Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Nucleic Acids Res ; 2024 May 06.
Article En | MEDLINE | ID: mdl-38709899

Emerging evidence indicates that arginine methylation promotes the stability of arginine-glycine-rich (RGG) motif-containing RNA-binding proteins (RBPs) and regulates gene expression. Here, we report that post-translational modification of FXR1 enhances the binding with mRNAs and is involved in cancer cell growth and proliferation. Independent point mutations in arginine residues of FXR1's nuclear export signal (R386 and R388) and RGG (R453, R455 and R459) domains prevent it from binding to RNAs that form G-quadruplex (G4) RNA structures. Disruption of G4-RNA structures by lithium chloride failed to bind with FXR1, indicating its preference for G4-RNA structure containing mRNAs. Furthermore, loss-of-function of PRMT5 inhibited FXR1 methylation both in vivo and in vitro, affecting FXR1 protein stability, inhibiting RNA-binding activity and cancer cell growth and proliferation. Finally, the enhanced crosslinking and immunoprecipitation (eCLIP) analyses reveal that FXR1 binds with the G4-enriched mRNA targets such as AHNAK, MAP1B, AHNAK2, HUWE1, DYNC1H1 and UBR4 and controls its mRNA expression in cancer cells. Our findings suggest that PRMT5-mediated FXR1 methylation is required for RNA/G4-RNA binding, which promotes gene expression in cancer cells. Thus, FXR1's structural characteristics and affinity for RNAs preferentially G4 regions provide new insights into the molecular mechanism of FXR1 in oral cancer cells.

2.
Sci Rep ; 14(1): 11756, 2024 May 23.
Article En | MEDLINE | ID: mdl-38783024

Visual place recognition (VPR) involves obtaining robust image descriptors to cope with differences in camera viewpoints and drastic external environment changes. Utilizing multiscale features improves the robustness of image descriptors; however, existing methods neither exploit the multiscale features generated during feature extraction nor consider the feature redundancy problem when fusing multiscale information when image descriptors are enhanced. We propose a novel encoding strategy-convolutional multilayer perceptron orthogonal fusion of multiscale features (ConvMLP-OFMS)-for VPR. A ConvMLP is used to obtain robust and generalized global image descriptors and the multiscale features generated during feature extraction are used to enhance the global descriptors to cope with changes in the environment and viewpoints. Additionally, an attention mechanism is used to eliminate noise and redundant information. Compared to traditional methods that use tensor splicing for feature fusion, we introduced matrix orthogonal decomposition to eliminate redundant information. Experiments demonstrated that the proposed architecture outperformed NetVLAD, CosPlace, ConvAP, and other methods. On the Pittsburgh and MSLS datasets, which contained significant viewpoint and illumination variations, our method achieved 92.5% and 86.5% Recall@1, respectively. We also achieved good performances-80.6% and 43.2%-on the SPED and NordLand datasets, respectively, which have more extreme illumination and appearance variations.

3.
Genome Instab Dis ; 4(4): 197-209, 2023 Aug.
Article En | MEDLINE | ID: mdl-37663901

DNA double-strand breaks (DSBs) are widely accepted to be the most deleterious form of DNA lesions that pose a severe threat to genome integrity. Two predominant pathways are responsible for repair of DSBs, homologous recombination (HR) and non-homologous end-joining (NHEJ). HR relies on a template to faithfully repair breaks, while NHEJ is a template-independent and error-prone repair mechanism. Multiple layers of regulation have been documented to dictate the balance between HR and NHEJ, such as cell cycle and post-translational modifications (PTMs). Arginine methylation is one of the most common PTMs, which is catalyzed by protein arginine methyltransferases (PRMTs). PRMT1 and PRMT5 are the predominate PRMTs that promote asymmetric dimethylarginine and symmetric dimethylarginine, respectively. They have emerged to be crucial regulators of DNA damage repair. In this review, we summarize current understanding and unaddressed questions of PRMT1 and PRMT5 in regulation of HR and NHEJ, providing insights into their roles in DSB repair pathway choice and the potential of targeting them for cancer therapy.

