1023.e4, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32871104

ABSTRACT

TMPRSS2-ERG gene fusion occurs in approximately 50% of cases of prostate cancer (PCa), and the fusion product is a key driver of prostate oncogenesis. However, how to leverage cellular signaling to ablate TMPRSS2-ERG oncoprotein for PCa treatment remains elusive. Here, we demonstrate that DNA damage induces proteasomal degradation of wild-type ERG and TMPRSS2-ERG oncoprotein through ERG threonine-187 and tyrosine-190 phosphorylation mediated by GSK3ß and WEE1, respectively. The dual phosphorylation triggers ERG recognition and degradation by the E3 ubiquitin ligase FBW7 in a manner independent of a canonical degron. DNA damage-induced TMPRSS2-ERG degradation was abolished by cancer-associated PTEN deletion or GSK3ß inactivation. Blockade of DNA damage-induced TMPRSS2-ERG oncoprotein degradation causes chemotherapy-resistant growth of fusion-positive PCa cells in culture and in mice. Our findings uncover a previously unrecognized TMPRSS2-ERG protein destruction mechanism and demonstrate that intact PTEN and GSK3ß signaling are essential for effective targeting of ERG protein by genotoxic therapeutics in fusion-positive PCa.


Subject(s)
Cell Cycle Proteins/genetics , Glycogen Synthase Kinase 3 beta/genetics , Oncogene Proteins, Fusion/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , DNA Damage/drug effects , Drug Resistance, Neoplasm/genetics , Drug Therapy , F-Box-WD Repeat-Containing Protein 7/genetics , Heterografts , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Proteolysis/drug effects , Signal Transduction/drug effects
2.
Mol Cell ; 71(4): 592-605.e4, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30057199

ABSTRACT

The bromodomain and extra-terminal domain (BET) protein BRD4 is emerging as a promising anticancer therapeutic target. However, resistance to BET inhibitors often occurs, and it has been linked to aberrant degradation of BRD4 protein in cancer. Here, we demonstrate that the deubiquitinase DUB3 binds to BRD4 and promotes its deubiquitination and stabilization. Expression of DUB3 is transcriptionally repressed by the NCOR2-HDAC10 complex. The NCOR2 gene is frequently deleted in castration-resistant prostate cancer patient specimens, and loss of NCOR2 induces elevation of DUB3 and BRD4 proteins in cancer cells. DUB3-proficient prostate cancer cells are resistant to the BET inhibitor JQ1 in vitro and in mice, but this effect is diminished by DUB3 inhibitory agents such as CDK4/6 inhibitor in a RB-independent manner. Our findings identify a previously unrecognized mechanism causing BRD4 upregulation and drug resistance, suggesting that DUB3 is a viable therapeutic target to overcome BET inhibitor resistance in cancer.


Subject(s)
Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Endopeptidases/genetics , Gene Expression Regulation, Neoplastic , Nuclear Proteins/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Transcription Factors/genetics , Animals , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Disease Progression , Drug Resistance, Neoplasm/genetics , Endopeptidases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Male , Mice , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 2/deficiency , Nuclear Receptor Co-Repressor 2/genetics , Piperazines/pharmacology , Prostate/drug effects , Prostate/enzymology , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Kinase Inhibitors/pharmacology , Proteolysis , Pyridines/pharmacology , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription, Genetic , Triazoles/pharmacology , Xenograft Model Antitumor Assays
3.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33593912

