Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Epigenetics ; 18(1): 2195305, 2023 12.
Article En | MEDLINE | ID: mdl-36994860

Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease with an unclear pathogenesis. This study aimed to elucidate the function and potential mechanisms of TUG1 in IPF progression. Cell viability and migration were detected by CCK-8 and transwell assays. Autophagy, fibrosis, or EMT-related proteins were measured by Western blotting. Pro-inflammatory cytokine levels were assessed by ELISA kits. The subcellular localization of TUG1 was observed by FISH assay. RIP assay detected the interaction between TUG1 and CDC27. TUG1 and CDC27 was up-regulated in TGF-ß1-induced RLE-6TN cells. TUG1 depletion suppressed pulmonary fibrosis via attenuating inflammation, EMT, inducing autophagy and inactivating PI3K/Akt/mTOR pathway in vitro and in vivo. TUG1 knockdown prevented CDC27 expression. TUG1 silencing ameliorated pulmonary fibrosis by reducing CDC27 expression and inhibiting PI3K/Akt/mTOR pathway.


Pulmonary Fibrosis , RNA, Long Noncoding , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , DNA Methylation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Animals
2.
Pathol Res Pract ; 235: 153908, 2022 Jul.
Article En | MEDLINE | ID: mdl-35561648

BACKGROUND: Hepatic carcinoma (HC) is one of the leading causes of cancer-related death, and the incidence keeps high in the world. The vital role of circular RNAs (circRNAs) in HC development has been revealed to some extent. Circ_0000775, a novel circRNA, has never been thoroughly studied regarding HC. METHODS: Online datasets were utilized to obtain expression pattern of genes in tumor tissues. RT-qPCR and western blot examined the RNA and protein levels of indicated genes. ChIP, DNA pull down, RNA pull down, RIP and luciferase reporter assays were carried out to verify correlation between different factors. Supported by RT-qPCR and western blot analyses, transwell and wound healing assay were implemented for detecting cell migration and invasion and EMT. Additionally, cell EMT was also evaluated via cell morphology observation for calculation of spindle cell number. RESULTS: High expression of circ_0000775 in HC cells was induced by transcriptionally stimulation by TCF7L2. Circ_0000775 in cytoplasm recruited IGF2BP2 to enhance the mRNA stability of CDC27, thus positively modulating CDC27 expression. Circ_0000775 exacerbated HC cell migration, invasion and EMT through CDC27. CONCLUSION: TCF7L2 promoted the transcription of circ_0000775, and circ_0000775 recruited IGF2BP2 to maintain CDC27 mRNA stability, thereby facilitating HC cell migration, invasion and EMT.


Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome , Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , RNA, Circular , RNA-Binding Proteins , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplastic Processes , RNA, Circular/genetics , RNA, Circular/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
3.
Metab Brain Dis ; 37(4): 1015-1023, 2022 04.
Article En | MEDLINE | ID: mdl-35098413

Circular RNA circSLC8A1 is one of the cancer-related circRNAs that is implicated in various cancers. However, studies focusing on the role of circSLC8A1 in glioma is rare. Here we attempted to evaluate the biological function of circSLC8A1 in glioma and explore the potential mechanism. The relative expression of circSLC8A1, miR-214-5p and CDC27 in tissues and cell lines was determined by qRT-PCR. Cell proliferation and invasion were respectively measured by CCK-8 and transwell assays. Protein level of CDC27 was analyzed by western blot. Luciferase reporter assay was performed to confirm the regulatory interaction of cirRNA-miRNA-mRNA. Lowly expressed circSLC8A1 was observed in both glioma tissues and cell lines. Further biological analyses showed that circSLC8A1 inhibits the cell proliferation and invasion of glioma cells. CircSLC8A1 directly sponged miR-214-5p and inhibited miR-214-5p expression in glioma cells. CDC27 was a direct target of miR-214-5p and could be regulated by miR-214-5p. Moreover, miR-214-5p mimics and CDC27 knockdown reversed the inhibitory effects of circSLC8A1 on cell proliferation and invasion. Taken together, our results demonstrated a tumor suppressive role of circSLC8A1 in glioma through regulation of glioma cells proliferation and invasion. The effects of circSLC8A1 were mediated by miR-214-5p/CDC27 axis. Our study provided a new understanding of the occurrence and development of glioma.