4.
Mol Cancer Res ; 21(12): 1317-1328, 2023 12 01.
Article En | MEDLINE | ID: mdl-37606694

Although androgen deprivation treatment often effectively decreases prostate cancer, incurable metastatic castration-resistant prostate cancer (CRPC) eventually occurs. It is important to understand how CRPC metastasis progresses, which is not clearly defined. The loss of PTEN, a phosphatase to dephosphorylate phosphatidylinositol 3,4,5-trisphosphate in the PI3K pathway, occurs in up to 70% to 80% of CRPC. We generated a mouse androgen-independent prostate cancer cell line (PKO) from PTEN null and Hi-Myc transgenic mice in C57BL/6 background. We confirmed that this PKO cell line has an activated PI3K pathway and can metastasize into the femur and tibia of immunodeficient nude and immunocompetent C57BL/6 mice. In vitro, we found that androgen deprivation significantly enhanced PKO cell migration/invasion via the p110ß isoform-depended PAK1-MAPK activation. Inhibition of the p110ß-PAK1 axis significantly decreased prostate cancer cell migration/invasion. Of note, our analysis using clinical samples showed that PAK1 is more activated in CRPC than in advanced prostate cancer; high PAK1/phosphorylated-PAK1 levels are associated with decreased survival rates in patients with CRPC. All the information suggests that this cell line reflects the characteristics of CRPC cells and can be applied to dissect the mechanism of CRPC initiation and progression. This study also shows that PAK1 is a potential target for CRPC treatment. IMPLICATIONS: This study uses a newly generated PTEN null prostate cancer cell line to define a critical functional role of p110ß-PAK1 in CRPC migration/invasion. This study also shows that the p110ß-PAK1 axis can potentially be a therapeutic target in CRPC metastasis.


Prostatic Neoplasms, Castration-Resistant , Animals , Humans , Male , Mice , Androgen Antagonists , Androgens/therapeutic use , Cell Line, Tumor , Mice, Inbred C57BL , Mice, Transgenic , p21-Activated Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Receptors, Androgen/metabolism
5.
Sci Rep ; 13(1): 10752, 2023 07 03.
Article En | MEDLINE | ID: mdl-37400460

Protein arginine methyltransferase 5 (PRMT5) catalyzes mono-methylation and symmetric di-methylation on arginine residues and has emerged as a potential antitumor target with inhibitors being tested in clinical trials. However, it remains unknown how the efficacy of PRMT5 inhibitors is regulated. Here we report that autophagy blockage enhances cellular sensitivity to PRMT5 inhibitor in triple negative breast cancer cells. Genetic ablation or pharmacological inhibition of PRMT5 triggers cytoprotective autophagy. Mechanistically, PRMT5 catalyzes monomethylation of ULK1 at R532 to suppress ULK1 activation, leading to attenuation of autophagy. As a result, ULK1 inhibition blocks PRMT5 deficiency-induced autophagy and sensitizes cells to PRMT5 inhibitor. Our study not only identifies autophagy as an inducible factor that dictates cellular sensitivity to PRMT5 inhibitor, but also unearths a critical molecular mechanism by which PRMT5 regulates autophagy through methylating ULK1, providing a rationale for the combination of PRMT5 and autophagy inhibitors in cancer therapy.


Protein-Arginine N-Methyltransferases , Triple Negative Breast Neoplasms , Humans , Protein-Arginine N-Methyltransferases/metabolism , Methylation , Enzyme Inhibitors/pharmacology , Autophagy
6.
Cancers (Basel) ; 15(9)2023 Apr 27.
Article En | MEDLINE | ID: mdl-37173967

Protein arginine methyltransferase 5 (PRMT5) is the primary enzyme generating symmetric dimethylarginine (sDMA) on numerous substrates, through which it regulates many cellular processes, such as transcription and DNA repair. Aberrant expression and activation of PRMT5 is frequently observed in various human cancers and associated with poor prognosis and survival. However, the regulatory mechanisms of PRMT5 remain poorly understood. Here, we report that TRAF6 serves as an upstream E3 ubiquitin ligase to promote PRMT5 ubiquitination and activation. We find that TRAF6 catalyzes K63-linked ubiquitination of PRMT5 and interacts with PRMT5 in a TRAF6-binding-motif-dependent manner. Moreover, we identify six lysine residues located at the N-terminus as the primarily ubiquitinated sites. Disruption of TRAF6-mediated ubiquitination decreases PRMT5 methyltransferase activity towards H4R3 in part by impairing PRMT5 interaction with its co-factor MEP50. As a result, mutating the TRAF6-binding motifs or the six lysine residues significantly suppresses cell proliferation and tumor growth. Lastly, we show that TRAF6 inhibitor enhances cellular sensitivity to PRMT5 inhibitor. Therefore, our study reveals a critical regulatory mechanism of PRMT5 in cancers.