ABSTRACT

Both gene repressor (Polycomb-dependent) and activator (Polycomb-independent) functions of the Polycomb protein enhancer of zeste homolog 2 (EZH2) are implicated in cancer progression. EZH2 protein can be phosphorylated at various residues, such as threonine 487 (T487), by CDK1 kinase, and such phosphorylation acts as a Polycomb repressive complex 2 (PRC2) suppression "code" to mediate the gene repressor-to-activator switch of EZH2 functions. Here we demonstrate that the histone reader protein ZMYND8 is overexpressed in human clear cell renal cell carcinoma (ccRCC). ZMYND8 binds to EZH2, and their interaction is largely enhanced by CDK1 phosphorylation of EZH2 at T487. ZMYND8 depletion not only enhances Polycomb-dependent function of EZH2 in hypoxia-exposed breast cancer cells or von Hippel-Lindau (VHL)-deficient ccRCC cells, but also suppresses the FOXM1 transcription program. We further show that ZMYND8 is required for EZH2-FOXM1 interaction and is important for FOXM1-dependent matrix metalloproteinase (MMP) gene expression and EZH2-mediated migration and invasion of VHL-deficient ccRCC cells. Our results identify a previously uncharacterized role of the chromatin reader ZMYND8 in recognizing the PRC2-inhibitory phosphorylation "code" essential for the Polycomb-dependent to -independent switch of EZH2 functions. They also reveal an oncogenic pathway driving cell migration and invasion in hypoxia-inducible factor-activated (hypoxia or VHL-deficient) cancer.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Movement , Enhancer of Zeste Homolog 2 Protein/metabolism , Hypoxia/physiopathology , Kidney Neoplasms/pathology , Polycomb Repressive Complex 2/metabolism , Tumor Suppressor Proteins/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Proliferation , Chromatin/genetics , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Phosphorylation , Polycomb Repressive Complex 2/genetics , Prognosis , Survival Rate , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics
4.
EMBO J ; 38(5)2019 03 01.
Article in English | MEDLINE | ID: mdl-30723117

ABSTRACT

In light of the increasing number of identified cancer-driven gain-of-function (GOF) mutants of p53, it is important to define a common mechanism to systematically target several mutants, rather than developing strategies tailored to inhibit each mutant individually. Here, using RNA immunoprecipitation-sequencing (RIP-seq), we identified the Polycomb-group histone methyltransferase EZH2 as a p53 mRNA-binding protein. EZH2 bound to an internal ribosome entry site (IRES) in the 5'UTR of p53 mRNA and enhanced p53 protein translation in a methyltransferase-independent manner. EZH2 augmented p53 GOF mutant-mediated cancer growth and metastasis by increasing protein levels of mutant p53. EZH2 overexpression was associated with worsened outcome selectively in patients with p53-mutated cancer. Depletion of EZH2 by antisense oligonucleotides inhibited p53 GOF mutant-mediated cancer growth. Our findings reveal a non-methyltransferase function of EZH2 that controls protein translation of p53 GOF mutants, inhibition of which causes synthetic lethality in cancer cells expressing p53 GOF mutants.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Gain of Function Mutation , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/pathology , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Internal Ribosome Entry Sites , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Stability , RNA, Messenger/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
5.
Eat Weight Disord ; 28(1): 11, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36790552

ABSTRACT

OBJECTIVES: To determine the causal association between genetically predicted obesity and the risk of hip osteoarthritis. METHODS: We performed two-sample Mendelian randomization (MR) analysis to analyze the association between body mass index (BMI) and hip osteoarthritis using pooled-level genome-wide association study (GWAS) data. The inverse variance weighted (IVW), MR‒Egger, and weighted median methods were used to estimate the causal association. In addition, we applied the MR Steiger filtering method, MR robust adjusted profile score (MR.RAPS) methods, and the MR Pleiotropy RESidual Sum and Outlier (MR-PRESSO) global test to examine and address potential horizontal pleiotropy. RESULTS: We found a causal relationship between genetically predicted BMI and the risk of hip osteoarthritis by the IVW method [OR = 1.45, 95% confidence interval (CI) = 1.04-2.00, P = 0.02]. In the sensitivity analysis, the results of the MR‒Egger and weighted median methods revealed similar estimations but with a wide CI with lower precision. The funnel plot, MR-Egger intercept, and MR-PRESSO all indicated the absence of a directional pleiotropic effect. In addition, no heterogeneity was observed in the present analysis. Therefore, the result of IVW is most suitable and reliable for the present MR analysis. CONCLUSION: There is a causal relationship between obesity and a higher risk of hip osteoarthritis, suggesting that weight management may be an intervention for the prevention and management of hip osteoarthritis. LEVEL OF EVIDENCE: Bioinformatics, Basic science.