Glioma , MicroRNAs , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics
4.
Cancer Lett ; 499: 109-121, 2021 02 28.
Article En | MEDLINE | ID: mdl-33259899

Hepatocellular carcinoma (HCC) is a malignancy found at high frequency around the world. Unfortunately, the scarcity of effective early diagnostic methods invariably results in poor outcomes. Long noncoding RNAs (lncRNAs) are known to regulate the progression of hepatocellular carcinoma (HCC). A novel lncRNA RP11-286H15.1(OTTHUMG00000186042) has been identified and associated with HCC; however, the potential role of RP11-286H15.1 in HCC remains undefined. The transcript abundance of RP11-286H15.1 in 80 pairs of HCC samples and cell lines was evaluated by qRT-PCR analysis. The functional role of RP11-286H15.1 in HCC was tested in vivo and in vitro. The mechanisms underlying the role of RP11-286H15.1 in HCC were explored by RNA pulldown, transcriptome sequencing, and RNA immunoprecipitation (RIP), ubiquitination and fluorescence in situ hybridization (FISH) assays as well as Western blot analysis. The qRT-PCR and FISH assays revealed that RP11-286H15.1 was significantly decreased in HCC, and implied a shorter survival time. RP11-286H15.1 overexpression inhibited HCC cell proliferation and metastasis in vitro and in vivo, whereas RP11-286H15.1 knockdown produced the opposite results. Furthermore, we confirmed that RP11-286H15.1 (620-750 nucleotides) binds to poly(A) binding protein 4 (PABPC4) and promotes its ubiquitination, thus, reducing the stability of TRIM37 and CDC27 mRNAs. Our study demonstrates that a novel lncRNA, RP11-286H15.1, represses HCC progression by promoting PABPC4 ubiquitination. These findings highlight potential therapeutic targets for HCC.


Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA, Long Noncoding/metabolism , Ubiquitination/genetics , Aged , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Liver/pathology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , RNA Stability/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism , RNA-Seq , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
PLoS Genet ; 16(11): e1009184, 2020 11.
Article En | MEDLINE | ID: mdl-33137813

In mitosis and meiosis, chromosome segregation is triggered by the Anaphase-Promoting Complex/Cyclosome (APC/C), a multi-subunit ubiquitin ligase that targets proteins for degradation, leading to the separation of chromatids. APC/C activation requires phosphorylation of its APC3 and APC1 subunits, which allows the APC/C to bind its co-activator Cdc20. The identity of the kinase(s) responsible for APC/C activation in vivo is unclear. Cyclin B3 (CycB3) is an activator of the Cyclin-Dependent Kinase 1 (Cdk1) that is required for meiotic anaphase in flies, worms and vertebrates. It has been hypothesized that CycB3-Cdk1 may be responsible for APC/C activation in meiosis but this remains to be determined. Using Drosophila, we found that mutations in CycB3 genetically enhance mutations in tws, which encodes the B55 regulatory subunit of Protein Phosphatase 2A (PP2A) known to promote mitotic exit. Females heterozygous for CycB3 and tws loss-of-function alleles lay embryos that arrest in mitotic metaphase in a maternal effect, indicating that CycB3 promotes anaphase in mitosis in addition to meiosis. This metaphase arrest is not due to the Spindle Assembly Checkpoint (SAC) because mutation of mad2 that inactivates the SAC does not rescue the development of embryos from CycB3-/+, tws-/+ females. Moreover, we found that CycB3 promotes APC/C activity and anaphase in cells in culture. We show that CycB3 physically associates with the APC/C, is required for phosphorylation of APC3, and promotes APC/C association with its Cdc20 co-activators Fizzy and Cortex. Our results strongly suggest that CycB3-Cdk1 directly activates the APC/C to promote anaphase in both meiosis and mitosis.