7.
Cell Rep ; 42(4): 112316, 2023 04 25.
Article En | MEDLINE | ID: mdl-36995937

The mammalian target of rapamycin complex1 (mTORC1) is a central regulator of metabolism and cell growth by sensing diverse environmental signals, including amino acids. The GATOR2 complex is a key component linking amino acid signals to mTORC1. Here, we identify protein arginine methyltransferase 1 (PRMT1) as a critical regulator of GATOR2. In response to amino acids, cyclin-dependent kinase 5 (CDK5) phosphorylates PRMT1 at S307 to promote PRMT1 translocation from nucleus to cytoplasm and lysosome, which in turn methylates WDR24, an essential component of GATOR2, to activate the mTORC1 pathway. Disruption of the CDK5-PRMT1-WDR24 axis suppresses hepatocellular carcinoma (HCC) cell proliferation and xenograft tumor growth. High PRMT1 protein expression is associated with elevated mTORC1 signaling in patients with HCC. Thus, our study dissects a phosphorylation- and arginine methylation-dependent regulatory mechanism of mTORC1 activation and tumor growth and provides a molecular basis to target this pathway for cancer therapy.


Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Amino Acids/metabolism , Cyclin-Dependent Kinase 5 , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism
8.
Sci Adv ; 8(49): eadd8928, 2022 12 09.
Article En | MEDLINE | ID: mdl-36475791

BRD4 functions as an epigenetic reader and plays a crucial role in regulating transcription and genome stability. Dysregulation of BRD4 is frequently observed in various human cancers. However, the molecular details of BRD4 regulation remain largely unknown. Here, we report that PRMT2- and PRMT4-mediated arginine methylation is pivotal for BRD4 functions on transcription, DNA repair, and tumor growth. Specifically, PRMT2/4 interacts with and methylates BRD4 at R179, R181, and R183. This arginine methylation selectively controls a transcriptional program by promoting BRD4 recruitment to acetylated histones/chromatin. Moreover, BRD4 arginine methylation is induced by DNA damage and thereby promotes its binding to chromatin for DNA repair. Deficiency in BRD4 arginine methylation significantly suppresses tumor growth and sensitizes cells to BET inhibitors and DNA damaging agents. Therefore, our findings reveal an arginine methylation-dependent regulatory mechanism of BRD4 and highlight targeting PRMT2/4 for better antitumor effect of BET inhibitors and DNA damaging agents.


Neoplasms , Nuclear Proteins , Humans , Nuclear Proteins/genetics , Arginine , Transcription Factors/genetics , DNA Repair , DNA , Chromatin , Protein-Arginine N-Methyltransferases/genetics , Intracellular Signaling Peptides and Proteins , Cell Cycle Proteins/genetics
9.
Int J Mol Sci ; 23(17)2022 Aug 29.
Article En | MEDLINE | ID: mdl-36077176

In response to DNA damage, cells have developed a sophisticated signaling pathway, consisting of DNA damage sensors, transducers, and effectors, to ensure efficient and proper repair of damaged DNA. During this process, posttranslational modifications (PTMs) are central events that modulate the recruitment, dissociation, and activation of DNA repair proteins at damage sites. Emerging evidence reveals that protein arginine methylation is one of the common PTMs and plays critical roles in DNA damage response. Protein arginine methyltransferases (PRMTs) either directly methylate DNA repair proteins or deposit methylation marks on histones to regulate their transcription, RNA splicing, protein stability, interaction with partners, enzymatic activities, and localization. In this review, we summarize the substrates and roles of each PRMTs in DNA damage response and discuss the synergistic anticancer effects of PRMTs and DNA damage pathway inhibitors, providing insight into the significance of arginine methylation in the maintenance of genome integrity and cancer therapies.