Subject(s)
Genome-Wide Association Study , Osteoarthritis, Hip , Humans , Osteoarthritis, Hip/genetics , Body Mass Index , Nonoxynol , Obesity/complications , Obesity/genetics , Polymorphism, Single Nucleotide
6.
J Biol Chem ; 292(15): 6269-6280, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28242758

ABSTRACT

Pancreatic cancer is one of the most lethal cancer types. Enhancer of zeste homolog 2 (EZH2) is an oncogenic protein overexpressed in pancreatic cancer, and EZH2 could be a potential therapeutic target for the treatment of pancreatic cancer. Although significant progress has been made toward understanding the function and deregulation of EZH2 in cancer cells, the posttranslational regulation of EZH2 in cancer cells is still unclear. F-box and WD repeat domain-containing 7 (FBW7) acts as a tumor suppressor by targeting multiple oncoprotein substrates for ubiquitination and degradation. Here we demonstrate that EZH2 is a bona fide substrate of FBW7 in pancreatic cancer cells. We provide evidence that the activated CDK5 kinase is involved in the EZH2 phosphorylation that is required for FBW7-mediated degradation. We further show that FBW7 suppresses EZH2 activity and inhibits tumor migration and invasion via degradation of EZH2 in pancreatic cancer cells. Furthermore, immunohistochemistry analysis revealed that expression of EZH2 protein negatively correlates with FBW7 protein levels in a cohort of human pancreatic cancer specimens. Collectively, our findings demonstrate that FBW7 is a novel E3 ligase of EZH2 that regulates the EZH2 protein level in pancreatic cancer and represents a viable strategy for effective treatment of pancreatic cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Movement , Cyclin-Dependent Kinase 5/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , F-Box Proteins/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 5/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Humans , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Ubiquitin-Protein Ligases/genetics
7.
BMC Anesthesiol ; 18(1): 138, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30285627

ABSTRACT

BACKGROUND: The goal of this study was to evaluate the accuracy and interchangeability between continuous cardiac output (CO) measured by electrical velocimetry (COEv) and continuous cardiac output obtained using the pulmonary thermodilution method (COPAC) during living donor liver transplantation (LDLT). METHOD: Twenty-three patients were enrolled in this prospective observational study. CO was recorded by both two methods and compared at nine specific time points. The data were analyzed using correlation coefficients, Bland-Altman analysis for the percentage errors, and the concordance rate for trend analysis using a four-quadrant plot. RESULTS: In total, 207 paired datasets were recorded during LDLT. CO data were in the range of 2.8-12.7 L/min measured by PAC and 3.4-14.9 L/min derived from the EV machine. The correction coefficient between COPAC and COEv was 0.415 with p < 0.01. The 95% limitation agreement was - 5.9 to 3.4 L/min and the percentage error was 60%. The concordance rate was 56.5%. CONCLUSIONS: The Aesculon™ monitor is not yet interchangeable with continuous thermodilution CO monitoring during LDLT. TRIAL REGISTRATION: The study was approved by the Institutional Review Board of Chang Gung Medical Foundation in Taiwan (registration number: 201600264B0 ).


Subject(s)
Cardiac Output/physiology , Liver Transplantation/methods , Lung/physiology , Monitoring, Intraoperative/methods , Rheology/methods , Adult , Aged , Female , Humans , Liver Transplantation/adverse effects , Liver Transplantation/standards , Male , Middle Aged , Monitoring, Intraoperative/standards , Prospective Studies , Rheology/standards , Thermodilution/methods , Thermodilution/standards
8.
J Cell Sci ; 128(5): 992-1000, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25588844

ABSTRACT

A hallmark of neutrophil polarization is the back localization of active RHOA and phosphorylated myosin light chain (pMLC, also known as MYL2). However, the mechanism for the polarization is not entirely clear. Here, we show that FAM65B, a newly identified RHOA inhibitor, is important for the polarization. When FAM65B is phosphorylated, it binds to 14-3-3 family proteins and becomes more stable. In neutrophils, chemoattractants stimulate FAM65B phosphorylation largely depending on the signals from the front of the cells that include those mediated by phospholipase Cß (PLCß) and phosphoinositide 3-kinase γ (PI3Kγ), leading to FAM65B accumulation at the leading edge. Concordantly, FAM65B deficiency in neutrophils resulted in an increase in RHOA activity and localization of pMLC to the front of cells, as well as defects in chemotaxis directionality and adhesion to endothelial cells under flow. These data together elucidate a mechanism for RHOA and pMLC polarization in stimulated neutrophils through direct inhibition of RHOA by FAM65B at the leading edge.