Anaphase-Promoting Complex-Cyclosome/metabolism , Anaphase/physiology , CDC2 Protein Kinase/metabolism , Cyclin B/metabolism , Drosophila Proteins/metabolism , Animals , Animals, Genetically Modified , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Cdc20 Proteins/metabolism , Cell Line , Chromosome Segregation/physiology , Cyclin B/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Loss of Function Mutation , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Male , Metaphase/physiology , Models, Animal , Mutagenesis , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphorylation
6.
Cell Physiol Biochem ; 50(2): 501-511, 2018.
Article En | MEDLINE | ID: mdl-30308498

BACKGROUND/AIMS: Lymph node metastasis is the primary cause of cancer-related death among patients with gastric cancer (GC), and cell division cycle 27 (CDC27) promotes the metastasis and epithelial-mesenchymal transition in many cancers. Till now, the mechanisms underlying CDC27-induced the epithelial-mesenchymal transition (EMT) of GC are still unclear. METHODS: We analyzed the expression levels of CDC27 and EMT-related biomarkers using immunohistochemistry and Western blot in 60 cases of GC tissues, and then GC cells with CDC27 shRNAs or plasmids were subjected to in vitro and in vivo assays, including CCK-8, wound healing and transwell assays. RESULTS: The CDC27 expression was obviously increased in GC tissues, and significantly correlates with EMT-related biomarkers, lymph node metastasis and poor 5-year overall survival. Additionally, in vitro and in vivo assays demonstrated that silencing of CDC27 expression effectively inhibited GC cell proliferation, invasion and metastasis. Conversely, CDC27 overexpression led to the opposite results. Finally, we demonstrated that Twist shRNA inhibited CDC27-meditated invasion and EMT of GC cells. CONCLUSION: CDC27 facilitates gastric cancer cell proliferation, invasion and metastasis via Twist-induced EMT; thus, this study offered a new therapy method for GC patients.


Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Nuclear Proteins/metabolism , Stomach Neoplasms/pathology , Twist-Related Protein 1/metabolism , Adult , Aged , Animals , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/antagonists & inhibitors , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Lymphatic Metastasis , Male , Mice , Mice, Nude , Middle Aged , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Survival Rate , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics
7.
Biochem Biophys Res Commun ; 485(4): 820-825, 2017 04 15.
Article En | MEDLINE | ID: mdl-28257844

Ubiquitin-conjugating enzyme E2S (UBE2S), a family of E2 protein in the ubiquitin-proteasome system, is highly expressed in several types of cancers; however, its roles in oral squamous cell carcinoma (OSCC) have not yet been well elucidated. The purpose of this study was to clarify the functional activities of UBE2S in OSCCs. We analyzed the expression levels of UBE2S in nine OSCC cell lines and primary OSCC tissues by quantitative reverse transcriptase-polymerase chain reaction, Western blotting, and immunohistochemistry (IHC). The correlations between UBE2S expression and clinical classifications of OSCCs were analyzed using the IHC scoring system. We also used UBE2S knockdown OSCC cells for functional assays (proliferation assay, flow cytometry, and Western blotting). UBE2S was overexpressed in OSCCs in vitro and in vivo and was correlated significantly (P < 0.05) with the primary tumoral size. The cellular growth was decreased and the cell-cycle was arrested in the G2/M phase in the UBE2S knockdown (shUBE2S) cells. The expression level of P21, a target of the ubiquitin-proteasome system, was increased in the shUBE2S cells because of lower anaphase activity that promotes complex subunit 3 (APC3), an E3 ubiquitin ligase, compared with shMock cells. These findings might promote the understanding of the relationship between UBE2S overexpression and oral cancer proliferation, indicating that UBE2S would be a potential biomarker of and therapeutic target in OSCCs.


Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Mouth Neoplasms/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Aged , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/genetics
8.
Cancer Discov ; 7(2): 218-233, 2017 02.
Article En | MEDLINE | ID: mdl-28069571