Histones , Protein-Arginine N-Methyltransferases , Arginine/metabolism , DNA Damage , Histones/metabolism , Methylation , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism
10.
Sci Signal ; 15(715): eabh2290, 2022 01 04.
Article En | MEDLINE | ID: mdl-34982576

The kinase AKT (also known as protein kinase B) is a key regulator of cell proliferation, survival, and metabolism. In addition to being activated by growth factors, AKT is activated in response to DNA damage. Here, we found that the DNA damage response kinase DNA-PK sustains cell survival through a phosphorylation event that leads to increased AKT activity. In various cancer and noncancer cells in culture, DNA damage caused by ionizing radiation or topoisomerase inhibitors triggered DNA-PK­dependent phosphorylation of the mTOR complex 2 (mTORC2) subunit Sin1, which enabled its interaction with the guanine nucleotide exchange factor ECT2. Depleting Sin1 or ECT2 or disrupting the protein interaction or catalytic function of ECT2 attenuated DNA damage­induced AKT activation, thereby enhancing cellular sensitivity to DNA-damaging agents. Our findings elucidate a mechanism mediating DNA damage­induced AKT activation and cell survival.


Proto-Oncogene Proteins c-akt , TOR Serine-Threonine Kinases , Adaptor Proteins, Signal Transducing/metabolism , DNA Damage , Mechanistic Target of Rapamycin Complex 2/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
11.
Nat Commun ; 12(1): 6704, 2021 11 18.
Article En | MEDLINE | ID: mdl-34795215

Chromosomal rearrangements can generate genetic fusions composed of two distinct gene sequences, many of which have been implicated in tumorigenesis and progression. Our study proposes a model whereby oncogenic gene fusions frequently alter the protein stability of the resulting fusion products, via exchanging protein degradation signal (degron) between gene sequences. Computational analyses of The Cancer Genome Atlas (TCGA) identify 2,406 cases of degron exchange events and reveal an enrichment of oncogene stabilization due to loss of degrons from fusion. Furthermore, we identify and experimentally validate that some recurrent fusions, such as BCR-ABL, CCDC6-RET and PML-RARA fusions, perturb protein stability by exchanging internal degrons. Likewise, we also validate that EGFR or RAF1 fusions can be stabilized by losing a computationally-predicted C-terminal degron. Thus, complementary to enhanced oncogene transcription via promoter swapping, our model of degron loss illustrates another general mechanism for recurrent fusion proteins in driving tumorigenesis.


Amino Acid Motifs/genetics , Carcinogenesis/genetics , Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Oncogenes/genetics , Animals , Carcinogenesis/metabolism , Cell Line, Tumor , Cells, Cultured , Computational Biology/methods , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Male , Mice, Knockout , Mice, Nude , Models, Genetic , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Oncogene Proteins, Fusion/metabolism , Proteolysis , Transplantation, Heterologous
12.
Nat Commun ; 12(1): 3444, 2021 06 08.
Article En | MEDLINE | ID: mdl-34103528

AKT is involved in a number of key cellular processes including cell proliferation, apoptosis and metabolism. Hyperactivation of AKT is associated with many pathological conditions, particularly cancers. Emerging evidence indicates that arginine methylation is involved in modulating AKT signaling pathway. However, whether and how arginine methylation directly regulates AKT kinase activity remain unknown. Here we report that protein arginine methyltransferase 5 (PRMT5), but not other PRMTs, promotes AKT activation by catalyzing symmetric dimethylation of AKT1 at arginine 391 (R391). Mechanistically, AKT1-R391 methylation cooperates with phosphatidylinositol 3,4,5 trisphosphate (PIP3) to relieve the pleckstrin homology (PH)-in conformation, leading to AKT1 membrane translocation and subsequent activation by phosphoinositide-dependent kinase-1 (PDK1) and the mechanistic target of rapamycin complex 2 (mTORC2). As a result, deficiency in AKT1-R391 methylation significantly suppresses AKT1 kinase activity and tumorigenesis. Lastly, we show that PRMT5 inhibitor synergizes with AKT inhibitor or chemotherapeutic drugs to enhance cell death. Altogether, our study suggests that R391 methylation is an important step for AKT activation and its oncogenic function.