Subject(s)
Neutrophils/metabolism , Proteins/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules , Chemotaxis/physiology , Class Ib Phosphatidylinositol 3-Kinase , Endothelial Cells/cytology , Endothelial Cells/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Neutrophils/cytology , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Phosphorylation/physiology , Proteins/genetics , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics
9.
Hum Mutat ; 35(9): 1142-51, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24990631

ABSTRACT

Characterization of the exome and genome of prostate cancers by next-generation sequencing has identified numerous genetic alternations. SPOP (speckle-type POZ protein) was identified as one of the most frequently affected genes by somatic point mutations in prostate cancer, suggesting SPOP is potentially a key driver for prostate cancer development and progression. However, how SPOP mutations contribute to prostate cancer remains to be elucidated. SPOP acts as an adaptor protein of the CUL3-RBX1 E3 ubiquitin ligase complex and selectively recruits substrates for their ubiquitination and subsequent degradation. DDIT3 is an endoplasmic reticulum (ER) stress-responsive transcription factor playing an essential role in apoptotic execution pathways triggered by ER stress. Here, we identified DDIT3/CHOP as a bona fide substrate for the SPOP-CUL3-RBX1 E3 ubiquitin ligase complex. SPOP recognizes a Ser/Thr-rich degron in the transactivation domain of DDIT3 and triggers DDIT3 degradation via the ubiquitin-proteasome pathway. Strikingly, prostate cancer-associated mutants of SPOP are defective in promoting DDIT3 degradation. This study reveals novel molecular events underlying the regulation of DDIT3 protein homeostasis and provides insight in understanding the relationship between SPOP mutations and ER stress dysregulation in prostate cancer.


Subject(s)
Mutation , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Repressor Proteins/genetics , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Amino Acid Motifs , Amino Acid Sequence , Apoptosis/genetics , Carrier Proteins/metabolism , Cullin Proteins/metabolism , Endoplasmic Reticulum Stress , Humans , Male , Multiprotein Complexes , Protein Binding , Protein Interaction Domains and Motifs , Proteolysis , Transcription Factor CHOP/chemistry , Transcription, Genetic , Ubiquitination
10.
J Biol Chem ; 288(1): 529-39, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23150668

ABSTRACT

Polycomb group protein PHF1 is well known as a component of a novel EED-EZH2·Polycomb repressive complex 2 complex and plays important roles in H3K27 methylation and Hox gene silencing. PHF1 is also involved in the response to DNA double-strand breaks in human cells, promotes nonhomologous end-joining processes through interaction with Ku70/Ku80. Here, we identified another function of PHF1 as a potential p53 pathway activator in a pathway screen using luminescence reporter assay. Subsequent studies showed PHF1 directly interacts with p53 proteins both in vivo and in vitro and co-localized in nucleus. PHF1 binds to the C-terminal regulatory domain of p53. Overexpression of PHF1 elevated p53 protein level and prolonged its turnover. Knockdown of PHF1 reduced p53 protein level and its target gene expression both in normal state and DNA damage response. Mechanically, PHF1 protects p53 proteins from MDM2-mediated ubiquitination and degradation. Furthermore, we showed that PHF1 regulates cell growth arrest and etoposide-induced apoptosis in a p53-dependent manner. Finally, PHF1 expression was significantly down-regulated in human breast cancer samples. Taken together, we establish PHF1 as a novel positive regulator of the p53 pathway. These data shed light on the potential roles of PHF1 in tumorigenesis and/or tumor progression.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Polycomb-Group Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , DNA Damage , DNA-Binding Proteins/chemistry , Female , Gene Silencing , Humans , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , Transcription Factors/chemistry , Ubiquitin/chemistry
11.
Biochem J ; 444(2): 279-89, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22385262