Intercellular heterogeneity, exacerbated by chromosomal instability (CIN), fosters tumor heterogeneity and drug resistance. However, extreme CIN correlates with improved cancer outcome, suggesting that karyotypic diversity required to adapt to selection pressures might be balanced in tumors against the risk of excessive instability. Here, we used a functional genomics screen, genome editing, and pharmacologic approaches to identify CIN-survival factors in diploid cells. We find partial anaphase-promoting complex/cyclosome (APC/C) dysfunction lengthens mitosis, suppresses pharmacologically induced chromosome segregation errors, and reduces naturally occurring lagging chromosomes in cancer cell lines or following tetraploidization. APC/C impairment caused adaptation to MPS1 inhibitors, revealing a likely resistance mechanism to therapies targeting the spindle assembly checkpoint. Finally, CRISPR-mediated introduction of cancer somatic mutations in the APC/C subunit cancer driver gene CDC27 reduces chromosome segregation errors, whereas reversal of an APC/C subunit nonsense mutation increases CIN. Subtle variations in mitotic duration, determined by APC/C activity, influence the extent of CIN, allowing cancer cells to dynamically optimize fitness during tumor evolution. SIGNIFICANCE: We report a mechanism whereby cancers balance the evolutionary advantages associated with CIN against the fitness costs caused by excessive genome instability, providing insight into the consequence of CDC27 APC/C subunit driver mutations in cancer. Lengthening of mitosis through APC/C modulation may be a common mechanism of resistance to cancer therapeutics that increase chromosome segregation errors. Cancer Discov; 7(2); 218-33. ©2017 AACR.See related commentary by Burkard and Weaver, p. 134This article is highlighted in the In This Issue feature, p. 115.


Anaphase-Promoting Complex-Cyclosome/metabolism , Chromosomal Instability , Gene Editing/methods , Genomics/methods , Neoplasms/genetics , Anaphase-Promoting Complex-Cyclosome/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , HCT116 Cells , HT29 Cells , Humans , Mitosis , Neoplasms/metabolism
9.
Oncotarget ; 8(4): 6304-6318, 2017 Jan 24.
Article En | MEDLINE | ID: mdl-27974673

Glioma has become a significant global health problem with substantial morbidity and mortality, underscoring the importance of elucidating its underlying molecular mechanisms. Recent studies have identified mir-218 as an anti-oncogene; however, the specific functions of mir-218-1 and mir-218-2 remain unknown, especially the latter. The objective of this study was to further investigate the role of mir-218-2 in glioma. Our results indicated that mir-218-2 is highly overexpressed in glioma. Furthermore, we showed that mir-218-2 is positively correlated with the growth, invasion, migration, and drug susceptibility (to ß-lapachone) of glioma cells. In vitro, the overexpression of mir-218-2 promoted glioma cell proliferation, invasion, and migration. In addition, the overexpression of mir-218-2 in vivo was found to increase glioma tumor growth. Accordingly, the inhibition of mir-218-2 resulted in the opposite effects. Cell division cycle 27 (CDC27), the downstream target of mir-218-2, is involved in the regulation of glioma cells. Our results indicate that the overexpression of CDC27 counteracted the function of mir-218-2 in glioma cells. These novel findings provide new insight in the application of mir-218-2 as a potential glioma treatment.


Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , MicroRNAs/metabolism , Naphthoquinones/pharmacology , Animals , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
10.
J Cell Biochem ; 118(10): 3150-3157, 2017 10.
Article En | MEDLINE | ID: mdl-28004426

The aim of this study was to investigate the possibility of APC/CCdh1 as a potential therapeutic target in the radiosensitivity of nasopharyngeal carcinoma (NPC) cell CNE-1, and explain the role of APC subunits after silence of Cdh1 combined with radiotherapy. Transfection with Cdh1 shRNA significantly increased the radiosensitivity of CNE-1 cells and the radiation enhancement ratio (RER) of sh-Cdh1 cells was 1.76. Knockdown of Cdh1 in CNE-1 cells increased irradiation induced apoptosis and G2/M phase cell cycle arrest. The levels of CDC20 and CylinB1 increased and the levels of Ku70 and APC3 decreased after irradiation. APC/CCdh1 is involved in regulation of radiosensitivity in human NPC CNE-1 cells. Our study may provide a promising therapeutic strategy for NPC by targeting Cdh1. J. Cell. Biochem. 118: 3150-3157, 2017. © 2016 Wiley Periodicals, Inc.


Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apoptosis , Cadherins/metabolism , Carcinoma/metabolism , Nasopharyngeal Neoplasms/metabolism , Radiation Tolerance , Antigens, CD , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Cadherins/genetics , Carcinoma/genetics , Carcinoma/radiotherapy , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/radiotherapy
11.
Nature ; 533(7602): 260-264, 2016 05 12.
Article En | MEDLINE | ID: mdl-27120157

In eukaryotes, the anaphase-promoting complex (APC/C, also known as the cyclosome) regulates the ubiquitin-dependent proteolysis of specific cell-cycle proteins to coordinate chromosome segregation in mitosis and entry into the G1 phase. The catalytic activity of the APC/C and its ability to specify the destruction of particular proteins at different phases of the cell cycle are controlled by its interaction with two structurally related coactivator subunits, Cdc20 and Cdh1. Coactivators recognize substrate degrons, and enhance the affinity of the APC/C for its cognate E2 (refs 4-6). During mitosis, cyclin-dependent kinase (Cdk) and polo-like kinase (Plk) control Cdc20- and Cdh1-mediated activation of the APC/C. Hyperphosphorylation of APC/C subunits, notably Apc1 and Apc3, is required for Cdc20 to activate the APC/C, whereas phosphorylation of Cdh1 prevents its association with the APC/C. Since both coactivators associate with the APC/C through their common C-box and Ile-Arg tail motifs, the mechanism underlying this differential regulation is unclear, as is the role of specific APC/C phosphorylation sites. Here, using cryo-electron microscopy and biochemical analysis, we define the molecular basis of how phosphorylation of human APC/C allows for its control by Cdc20. An auto-inhibitory segment of Apc1 acts as a molecular switch that in apo unphosphorylated APC/C interacts with the C-box binding site and obstructs engagement of Cdc20. Phosphorylation of the auto-inhibitory segment displaces it from the C-box-binding site. Efficient phosphorylation of the auto-inhibitory segment, and thus relief of auto-inhibition, requires the recruitment of Cdk-cyclin in complex with a Cdk regulatory subunit (Cks) to a hyperphosphorylated loop of Apc3. We also find that the small-molecule inhibitor, tosyl-l-arginine methyl ester, preferentially suppresses APC/C(Cdc20) rather than APC/C(Cdh1), and interacts with the binding sites of both the C-box and Ile-Arg tail motifs. Our results reveal the mechanism for the regulation of mitotic APC/C by phosphorylation and provide a rationale for the development of selective inhibitors of this state.


Anaphase-Promoting Complex-Cyclosome/metabolism , Mitosis , Phosphoproteins/metabolism , Amino Acid Motifs , Anaphase-Promoting Complex-Cyclosome/chemistry , Anaphase-Promoting Complex-Cyclosome/ultrastructure , Antigens, CD , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apoenzymes/metabolism , Binding Sites , Cadherins/chemistry , Cadherins/metabolism , Cadherins/ultrastructure , Cdc20 Proteins/antagonists & inhibitors , Cdc20 Proteins/chemistry , Cdc20 Proteins/metabolism , Cdc20 Proteins/ultrastructure , Cryoelectron Microscopy , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Enzyme Activation , Humans , Models, Molecular , Phosphoproteins/chemistry , Phosphoproteins/ultrastructure , Phosphorylation , Protein Binding , Protein Conformation , Tosylarginine Methyl Ester/pharmacology
12.
Science ; 352(6289): 1121-4, 2016 May 27.
Article En | MEDLINE | ID: mdl-27103671

Error-free genome duplication and segregation are ensured through the timely activation of ubiquitylation enzymes. The anaphase-promoting complex or cyclosome (APC/C), a multisubunit E3 ubiquitin ligase, is regulated by phosphorylation. However, the mechanism remains elusive. Using systematic reconstitution and analysis of vertebrate APC/Cs under physiological conditions, we show how cyclin-dependent kinase 1 (CDK1) activates the APC/C through coordinated phosphorylation between Apc3 and Apc1. Phosphorylation of the loop domains by CDK1 in complex with p9/Cks2 (a CDK regulatory subunit) controlled loading of coactivator Cdc20 onto APC/C. A phosphomimetic mutation introduced into Apc1 allowed Cdc20 to increase APC/C activity in interphase. These results define a previously unrecognized subunit-subunit communication over a distance and the functional consequences of CDK phosphorylation. Cdc20 is a potential therapeutic target, and our findings may facilitate the development of specific inhibitors.


Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Cdh1 Proteins/metabolism , Anaphase , Animals , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Cdc20 Proteins/metabolism , Enzyme Activation , Humans , Mutation , Phosphorylation , Xenopus
13.
Biochem Biophys Res Commun ; 471(4): 497-502, 2016 Mar 18.
Article En | MEDLINE | ID: mdl-26882976

Elmo has no intrinsic catalytic activity but coordinate multiple cellular processes via their interactions with other proteins. Studies thus have been focused on identifying Elmo binding partners, but the number of characterized Elmo-interacting proteins remains limited. Here, we report Cdc27 as a novel Elmo1-interacting protein. In yeast and mammalian cells, Cdc27 specifically interacted with the C-terminal region of Elmo1 essential for Dock1 association and function. The interaction of Elmo1 with Dock1 abrogated binding between Elmo1 and Cdc27, but the Dock1-Elmo1 interaction was unaffected by Cdc27. Similarly, cellular phagocytotic functions mediated by the Elmo1-Dock1-Rac module were unaffected by Cdc27 levels. In summary, a novel binding partner, Cdc27, was identified for Elmo1 and they appear to be independent of Elmo-Dock1-Rac-mediated processes.


Adaptor Proteins, Signal Transducing/metabolism , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Binding Sites , HEK293 Cells/metabolism , Humans , Phagocytosis/physiology , Protein Interaction Mapping/methods , rac GTP-Binding Proteins/metabolism
14.
Cell Death Dis ; 7: e2074, 2016 Jan 28.
Article En | MEDLINE | ID: mdl-26821069

Dysregulated cell cycle progression has a critical role in tumorigenesis. Cell division cycle 27 (CDC27) is a core subunit of the anaphase-promoting complex/cyclosome, although the specific role of CDC27 in cancer remains unknown. In our study, we explored the biological and clinical significance of CDC27 in colorectal cancer (CRC) growth and progression and investigated the underlying molecular mechanisms. Results showed that CDC27 expression is significantly correlated with tumor progression and poor patient survival. Functional assays demonstrated that overexpression of CDC27 promoted proliferation in DLD1 cells, whereas knockdown of CDC27 in HCT116 cells inhibited proliferation both in vitro and in vivo. Further mechanistic investigation showed that CDC27 downregulation resulted in G1/S phase transition arrest via the significant accumulation of p21 in HCT116 cells, and the upregulation of CDC27 promoted G1/S phase transition via the attenuation of p21 in DLD1 cells. Furthermore, we also demonstrated that CDC27 regulated inhibitor of DNA binding 1 (ID1) protein expression in DLD1 and HCT116 cells, and rescue assays revealed that CDC27 regulated p21 expression through modulating ID1 expression. Taken together, our results indicate that CDC27 contributes to CRC cell proliferation via the modulation of ID1-mediated p21 regulation, which offers a novel approach to the inhibition of tumor growth. Indeed, these findings provide new perspectives for the future study of CDC27 as a target for CRC treatment.


Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Animals , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apoptosis/physiology , Cell Proliferation/physiology , Colorectal Neoplasms/genetics , Disease Progression , Down-Regulation , HCT116 Cells , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Transfection
15.
Nature ; 522(7557): 450-454, 2015 Jun 25.
Article En | MEDLINE | ID: mdl-26083744

The anaphase-promoting complex (APC/C) is a multimeric RING E3 ubiquitin ligase that controls chromosome segregation and mitotic exit. Its regulation by coactivator subunits, phosphorylation, the mitotic checkpoint complex and interphase early mitotic inhibitor 1 (Emi1) ensures the correct order and timing of distinct cell-cycle transitions. Here we use cryo-electron microscopy to determine atomic structures of APC/C-coactivator complexes with either Emi1 or a UbcH10-ubiquitin conjugate. These structures define the architecture of all APC/C subunits, the position of the catalytic module and explain how Emi1 mediates inhibition of the two E2s UbcH10 and Ube2S. Definition of Cdh1 interactions with the APC/C indicates how they are antagonized by Cdh1 phosphorylation. The structure of the APC/C with UbcH10-ubiquitin reveals insights into the initiating ubiquitination reaction. Our results provide a quantitative framework for the design of future experiments to investigate APC/C functions in vivo.


Anaphase-Promoting Complex-Cyclosome/metabolism , Anaphase-Promoting Complex-Cyclosome/ultrastructure , Ubiquitination , Anaphase-Promoting Complex-Cyclosome/chemistry , Antigens, CD , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/ultrastructure , Apc10 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc10 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc10 Subunit, Anaphase-Promoting Complex-Cyclosome/ultrastructure , Apc11 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc11 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc8 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc8 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc8 Subunit, Anaphase-Promoting Complex-Cyclosome/ultrastructure , Cadherins/chemistry , Cadherins/metabolism , Cadherins/ultrastructure , Catalytic Domain , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/ultrastructure , Cryoelectron Microscopy , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , F-Box Proteins/chemistry , F-Box Proteins/metabolism , F-Box Proteins/ultrastructure , Humans , Lysine/metabolism , Models, Molecular , Phosphorylation , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism , Structure-Activity Relationship , Substrate Specificity , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitin/ultrastructure , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/ultrastructure
16.
J Mol Biol ; 427(8): 1748-64, 2015 Apr 24.
Article En | MEDLINE | ID: mdl-25490258

The anaphase-promoting complex/cyclosome (APC/C) is a massive E3 ligase that controls mitosis by catalyzing ubiquitination of key cell cycle regulatory proteins. The APC/C assembly contains two subcomplexes: the "Platform" centers around a cullin-RING-like E3 ligase catalytic core; the "Arc Lamp" is a hub that mediates transient association with regulators and ubiquitination substrates. The Arc Lamp contains the small subunits APC16, CDC26, and APC13, and tetratricopeptide repeat (TPR) proteins (APC7, APC3, APC6, and APC8) that homodimerize and stack with quasi-2-fold symmetry. Within the APC/C complex, APC3 serves as center for regulation. APC3's TPR motifs recruit substrate-binding coactivators, CDC20 and CDH1, via their C-terminal conserved Ile-Arg (IR) tail sequences. Human APC3 also binds APC16 and APC7 and contains a >200-residue loop that is heavily phosphorylated during mitosis, although the basis for APC3 interactions and whether loop phosphorylation is required for ubiquitination are unclear. Here, we map the basis for human APC3 assembly with APC16 and APC7, report crystal structures of APC3Δloop alone and in complex with the C-terminal domain of APC16, and test roles of APC3's loop and IR tail binding surfaces in APC/C-catalyzed ubiquitination. The structures show how one APC16 binds asymmetrically to the symmetric APC3 dimer and, together with biochemistry and prior data, explain how APC16 recruits APC7 to APC3, show how APC3's C-terminal domain is rearranged in the full APC/C assembly, and visualize residues in the IR tail binding cleft important for coactivator-dependent ubiquitination. Overall, the results provide insights into assembly, regulation, and interactions of TPR proteins and the APC/C.


Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Proteins/chemistry , Proteins/metabolism , Amino Acid Sequence , Apc7 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc7 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Cycle , Cell Cycle Proteins , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , Protein Interaction Maps , Protein Multimerization , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
17.
Cell Res ; 24(6): 701-12, 2014 Jun.
Article En | MEDLINE | ID: mdl-24699064

Single-cell sequencing is a powerful tool for delineating clonal relationship and identifying key driver genes for personalized cancer management. Here we performed single-cell sequencing analysis of a case of colon cancer. Population genetics analyses identified two independent clones in tumor cell population. The major tumor clone harbored APC and TP53 mutations as early oncogenic events, whereas the minor clone contained preponderant CDC27 and PABPC1 mutations. The absence of APC and TP53 mutations in the minor clone supports that these two clones were derived from two cellular origins. Examination of somatic mutation allele frequency spectra of additional 21 whole-tissue exome-sequenced cases revealed the heterogeneity of clonal origins in colon cancer. Next, we identified a mutated gene SLC12A5 that showed a high frequency of mutation at the single-cell level but exhibited low prevalence at the population level. Functional characterization of mutant SLC12A5 revealed its potential oncogenic effect in colon cancer. Our study provides the first exome-wide evidence at single-cell level supporting that colon cancer could be of a biclonal origin, and suggests that low-prevalence mutations in a cohort may also play important protumorigenic roles at the individual level.


High-Throughput Nucleotide Sequencing , Symporters/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Genotype , Humans , Male , Middle Aged , Mutation , Oncogenes , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , Principal Component Analysis , Sequence Analysis, DNA , Symporters/chemistry , Symporters/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , K Cl- Cotransporters
18.
Biochemistry ; 52(43): 7618-27, 2013 Oct 29.
Article En | MEDLINE | ID: mdl-24073851

The breast and ovarian cancer susceptibility protein 1 (BRCA1) plays a central role in DNA damage response (DDR). Two tandem BRCA1 C-terminal (BRCT) domains interact with several proteins that function in DDR and contain the generally accepted motif pS-X-X-F (pS denoting phosphoserine and X any amino acid), including the ATR-interacting protein (ATRIP) and the BRCA1-associated protein required for ATM activation-1 (BAAT1). The crystal structures of the BRCA1 BRCTs bound to the phosphopeptides ATRIP (235-PEACpSPQFG-243) and BAAT1 (266-VARpSPVFSS-274) were determined at 1.75 Å and 2.2 Å resolution, respectively. The pSer and Phe(+3) anchor the phosphopeptides into the BRCT binding groove, with adjacent peptide residues contributing to the interaction. In the BRCA1-ATRIP structure, Gln(+2) is accommodated through a conformational change of the BRCA1 E1698 side chain. Importantly, isothermal titration calorimetry experiments showed that the size and charge of the side chains at peptide positions +1 and +2 contribute significantly to the BRCA1 BRCT-peptide binding affinity. In particular, the Asp(+1) and Glu(+2) in the human CDC27 peptide 816-HAAEpSDEF-823 abrogate the interaction with the BRCA1 BRCTs due in large part to electrostatic repulsion between Glu(+2) and E1698, indicating a preference of these domains for specific side chains at positions +1 and +2. These results emphasize the need for a systematic assessment of the contribution of the peptide residues surrounding pSer and Phe(+3) to the binding affinity and specificity of the BRCA1 BRCTs in order to elucidate the molecular mechanisms underlying the hierarchy of target selection by these versatile domains during DDR and tumorigenesis.


Adaptor Proteins, Signal Transducing/chemistry , BRCA1 Protein/chemistry , DNA-Binding Proteins/chemistry , Models, Molecular , Nuclear Proteins/chemistry , Peptide Fragments/chemistry , Phosphopeptides/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Aspartic Acid/chemistry , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Binding Sites , DNA Repair , DNA-Binding Proteins/metabolism , Humans , Kinetics , Ligands , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Phenylalanine/chemistry , Phosphopeptides/metabolism , Phosphoserine/chemistry , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , X-Ray Diffraction
19.
J Mol Biol ; 425(22): 4236-48, 2013 Nov 15.
Article En | MEDLINE | ID: mdl-23583778

The anaphase-promoting complex or cyclosome (APC/C) is a large E3 RING-cullin ubiquitin ligase composed of between 14 and 15 individual proteins. A striking feature of the APC/C is that only four proteins are involved in directly recognizing target proteins and catalyzing the assembly of a polyubiquitin chain. All other subunits, which account for >80% of the mass of the APC/C, provide scaffolding functions. A major proportion of these scaffolding subunits are structurally related. In metazoans, there are four canonical tetratricopeptide repeat (TPR) proteins that form homo-dimers (Apc3/Cdc27, Apc6/Cdc16, Apc7 and Apc8/Cdc23). Here, we describe the crystal structure of the N-terminal homo-dimerization domain of Schizosaccharomyces pombe Cdc23 (Cdc23(Nterm)). Cdc23(Nterm) is composed of seven contiguous TPR motifs that self-associate through a related mechanism to those of Cdc16 and Cdc27. Using the Cdc23(Nterm) structure, we generated a model of full-length Cdc23. The resultant "V"-shaped molecule docks into the Cdc23-assigned density of the human APC/C structure determined using negative stain electron microscopy (EM). Based on sequence conservation, we propose that Apc7 forms a homo-dimeric structure equivalent to those of Cdc16, Cdc23 and Cdc27. The model is consistent with the Apc7-assigned density of the human APC/C EM structure. The four canonical homo-dimeric TPR proteins of human APC/C stack in parallel on one side of the complex. Remarkably, the uniform relative packing of neighboring TPR proteins generates a novel left-handed suprahelical TPR assembly. This finding has implications for understanding the assembly of other TPR-containing multimeric complexes.


Anaphase-Promoting Complex-Cyclosome/chemistry , Protein Multimerization , Protein Subunits , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome/ultrastructure , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc6 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc6 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc7 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Cell Cycle Proteins/chemistry , Humans , Minichromosome Maintenance Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/chemistry , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Sequence Alignment
...