Arginine/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Protein-Arginine N-Methyltransferases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antineoplastic Agents/pharmacology , Biocatalysis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Female , HEK293 Cells , Humans , Methylation/drug effects , Mice, Nude , Mutation/genetics , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Protein-Arginine N-Methyltransferases/deficiency , Proto-Oncogene Proteins c-akt/chemistry , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
13.
Int J Mol Sci ; 22(4)2021 Feb 11.
Article En | MEDLINE | ID: mdl-33670113

The mechanistic target of rapamycin (mTOR) is a master regulator of cell growth, proliferation, and metabolism by integrating various environmental inputs including growth factors, nutrients, and energy, among others. mTOR signaling has been demonstrated to control almost all fundamental cellular processes, such as nucleotide, protein and lipid synthesis, autophagy, and apoptosis. Over the past fifteen years, mapping the network of the mTOR pathway has dramatically advanced our understanding of its upstream and downstream signaling. Dysregulation of the mTOR pathway is frequently associated with a variety of human diseases, such as cancers, metabolic diseases, and cardiovascular and neurodegenerative disorders. Besides genetic alterations, aberrancies in post-translational modifications (PTMs) of the mTOR components are the major causes of the aberrant mTOR signaling in a number of pathologies. In this review, we summarize current understanding of PTMs-mediated regulation of mTOR signaling, and also update the progress on targeting the mTOR pathway and PTM-related enzymes for treatment of human diseases.


Cardiovascular Diseases/metabolism , Metabolic Diseases/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Protein Processing, Post-Translational , TOR Serine-Threonine Kinases/metabolism , Cardiovascular Diseases/genetics , Cell Proliferation , Humans , Metabolic Diseases/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics
14.
Nat Cell Biol ; 22(2): 246-256, 2020 02.
Article En | MEDLINE | ID: mdl-32015438

The Hippo and mammalian target of rapamycin complex 1 (mTORC1) pathways are the two predominant growth-control pathways that dictate proper organ development. We therefore explored potential crosstalk between these two functionally relevant pathways to coordinate their growth-control functions. We found that the LATS1 and LATS2 kinases, the core components of the Hippo pathway, phosphorylate S606 of Raptor, an essential component of mTORC1, to attenuate mTORC1 activation by impairing the interaction of Raptor with Rheb. The phosphomimetic Raptor-S606D knock-in mutant led to a reduction in cell size and proliferation. Compared with Raptor+/+ mice, RaptorD/D knock-in mice exhibited smaller livers and hearts, and a significant inhibition of elevation in mTORC1 signalling induced by Nf2 or Lats1 and Lats2 loss. Thus, our study reveals a direct link between the Hippo and mTORC1 pathways to fine-tune organ growth.


Gene Expression Regulation, Developmental , Mechanistic Target of Rapamycin Complex 1/genetics , Protein Serine-Threonine Kinases/genetics , Ras Homolog Enriched in Brain Protein/genetics , Regulatory-Associated Protein of mTOR/genetics , Tumor Suppressor Proteins/genetics , Animals , CRISPR-Cas Systems , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Gene Editing , HCT116 Cells , HEK293 Cells , HeLa Cells , Heterografts , Hippo Signaling Pathway , Humans , Liver/abnormalities , Liver/metabolism , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Neurofibromin 2/deficiency , Neurofibromin 2/genetics , Organ Size , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction , Tumor Suppressor Proteins/deficiency
15.
Cancer Discov ; 9(9): 1306-1323, 2019 09.
Article En | MEDLINE | ID: mdl-31217297