ABSTRACT

AMOT (angiomotin) is a membrane-associated protein that is expressed in ECs (endothelial cells) and controls migration, TJ (tight junction) formation, cell polarity and angiogenesis. Recent studies have revealed that AMOT and two AMOT-like proteins, AMOTL1 and AMOTL2, play critical roles in the Hippo pathway by regulating the subcellular localization of the co-activators YAP (Yes-associated protein) and TAZ (transcriptional co-activator with PDZ-binding motif). However, it has been unclear how AMOT is regulated. In the present study, we report that AMOT undergoes proteasomal degradation. We identify three members of Nedd4 (neural-precursor-cell-expressed developmentally down-regulated)-like ubiquitin E3 ligases, Nedd4, Nedd4-2 and Itch, as the ubiquitin E3 ligases for the long isoform of AMOT, AMOT/p130. We demonstrate that Nedd4, Nedd4-2 and Itch mediate poly-ubiquitination of AMOT/p130 in vivo. Overexpression of Nedd4, Nedd4-2 or Itch leads to AMOT/p130 proteasomal degradation. Knockdown of Nedd4, Nedd4-2 and Itch causes an accumulation of steady-state level of AMOT/p130. We also show that three L/P-PXY motifs of AMOT/p130 and the WW domains of Nedd4 mediate their interaction. Furthermore, Nedd4-like ubiquitin E3 ligases might compete with YAP for the binding to AMOT/p130, and subsequently targeting AMOT/p130 for ubiquitin-dependent degradation. Together, these observations reveal a novel post-translational regulatory mechanism of AMOT/p130.


Subject(s)
Endosomal Sorting Complexes Required for Transport/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Proteolysis , Ubiquitin-Protein Ligases/physiology , Ubiquitin/metabolism , Amino Acid Sequence , Angiomotins , Endosomal Sorting Complexes Required for Transport/deficiency , Endosomal Sorting Complexes Required for Transport/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Microfilament Proteins , Molecular Sequence Data , Nedd4 Ubiquitin Protein Ligases , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/deficiency , Repressor Proteins/genetics , Signal Transduction/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
12.
BMC Genom Data ; 24(1): 56, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37749495

ABSTRACT

BACKGROUND: Recently accumulated evidence indicates a potential association between COVID-19 and elevated susceptibility to cancer, including male genital cancer. However, the causal nature of this relationship remains unclear. METHODS: In this Mendelian randomization (MR) study, we investigated the potential causal relationship between COVID-19 and male genital cancer using genetic variants as instrumental variables. We utilized summary statistics from two large-scale genome-wide association studies of COVID-19 hospitalized Vs. controls, as well as data from a population-based male genital cancer database based on European ancestry. We applied stringent quality control measures to select instrumental variables, including checking for linkage disequilibrium, removing low-quality variants, and assessing the strength of the instruments using the F-statistic. We conducted the MR  analysis using the inverse-variance weighted method and several sensitivity analyses (including MR Egger and Weighted Median MR analysis) to test the robustness of our results. RESULTS: Our MR analysis revealed no causal associations between COVID-19 hospitalization and the incidence of male genital cancer. In the inverse-variance weighted analysis, no causal associations were observed between patients with COVID-19 hospitalization and the incidence of male genital cancer (odds ratio = 1.000 and 95% confidence interval = 0.998-1.001, p = 0.668). The estimated causal effect was consistent across all sensitivity analyses (including the Weighted Median, the MR Egger analysis, and the MR PROSSO analysis). The leave-one-out analysis showed that there was no any sing Single-nucleotide polymorphism significantly influencing our results. CONCLUSIONS: Our study provides evidence that there is no causal association between COVID-19 hospitalization and male genital cancer.


Subject(s)
COVID-19 , Genital Neoplasms, Male , Humans , Male , Mendelian Randomization Analysis , Genome-Wide Association Study , COVID-19/epidemiology , COVID-19/genetics , Genitalia, Male
13.
Front Oncol ; 12: 911466, 2022.
Article in English | MEDLINE | ID: mdl-36237339

ABSTRACT

Rationale: Fructose-1,6-bisphosphatase (FBP1) is a tumor suppressor and a key enzyme negatively regulating Warburg effect in cancer. However, regulation of FBP1 protein expression and its exact role in prostate cancer (PCa) is largely unclear. Phosphatase and tensin homolog (PTEN) is one of the most frequently deleted tumor suppressor genes in human PCa. However, the role of PTEN loss in aberrant Warburg effect in cancer remains poorly understood. Methods: Expression of PTEN and FBP1 was analyzed in several PCa cell lines and prostate tumor tissues in mice. Western blot (WB) and RT-PCR approaches were used to examine how PTEN regulates FBP1 expression. Co-immunoprecipitation (co-IP) and in vivo ubiquitination assays were used to define the regulatory mechanisms. A PCa xenograft model was employed to determine the impact of PTEN regulation of FBP1 on PCa growth in vivo. Result: We demonstrated that in a manner dependent of PI3K/AKT signal pathway PTEN regulated FBP1 expression in various PCa cell lines and tumors in mice. We confirmed that this regulation took place at the protein level and was mediated by SKP2 E3 ubiquitin ligase. Mechanistically, we showed that serine 271 phosphorylation of FBP1 by cyclin-dependent kinases (CDKs) was essential for SKP2-mediated degradation of FBP1 protein induced by PTEN loss. Most importantly, we further showed that loss of PTEN expression enhanced Warburg effect and PCa growth in mice in a manner dependent, at least partially on FBP1 protein degradation. Conclusions: Our results reveal a novel tumor-suppressive feature of PTEN in restraining FBP1 degradation and the Warburg effect. These results also suggest that prohibiting FBP1 protein degradation could be a viable therapeutic strategy for PTEN-deficient PCa.

14.
J Biol Chem ; 285(12): 8869-79, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20068034

ABSTRACT

Thioredoxin interacting protein (TXNIP) was originally characterized as an endogenous inhibitor of thioredoxin, a key regulator in cellular redox homeostasis. TXNIP is also known to play important roles in tumor growth and metastasis, glucose and lipid metabolism. TXNIP expression is induced by various stress stimuli. However, it has been unclear how TXNIP is down-regulated. Here, we report that TXNIP undergoes proteasomal degradation in cells. We identify Itch as the E3 ubiquitin ligase for TXNIP. We demonstrate that Itch mediates polyubiquitination of TXNIP both in vitro and in vivo. Overexpression of Itch leads to TXNIP proteasomal degradation. Knockdown of Itch by small interfering RNA causes an accumulation of the steady-state level of TXNIP. We also show that the PPXY motifs of TXNIP and the WW domains of Itch mediate their interaction. Furthermore, the Itch-TXNIP interaction regulates intracellular reactive oxygen species levels and apoptosis. These findings establish a new mechanism for the negative regulation of TXNIP by Itch and shed new light on the regulation of cellular redox homeostasis.


Subject(s)
Apoptosis , Carrier Proteins/chemistry , Repressor Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Ubiquitin/chemistry , Amino Acid Motifs , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Glutathione Transferase/metabolism , Humans , Oxidation-Reduction , Proteasome Endopeptidase Complex/metabolism , RNA Interference , RNA, Small Interfering/metabolism
15.
Adv Sci (Weinh) ; 8(20): e2102555, 2021 10.
Article in English | MEDLINE | ID: mdl-34397171

ABSTRACT

DNA-binding proteins, including transcription factors (TFs), play essential roles in various cellular processes and pathogenesis of diseases, deeming to be potential therapeutic targets. However, these proteins are generally considered undruggable as they lack an enzymatic catalytic site or a ligand-binding pocket. Proteolysis-targeting chimera (PROTAC) technology has been developed by engineering a bifunctional molecule chimera to bring a protein of interest (POI) to the proximity of an E3 ubiquitin ligase, thus inducing the ubiquitination of POI and further degradation through the proteasome pathway. Here, the development of oligonucleotide-based PROTAC (O'PROTACs), a class of noncanonical PROTACs in which a TF-recognizing double-stranded oligonucleotide is incorporated as a binding moiety of POI is reported. It is demonstrated that O'PROTACs of lymphoid enhancer-binding factor 1 (LEF1) and ETS-related gene (ERG), two highly cancer-related transcription factors, successfully promote degradation of these proteins, impede their transcriptional activity, and inhibit cancer cell growth in vitro and in vivo. The programmable nature of O'PROTACs indicates that this approach is also applicable to destruct other TFs. O'PROTACs not only can serve as a research tool but also can be harnessed as a therapeutic arsenal to target DNA binding proteins for effective treatment of diseases such as cancer.