The function of PTEN in the cytoplasm largely depends on its lipid-phosphatase activity, though which it antagonizes the PI3K-AKT oncogenic pathway. However, molecular mechanisms underlying the role of PTEN in the nucleus remain largely elusive. Here, we report that DNA double-strand breaks (DSB) promote PTEN interaction with MDC1 upon ATM-dependent phosphorylation of T/S398-PTEN. Importantly, DNA DSBs enhance NSD2 (MMSET/WHSC1)-mediated dimethylation of PTEN at K349, which is recognized by the tudor domain of 53BP1 to recruit PTEN to DNA-damage sites, governing efficient repair of DSBs partly through dephosphorylation of γH2AX. Of note, inhibiting NSD2-mediated methylation of PTEN, either through expressing methylation-deficient PTEN mutants or through inhibiting NSD2, sensitizes cancer cells to combinatorial treatment with a PI3K inhibitor and DNA-damaging agents in both cell culture and in vivo xenograft models. Therefore, our study provides a novel molecular mechanism for PTEN regulation of DSB repair in a methylation- and protein phosphatase-dependent manner. SIGNIFICANCE: NSD2-mediated dimethylation of PTEN is recognized by the 53BP1 tudor domain to facilitate PTEN recruitment into DNA-damage sites, governing efficient repair of DNA DSBs. Importantly, inhibiting PTEN methylation sensitizes cancer cells to combinatorial treatment with a PI3K inhibitor combined with DNA-damaging agents in both cell culture and in vivo xenograft models.This article is highlighted in the In This Issue feature, p. 1143.


Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Neoplasms/genetics , PTEN Phosphohydrolase/metabolism , Repressor Proteins/metabolism , Animals , Binding Sites , Cell Line, Tumor , DNA Breaks, Double-Stranded , Female , HCT116 Cells , Humans , Methylation , Mice , NIH 3T3 Cells , Neoplasms/metabolism , PTEN Phosphohydrolase/chemistry , Phosphorylation , Tumor Suppressor p53-Binding Protein 1/metabolism
16.
Proc Natl Acad Sci U S A ; 116(19): 9423-9432, 2019 05 07.
Article En | MEDLINE | ID: mdl-31000600

The Hippo-YAP/TAZ signaling pathway plays a pivotal role in growth control during development and regeneration and its dysregulation is widely implicated in various cancers. To further understand the cellular and molecular mechanisms underlying Hippo signaling regulation, we have found that activities of core Hippo signaling components, large tumor suppressor (LATS) kinases and YAP/TAZ transcription factors, oscillate during mitotic cell cycle. We further identified that the anaphase-promoting complex/cyclosome (APC/C)Cdh1 E3 ubiquitin ligase complex, which plays a key role governing eukaryotic cell cycle progression, intrinsically regulates Hippo signaling activities. CDH1 recognizes LATS kinases to promote their degradation and, hence, YAP/TAZ regulation by LATS phosphorylation is under cell cycle control. As a result, YAP/TAZ activities peak in G1 phase. Furthermore, we show in Drosophila eye and wing development that Cdh1 is required in vivo to regulate the LATS homolog Warts with a conserved mechanism. Cdh1 reduction increased Warts levels, which resulted in reduction of the eye and wing sizes in a Yorkie dependent manner. Therefore, LATS degradation by APC/CCdh1 represents a previously unappreciated and evolutionarily conserved layer of Hippo signaling regulation.


Anaphase-Promoting Complex-Cyclosome/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cdh1 Proteins/metabolism , Drosophila Proteins/metabolism , G1 Phase/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Anaphase-Promoting Complex-Cyclosome/genetics , Animals , Antigens, CD/genetics , Cadherins/genetics , Cdh1 Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , HEK293 Cells , HeLa Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
17.
Nat Cell Biol ; 21(5): 662-663, 2019 05.
Article En | MEDLINE | ID: mdl-30783264

In the version of this Article originally published, the labels for Rictor and mTOR in the whole cell lysate (WCL) blots were swapped in Fig. 3b and the mTOR blot was placed upside down. Unprocessed blots of mTOR were also missing from Supplementary Fig. 9. The corrected Figs are shown below. In addition, control blots for the mTOR antibody (Cell Signalling Technology #2972) were also missing. These are now provided below, as Fig. 9, and show that the lower band is likely non-specific.

18.
Nat Cell Biol ; 21(2): 226-237, 2019 02.
Article En | MEDLINE | ID: mdl-30692625

Aberrant activation of AKT disturbs the proliferation, survival and metabolic homeostasis of various human cancers. Thus, it is critical to understand the upstream signalling pathways governing AKT activation. Here, we report that AKT undergoes SETDB1-mediated lysine methylation to promote its activation, which is antagonized by the Jumonji-family demethylase KDM4B. Notably, compared with wild-type mice, mice harbouring non-methylated mutant Akt1 not only exhibited reduced body size but were also less prone to carcinogen-induced skin tumours, in part due to reduced AKT activation. Mechanistically, the interaction of phosphatidylinositol (3,4,5)-trisphosphate with AKT facilitates its interaction with SETDB1 for subsequent AKT methylation, which in turn sustains AKT phosphorylation. Pathologically, genetic alterations, including SETDB1 amplification, aberrantly promote AKT methylation to facilitate its activation and oncogenic functions. Thus, AKT methylation is an important step, synergizing with PI3K signalling to control AKT activation. This suggests that targeting SETDB1 signalling could be a potential therapeutic strategy for combatting hyperactive AKT-driven cancers.


Carcinogenesis/metabolism , Protein Methyltransferases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Carcinogenesis/genetics , Cell Line, Tumor , Female , HEK293 Cells , Histone-Lysine N-Methyltransferase , Humans , Methylation , Mice, Nude , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Plicamycin/pharmacology , Protein Methyltransferases/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Sf9 Cells , Spodoptera , Xenograft Model Antitumor Assays/methods
19.
Dev Cell ; 48(3): 329-344.e5, 2019 02 11.
Article En | MEDLINE | ID: mdl-30595538

Frequent SPOP mutation defines the molecular feature underlying one of seven sub-types of human prostate cancer (PrCa). However, it remains largely elusive how SPOP functions as a tumor suppressor in PrCa. Here, we report that SPOP suppresses stem cell traits of both embryonic stem cells and PrCa cells through promoting Nanog poly-ubiquitination and subsequent degradation. Mechanistically, Nanog, but not other pluripotency-determining factors including Oct4, Sox2, and Klf4, specifically interacts with SPOP via a conservative degron motif. Importantly, cancer-derived mutations in SPOP or at the Nanog-degron (S68Y) disrupt SPOP-mediated destruction of Nanog, leading to elevated cancer stem cell traits and PrCa progression. Notably, we identify the Pin1 oncoprotein as an upstream Nanog regulator that impairs its recognition by SPOP and thereby stabilizes Nanog. Thus, Pin1 inhibitors promote SPOP-mediated destruction of Nanog, which provides the molecular insight and rationale to use Pin1 inhibitor(s) for targeted therapies of PrCa patients with wild-type SPOP.


Cell Proliferation/physiology , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Cullin Proteins/metabolism , Disease Progression , Humans , Kruppel-Like Factor 4 , Male , Mutation/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Interaction Domains and Motifs/genetics , Repressor Proteins/genetics , Ubiquitination
20.
Sci Signal ; 11(533)2018 06 05.
Article En | MEDLINE | ID: mdl-29871913

Polyubiquitylation is canonically viewed as a posttranslational modification that governs protein stability or protein-protein interactions, in which distinct polyubiquitin linkages ultimately determine the fate of modified protein(s). We explored whether polyubiquitin chains have any nonprotein-related function. Using in vitro pull-down assays with synthetic materials, we found that polyubiquitin chains with the Lys63 (K63) linkage bound to DNA through a motif we called the "DNA-interacting patch" (DIP), which is composed of the adjacent residues Thr9, Lys11, and Glu34 Upon DNA damage, the binding of K63-linked polyubiquitin chains to DNA enhanced the recruitment of repair factors through their interaction with an Ile44 patch in ubiquitin to facilitate DNA repair. Furthermore, experimental or cancer patient-derived mutations within the DIP impaired the DNA binding capacity of ubiquitin and subsequently attenuated K63-linked polyubiquitin chain accumulation at sites of DNA damage, thereby resulting in defective DNA repair and increased cellular sensitivity to DNA-damaging agents. Our results therefore highlight a critical physiological role for K63-linked polyubiquitin chains in binding to DNA to facilitate DNA damage repair.


DNA Damage , DNA Repair , DNA/metabolism , Lysine/metabolism , Neoplasms/metabolism , Polyubiquitin/metabolism , DNA/chemistry , Humans , Lysine/chemistry , Models, Molecular , Neoplasms/genetics , Neoplasms/pathology , Protein Processing, Post-Translational , Signal Transduction , Tumor Cells, Cultured , Ubiquitination
...