Subject(s)
DNA-Binding Proteins/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Neoplasms/genetics , Oligonucleotides/genetics , Humans , Neoplasms/therapy , Oligonucleotides/pharmacology , Proteasome Endopeptidase Complex/genetics , Proteolysis/drug effects , Transcriptional Regulator ERG/genetics , Ubiquitin-Protein Ligases/genetics
16.
Cell Rep ; 34(7): 108744, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33596421

ABSTRACT

Acquisition of resistance to phosphatidylinositol 3-kinase (PI3K)/AKT-targeted monotherapy implies the existence of common resistance mechanisms independent of cancer type. Here, we demonstrate that PI3K/AKT inhibitors cause glycolytic crisis, acetyl-coenzyme A (CoA) shortage, and a global decrease in histone acetylation. In addition, PI3K/AKT inhibitors induce drug resistance by selectively augmenting histone H3 lysine 27 acetylation (H3K27ac) and binding of CBP/p300 and BRD4 proteins at a subset of growth factor and receptor (GF/R) gene loci. BRD4 occupation at these loci and drug-resistant cell growth are vulnerable to both bromodomain and histone deacetylase (HDAC) inhibitors. Little or no occupation of HDAC proteins at the GF/R gene loci underscores the paradox that cells respond equivalently to the two classes of inhibitors with opposite modes of action. Targeting this unique acetyl-histone-related vulnerability offers two clinically viable strategies to overcome PI3K/AKT inhibitor resistance in different cancers.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Neoplasms/drug therapy , Nerve Tissue Proteins/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , HCT116 Cells , Humans , Male , Mice , Mice, SCID , Neoplasms/enzymology , Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/metabolism , Xenograft Model Antitumor Assays
17.
Sci Adv ; 7(25)2021 06.
Article in English | MEDLINE | ID: mdl-34144977

ABSTRACT

53BP1 activates nonhomologous end joining (NHEJ) and inhibits homologous recombination (HR) repair of DNA double-strand breaks (DSBs). Dissociation of 53BP1 from DSBs and consequent activation of HR, a less error-prone pathway than NHEJ, helps maintain genome integrity during DNA replication; however, the underlying mechanisms are not fully understood. Here, we demonstrate that E3 ubiquitin ligase SPOP promotes HR during S phase of the cell cycle by excluding 53BP1 from DSBs. In response to DNA damage, ATM kinase-catalyzed phosphorylation of SPOP causes a conformational change in SPOP, revealed by x-ray crystal structures, that stabilizes its interaction with 53BP1. 53BP1-bound SPOP induces polyubiquitination of 53BP1, eliciting 53BP1 extraction from chromatin by a valosin-containing protein/p97 segregase complex. Our work shows that SPOP facilitates HR repair over NHEJ during DNA replication by contributing to 53BP1 removal from chromatin. Cancer-derived SPOP mutations block SPOP interaction with 53BP1, inducing HR defects and chromosomal instability.


Subject(s)
Chromatin , DNA Breaks, Double-Stranded , Chromatin/genetics , DNA End-Joining Repair , DNA Replication , Nuclear Proteins , Recombinational DNA Repair , Repressor Proteins , Tumor Suppressor p53-Binding Protein 1
18.
Nat Commun ; 12(1): 5716, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34588438

ABSTRACT

Mutations in SPOP E3 ligase gene are reportedly associated with genome-wide DNA hypermethylation in prostate cancer (PCa) although the underlying mechanisms remain elusive. Here, we demonstrate that SPOP binds and promotes polyubiquitination and degradation of histone methyltransferase and DNMT interactor GLP. SPOP mutation induces stabilization of GLP and its partner protein G9a and aberrant upregulation of global DNA hypermethylation in cultured PCa cells and primary PCa specimens. Genome-wide DNA methylome analysis shows that a subset of tumor suppressor genes (TSGs) including FOXO3, GATA5, and NDRG1, are hypermethylated and downregulated in SPOP-mutated PCa cells. DNA methylation inhibitor 5-azacytidine effectively reverses expression of the TSGs examined, inhibits SPOP-mutated PCa cell growth in vitro and in mice, and enhances docetaxel anti-cancer efficacy. Our findings reveal the GLP/G9a-DNMT module as a mediator of DNA hypermethylation in SPOP-mutated PCa. They suggest that SPOP mutation could be a biomarker for effective treatment of PCa with DNA methylation inhibitor alone or in combination with taxane chemotherapeutics.


Subject(s)
DNA Methylation/genetics , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Repressor Proteins/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/pharmacology , Azacitidine/therapeutic use , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Docetaxel/pharmacology , Docetaxel/therapeutic use , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , Humans , Male , Mice , Mutation , Nuclear Proteins/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Stability/drug effects , Proteolysis/drug effects , Repressor Proteins/metabolism , Xenograft Model Antitumor Assays
19.
Gut Microbes ; 11(6): 1567-1589, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32588736

ABSTRACT

Helicobacter pylori (H. pylori) infection is the strongest known risk factor for the development of gastric cancer. DNA damage response (DDR) and autophagy play key roles in tumorigenic transformation. However, it remains unclear how H. pylori modulate DDR and autophagy in gastric carcinogenesis. Here we report that H. pylori infection promotes DNA damage via suppression of Rad51 expression through inhibition of autophagy and accumulation of p62 in gastric carcinogenesis. We find that H. pylori activated DNA damage pathway in concert with downregulation of repair protein Rad51 in gastric cells, C57BL/6 mice and Mongolian gerbils. In addition, autophagy was increased early and then decreased gradually during the duration of H. pylori infection in vitro in a CagA-dependent manner. Moreover, loss of autophagy led to promotion of DNA damage in H. pylori-infected cells. Furthermore, knockdown of autophagic substrate p62 upregulated Rad51 expression, and p62 promoted Rad51 ubiquitination via the direct interaction of its UBA domain. Finally, H. pylori infection was associated with elevated levels of p62 in gastric intestinal metaplasia and decreased levels of Rad51 in dysplasia compared to their H. pylori- counterparts. Our findings provide a novel mechanism into the linkage of H. pylori infection, autophagy, DNA damage and gastric tumorigenesis.


Subject(s)
Autophagy , DNA Damage , Helicobacter Infections/metabolism , Helicobacter pylori/physiology , Rad51 Recombinase/metabolism , Stomach Neoplasms/metabolism , Adult , Aged , Animals , Carcinogenesis , Female , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/physiopathology , Helicobacter pylori/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Rad51 Recombinase/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Stomach Neoplasms/physiopathology , Ubiquitination
20.
EMBO Mol Med ; 11(11): e10659, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31559706

ABSTRACT

CULLIN3-based E3 ubiquitin ligase substrate-binding adaptor gene SPOP is frequently mutated in prostate cancer (PCa). PCa harboring SPOP hotspot mutants (e.g., F133V) are resistant to BET inhibitors because of aberrant elevation of BET proteins. Here, we identified a previously unrecognized mutation Q165P at the edge of SPOP MATH domain in primary and metastatic PCa of a patient. The Q165P mutation causes structural changes in the MATH domain and impairs SPOP dimerization and substrate degradation. Different from F133V hotspot mutant tumors, Q165P mutant patient-derived xenografts (PDXs) and organoids were modestly sensitive to the BET inhibitor JQ1. Accordingly, protein levels of AR, BRD4 and downstream effectors such as RAC1 and phosphorylated AKT were not robustly elevated in Q165P mutant cells as in F133V mutant cells. However, NEO2734, a novel dual inhibitor of BET and CBP/p300, is active in both hotspot mutant (F133V) and non-hotspot mutant (Q165P) PCa cells in vitro and in vivo. These data provide a strong rationale to clinically investigate the anti-cancer efficacy of NEO2734 in SPOP-mutated PCa patients.


Subject(s)
Antineoplastic Agents/pharmacology , E1A-Associated p300 Protein/metabolism , Mutant Proteins/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Prostatic Neoplasms/pathology , Repressor Proteins/metabolism , Sialoglycoproteins/metabolism , Transcription Factors/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Disease Models, Animal , E1A-Associated p300 Protein/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Heterografts , Humans , Male , Mutant Proteins/genetics , Mutation, Missense , Neoplasm Transplantation , Nuclear Proteins/genetics , Peptide Fragments/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Repressor Proteins/genetics , Sialoglycoproteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL