Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Genes Dev ; 35(3-4): 261-272, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33446573

ABSTRACT

SUMO modification regulates diverse cellular processes by targeting hundreds of proteins. However, the limited number of sumoylation enzymes raises the question of how such a large number of substrates are efficiently modified. Specifically, how genome maintenance factors are dynamically sumoylated at DNA replication and repair sites to modulate their functions is poorly understood. Here, we demonstrate a role for the conserved yeast Esc2 protein in this process by acting as a SUMO E2 cofactor. Esc2 is required for genome stability and binds to Holliday junctions and replication fork structures. Our targeted screen found that Esc2 promotes the sumoylation of a Holliday junction dissolution complex and specific replisome proteins. Esc2 does not elicit these effects via stable interactions with substrates or their common SUMO E3. Rather, we show that a SUMO-like domain of Esc2 stimulates sumoylation by exploiting a noncovalent SUMO binding site on the E2 enzyme. This role of Esc2 in sumoylation is required for Holliday junction clearance and genome stability. Our findings thus suggest that Esc2 acts as a SUMO E2 cofactor at distinct DNA structures to promote the sumoylation of specific substrates and genome maintenance.


Subject(s)
Cell Cycle Proteins/metabolism , Genome, Fungal/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sumoylation/genetics , Coenzymes/metabolism , Genomic Instability/genetics , Protein Binding , Recombination, Genetic , Ubiquitin-Conjugating Enzymes/metabolism
2.
Mol Cell ; 75(2): 238-251.e5, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31348879

ABSTRACT

BRCT domains support myriad protein-protein interactions involved in genome maintenance. Although di-BRCT recognition of phospho-proteins is well known to support the genotoxic response, whether multi-BRCT domains can acquire distinct structures and functions is unclear. Here we present the tetra-BRCT structures from the conserved yeast protein Rtt107 in free and ligand-bound forms. The four BRCT repeats fold into a tetrahedral structure that recognizes unmodified ligands using a bi-partite mechanism, suggesting repeat origami enabling function acquisition. Functional studies show that Rtt107 binding of partner proteins of diverse activities promotes genome replication and stability in both distinct and concerted manners. A unified theme is that tetra- and di-BRCT domains of Rtt107 collaborate to recruit partner proteins to chromatin. Our work thus illustrates how a master regulator uses two types of BRCT domains to recognize distinct genome factors and direct them to chromatin for constitutive genome protection.


Subject(s)
Genomic Instability/genetics , Nuclear Proteins/ultrastructure , Protein Interaction Domains and Motifs/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Saccharomyces cerevisiae/genetics , Chromatin/genetics , DNA Damage/genetics , Ligands , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphorylation , Protein Binding , Protein Domains/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
3.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Article in English | MEDLINE | ID: mdl-33941673

ABSTRACT

Structural maintenance of chromosomes (SMC) complexes are critical chromatin modulators. In eukaryotes, the cohesin and condensin SMC complexes organize chromatin, while the Smc5/6 complex directly regulates DNA replication and repair. The molecular basis for the distinct functions of Smc5/6 is poorly understood. Here, we report an integrative structural study of the budding yeast Smc5/6 holo-complex using electron microscopy, cross-linking mass spectrometry, and computational modeling. We show that the Smc5/6 complex possesses several unique features, while sharing some architectural characteristics with other SMC complexes. In contrast to arm-folded structures of cohesin and condensin, Smc5 and Smc6 arm regions do not fold back on themselves. Instead, these long filamentous regions interact with subunits uniquely acquired by the Smc5/6 complex, namely the Nse2 SUMO ligase and the Nse5/Nse6 subcomplex, with the latter also serving as a linchpin connecting distal parts of the complex. Our 3.0-Å resolution cryoelectron microscopy structure of the Nse5/Nse6 core further reveals a clasped-hand topology and a dimeric interface important for cell growth. Finally, we provide evidence that Nse5/Nse6 uses its SUMO-binding motifs to contribute to Nse2-mediated sumoylation. Collectively, our integrative study identifies distinct structural features of the Smc5/6 complex and functional cooperation among its coevolved unique subunits.


Subject(s)
Cell Cycle Proteins/chemistry , Multiprotein Complexes/chemistry , Protein Domains , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Binding Sites , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Cryoelectron Microscopy/methods , Mass Spectrometry/methods , Models, Molecular , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Binding , Saccharomyces cerevisiae Proteins/metabolism , Sumoylation
4.
Mikrochim Acta ; 191(6): 340, 2024 05 24.
Article in English | MEDLINE | ID: mdl-38787447

ABSTRACT

A new sandwich-type electrochemical biosensing platform was developed by gold @polyphthalenediamine nanohybrids (AuNP@PoPD) as the sensing platform and phosphorus doped reduced graphene oxide-hemin-palladium nanoparticles (PrGO-Hemin-PdNP) as the signal amplifier for phosphatidylinositol proteoglycan 3 (GPC3). AuNP@PoPD, co-electrodeposited into the screen printed electrode with high conductivity and stability, is dedicated to assembling the primary GPC3 aptamer (GPC3Apt). The second GPC3Apt immobilized on the high conductivity and large surface area of PrGO-Hemin-PdNP was utilized as an electrochemical signal reporter by hemin oxidation (PrGO-Hemin-PdNP-GPC3Apt). In the range 0.001-10.0 ng/mL, the hemin oxidation current signal of the electrochemical aptasensor increased log-linearly with the concentration of GPC3, the lowest detection limit was 0.13 pg/mL, and the sensitivity was 2.073 µA/µM/cm2. The aptasensor exhibited good sensing performance in a human serum sample with the relative error of 4.31-8.07%. The sandwich sensor showed good selectivity and stability for detection GPC3 in human serum samples, providing a new efficient and sensitive method for detecting HCC markers.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Electrochemical Techniques , Glypicans , Gold , Graphite , Hemin , Limit of Detection , Metal Nanoparticles , Palladium , Glypicans/blood , Humans , Electrochemical Techniques/methods , Electrochemical Techniques/instrumentation , Aptamers, Nucleotide/chemistry , Hemin/chemistry , Graphite/chemistry , Palladium/chemistry , Gold/chemistry , Biosensing Techniques/methods , Metal Nanoparticles/chemistry , Electrodes
5.
Int J Mol Sci ; 24(3)2023 Jan 29.
Article in English | MEDLINE | ID: mdl-36768882

ABSTRACT

Gametogenesis is an essential step for malaria parasite transmission and is activated in mosquito by signals including temperature drop, pH change, and mosquito-derived xanthurenic acid (XA). Recently, a membrane protein gametogenesis essential protein 1 (GEP1) was found to be responsible for sensing these signals and interacting with a giant guanylate cyclase α (GCα) to activate the cGMP-PKG-Ca2+ signaling pathway for malaria parasite gametogenesis. However, the molecular mechanisms for this process remain unclear. In this study, we used AlphaFold2 to predict the structure of GEP1 and found that it consists of a conserved N-terminal helical domain and a transmembrane domain that adopts a structure similar to that of cationic amino acid transporters. Molecular docking results showed that XA binds to GEP1 via a pocket similar to the ligand binding sites of known amino acid transporters. In addition, truncations of this N-terminal sequence significantly enhanced the expression, solubility, and stability of GEP1. In addition, we found that GEP1 interacts with GCα via its C-terminal region, which is interrupted by mutations of a few conserved residues. These findings provide further insights into the molecular mechanism for the XA recognition by GEP1 and the activation of the gametogenesis of malaria parasites through GEP1-GCα interaction.


Subject(s)
Malaria , Parasites , Animals , Guanylate Cyclase/metabolism , Parasites/metabolism , Molecular Docking Simulation , Signal Transduction , Gametogenesis , Cyclic GMP/metabolism , Malaria/parasitology
6.
J Clin Biochem Nutr ; 72(2): 139-146, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36936869

ABSTRACT

M2-type polarization of tumor associated-macrophage (TAM) is involved in the malignancy of gastrointestinal stromal tumor (GIST) progression. ETS variant 1 (ETV1) has been previously validated to regulate GIST pathogenesis. Our study intended to explore the role and mechanism of ETV1 in mediating the M2-polarization of TAM in GIST progression. First, we analyzed the correlation between ETV1 expression and M2-polarization in GIST tissues. IL-4 was used to treat THP-1-derived TAM cells and IL-4-stimulated TAM were co-cultured with GIST-T1 cells to mimic the GIST microenvironment. A loss-of-function assay was performed to explore the role of ETV1. Results showed that ETV1 elevation was positively correlated with M2-polarization. IL-4-induced TAM promoted ETV1 expression, silencing ETV1 inhibited proliferation, invasion and KIT activation in IL-4-treated GIST cells, while cell apoptosis was enhanced. Besides, co-culture of ETV1-silenced GIST cells significantly depressed M2-polarization in TAM, presented as decreased levels of CD206, Agr-1 and cytokines, as well as the proportion of CD206-positive TAM. PDE3A was positively correlated with ETV1 and M2-polarization. Overexpressing PDE3A reversed the inhibitory effects of ETV1 silencing. Generally, ETV1 inhibition depressed M2-polarization of TAM in GIST and its promotion on pathological aggravation via down-regulating PDE3A. This evidence may provide a new target for GIST regulation.

7.
Nature ; 482(7386): 542-6, 2012 Feb 12.
Article in English | MEDLINE | ID: mdl-22327296

ABSTRACT

Menin is a tumour suppressor protein whose loss or inactivation causes multiple endocrine neoplasia 1 (MEN1), a hereditary autosomal dominant tumour syndrome that is characterized by tumorigenesis in multiple endocrine organs. Menin interacts with many proteins and is involved in a variety of cellular processes. Menin binds the JUN family transcription factor JUND and inhibits its transcriptional activity. Several MEN1 missense mutations disrupt the menin-JUND interaction, suggesting a correlation between the tumour-suppressor function of menin and its suppression of JUND-activated transcription. Menin also interacts with mixed lineage leukaemia protein 1 (MLL1), a histone H3 lysine 4 methyltransferase, and functions as an oncogenic cofactor to upregulate gene transcription and promote MLL1-fusion-protein-induced leukaemogenesis. A recent report on the tethering of MLL1 to chromatin binding factor lens epithelium-derived growth factor (LEDGF) by menin indicates that menin is a molecular adaptor coordinating the functions of multiple proteins. Despite its importance, how menin interacts with many distinct partners and regulates their functions remains poorly understood. Here we present the crystal structures of human menin in its free form and in complexes with MLL1 or with JUND, or with an MLL1-LEDGF heterodimer. These structures show that menin contains a deep pocket that binds short peptides of MLL1 or JUND in the same manner, but that it can have opposite effects on transcription. The menin-JUND interaction blocks JUN N-terminal kinase (JNK)-mediated JUND phosphorylation and suppresses JUND-induced transcription. In contrast, menin promotes gene transcription by binding the transcription activator MLL1 through the peptide pocket while still interacting with the chromatin-anchoring protein LEDGF at a distinct surface formed by both menin and MLL1.


Subject(s)
Myeloid-Lymphoid Leukemia Protein/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Chromatin/metabolism , Crystallography, X-Ray , Fibroblasts , HEK293 Cells , Histone-Lysine N-Methyltransferase , Humans , Intercellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein/chemistry , Phosphorylation , Protein Binding , Protein Multimerization , Proto-Oncogene Proteins c-jun/chemistry , Structure-Activity Relationship
8.
Nucleic Acids Res ; 44(10): 4871-80, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27131364

ABSTRACT

The Fanconi anemia protein SLX4 assembles a genome and telomere maintenance toolkit, consisting of the nucleases SLX1, MUS81 and XPF. Although it is known that SLX4 acts as a scaffold for building this complex, the molecular basis underlying this function of SLX4 remains unclear. Here, we report that functioning of SLX4 is dependent on its dimerization via an oligomerization motif called the BTB domain. We solved the crystal structure of the SLX4BTB dimer, identifying key contacts (F681 and F708) that mediate dimerization. Disruption of BTB dimerization abrogates nuclear foci formation and telomeric localization of not only SLX4 but also of its associated nucleases. Furthermore, dimerization-deficient SLX4 mutants cause defective cellular response to DNA interstrand crosslinking agent and telomere maintenance, underscoring the contribution of BTB domain-mediated dimerization of SLX4 in genome and telomere maintenance.


Subject(s)
Endonucleases/metabolism , Recombinases/chemistry , Cell Line , Hydrophobic and Hydrophilic Interactions , Mitomycin/toxicity , Protein Domains , Protein Multimerization , Recombinases/metabolism , Telomere/enzymology , Telomere/ultrastructure
9.
Nucleic Acids Res ; 43(12): 5912-23, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-25990736

ABSTRACT

SLX4 assembles a toolkit of endonucleases SLX1, MUS81 and XPF, which is recruited to telomeres via direct interaction of SLX4 with TRF2. Telomeres present an inherent obstacle for DNA replication and repair due to their high propensity to form branched DNA intermediates. Here we provide novel insight into the mechanism and regulation of the SLX4 complex in telomere preservation. SLX4 associates with telomeres throughout the cell cycle, peaking in late S phase and under genotoxic stress. Disruption of SLX4's interaction with TRF2 or SLX1 and SLX1's nuclease activity independently causes telomere fragility, suggesting a requirement of the SLX4 complex for nucleolytic resolution of branched intermediates during telomere replication. Indeed, the SLX1-SLX4 complex processes a variety of telomeric joint molecules in vitro. The nucleolytic activity of SLX1-SLX4 is negatively regulated by telomeric DNA-binding proteins TRF1 and TRF2 and is suppressed by the RecQ helicase BLM in vitro. In vivo, in the presence of functional BLM, telomeric circle formation and telomere sister chromatid exchange, both arising out of nucleolytic processing of telomeric homologous recombination intermediates, are suppressed. We propose that the SLX4-toolkit is a telomere accessory complex that, in conjunction with other telomere maintenance proteins, ensures unhindered, but regulated telomere maintenance.


Subject(s)
Recombinases/metabolism , Telomere/metabolism , Cell Cycle , DNA/metabolism , Endodeoxyribonucleases , Endonucleases/metabolism , HeLa Cells , Homologous Recombination , Humans , RecQ Helicases/metabolism , Sister Chromatid Exchange , Telomere-Binding Proteins/metabolism
10.
J Biol Chem ; 289(29): 19928-41, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24895130

ABSTRACT

Understanding how cellular machinery deals with chromosomal genome complexity is an important question because protein bound to DNA may affect various cellular processes of nucleic acid metabolism. DNA helicases are at the forefront of such processes, yet there is only limited knowledge how they remodel protein-DNA complexes and how these mechanisms are regulated. We have determined that representative human RecQ and Fe-S cluster DNA helicases are potently blocked by a protein-DNA interaction. The Fanconi anemia group J (FANCJ) helicase partners with the single-stranded DNA-binding protein replication protein A (RPA) to displace BamHI-E111A bound to duplex DNA in a specific manner. Protein displacement was dependent on the ATPase-driven function of the helicase and unique properties of RPA. Further biochemical studies demonstrated that the shelterin proteins TRF1 and TRF2, which preferentially bind the telomeric repeat found at chromosome ends, effectively block FANCJ from unwinding the forked duplex telomeric substrate. RPA, but not the Escherichia coli single-stranded DNA-binding protein or shelterin factor Pot1, stimulated FANCJ ejection of TRF1 from the telomeric DNA substrate. FANCJ was also able to displace TRF2 from the telomeric substrate in an RPA-dependent manner. The stimulation of helicase-catalyzed protein displacement is also observed with the DNA helicase RECQ1, suggesting a conserved functional interaction of RPA-interacting helicases. These findings suggest that partnerships between RPA and interacting human DNA helicases may greatly enhance their ability to dislodge proteins bound to duplex DNA, an activity that is likely to be highly relevant to their biological roles in DNA metabolism.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , DNA/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , RecQ Helicases/metabolism , Replication Protein A/metabolism , Amino Acid Substitution , Base Sequence , DNA/chemistry , DNA/genetics , Deoxyribonuclease BamHI/metabolism , Exodeoxyribonucleases/metabolism , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Nucleic Acid Conformation , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Protein A/genetics , Substrate Specificity , Telomeric Repeat Binding Protein 1/metabolism , Werner Syndrome Helicase
11.
Res Sq ; 2024 May 24.
Article in English | MEDLINE | ID: mdl-38826278

ABSTRACT

The DNA damage checkpoint is a highly conserved signaling pathway induced by genotoxin exposure or endogenous genome stress. It alters many cellular processes such as arresting the cell cycle progression and increasing DNA repair capacities. However, cells can downregulate the checkpoint after prolonged stress exposure to allow continued growth and alternative repair. Strategies that can dampen the DNA damage checkpoint are not well understood. Here, we report that budding yeast employs a pathway composed of the scaffold protein Rtt107, its binding partner Mms22, and an Mms22-associated ubiquitin ligase complex to downregulate the DNA damage checkpoint. Mechanistically, this pathway promotes the proteasomal degradation of a key checkpoint factor, Rad9. Furthermore, Rtt107 binding to Mms22 helps to enrich the ubiquitin ligase complex on chromatin and target the chromatin-bound form of Rad9. Finally, we provide evidence that the Rtt107-Mms22 axis operates in parallel with the Rtt107-Slx4 axis, which displaces Rad9 from chromatin. We thus propose that Rtt107 enables a bifurcated "anti-Rad9" strategy to optimally downregulate the DNA damage checkpoint.

12.
MedComm (2020) ; 5(5): e548, 2024 May.
Article in English | MEDLINE | ID: mdl-38645664

ABSTRACT

Identifying new targets for overcoming radioresistance is crucial for improving the efficacy of lung cancer radiotherapy, given that tumor cell resistance is a leading cause of treatment failure. Recent research has spotlighted the significance of Musashi2 (MSI2) in cancer biology. In this study, we first demonstrated that MSI2 plays a key function in regulating the radiosensitivity of lung cancer. The expression of MSI2 is negatively correlated with overall survival in cancer patients, and the knockdown of MSI2 inhibits tumorigenesis and increases radiosensitivity of lung cancer cells. Cellular radiosensitivity, which is closely linked to DNA damage, is influenced by MSI2 interaction with ataxia telangiectasia mutated and Rad3-related kinase (ATR) and checkpoint kinase 1 (CHK1) post-irradiation; moreover, knockdown of MSI2 inhibits the ATR-mediated DNA damage response pathway. RNA-binding motif protein 17 (RBM17), which is implicated in DNA damage repair, exhibits increased interaction with MSI2 post-irradiation. We found that knockdown of RBM17 disrupted the interaction between MSI2 and ATR post-irradiation and increased the radiosensitivity of lung cancer cells. Furthermore, we revealed the potential mechanism of MSI2 recruitment into the nucleus with the assistance of RBM17 to activate ATR to promote radioresistance. This study provides novel insights into the potential application of MSI2 as a new target in lung cancer radiotherapy.

13.
EMBO Rep ; 12(8): 797-803, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21660059

ABSTRACT

Ash2L is a core component of the MLL family histone methyltransferases and has an important role in regulating the methylation of histone H3 on lysine 4. Here, we report the crystal structure of the N-terminal domain of Ash2L and reveal a new function of Ash2L. The structure shows that Ash2L contains an atypical PHD finger that does not have histone tail-binding activity. Unexpectedly, the structure shows a previously unrecognized winged-helix motif that directly binds to DNA. The DNA-binding-deficient mutants of Ash2L reduced Ash2L localization to the HOX locus. Strikingly, a single mutation in Ash2L(WH) (K131A) breaks the chromatin domain boundary, suggesting that Ash2L also has a role in chromosome demarcation.


Subject(s)
Amino Acid Motifs , DNA-Binding Proteins/chemistry , Nuclear Proteins/chemistry , Transcription Factors/chemistry , Winged-Helix Transcription Factors/chemistry , Amino Acid Sequence , Chromatin/metabolism , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genetic Loci , Histones/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Structure, Tertiary , Transcription Factors/genetics , Transcription Factors/metabolism , Winged-Helix Transcription Factors/genetics , Winged-Helix Transcription Factors/metabolism
14.
Cell Mol Biol Lett ; 18(2): 297-314, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23666597

ABSTRACT

PTPN4 is a widely expressed non-receptor protein tyrosine phosphatase. Although its overexpression inhibits cell growth, the proteins with which it interacts to regulate cell growth are unknown. In this study, we identified CrkI as a PTPN4-interacting protein using a yeast two-hybrid, and confirmed this interaction using in vitro GST pull-down and co-immunoprecipitation and co-localization assays. We further determined the interactional regions as the SH3 domain of CrkI and the proline-rich region between amino acids 462 and 468 of PTPN4. Notably, overexpression of PTPN4 inhibits CrkI-mediated proliferation and wound healing of HEK293T cells, while knockdown of PTPN4 by siRNA in Hep3B cells enhances CrkI-mediated cell growth and motility. Moreover, our data show that ectopic expression of PTPN4 reduces the phosphorylation level of CrkI in HEK293T cells. These findings suggest that PTPN4 negatively regulates cell proliferation and motility through dephosphorylation of CrkI.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 4/metabolism , Proto-Oncogene Proteins c-crk/metabolism , Amino Acid Sequence , Cell Movement , Cell Proliferation , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 4/chemistry , RNA Interference , Reproducibility of Results
15.
Cancer Lett ; 575: 216404, 2023 Oct 28.
Article in English | MEDLINE | ID: mdl-37739210

ABSTRACT

Elevated expression and genetic aberration of IRTKS, also named as BAIAP2L1, have been observed in many tumors, especially in tumor progression. however, the molecular and cellular mechanisms involved in the IRTKS-enhanced tumor progression are obscure. Here we show that higher IRTKS level specifically increases histone H3 lysine 9 trimethylation (H3K9me3) by promoting accumulation of the histone methyltransferase SETDB1. Furthermore, we reveal that IRTKS recruits the deubiquitinase OTUD4 to remove Lys48-linked polyubiquitination at K182/K1050 sites of SETDB1, thus blocking SETDB1 degradation via the ubiquitin-proteasome pathway. Interestingly, the enhanced IRTKS-OTUD4-SETDB1-H3K9me3 axis leads to a general decrease in chromatin accessibility, which inhibits transcription of CDH1 encoding E-cadherin, a key molecule essential for maintaining epithelial cell phenotype, and therefore results in epithelial-mesenchymal transition (EMT) and malignant cell metastasis. Clinically, the elevated IRTKS levels in tumor specimens correlate with SETDB1 levels, but negatively associate with survival time. Our data reveal a novel mechanism for the IRTKS-enhanced tumor progression, where IRTKS cooperates with OTUD4 to enhance SETDB1-mediated H3K9 trimethylation that promotes tumor metastasis via suppressing E-cadherin expression. This study also provides a potential approach to reduce the activity and stability of the known therapeutic target SETDB1 possibly through regulating IRTKS or deubiquitinase OTUD4.

16.
Pathol Res Pract ; 237: 154020, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35868058

ABSTRACT

Lysine acetyltransferase 7 (KAT7) was upregulated in gastric cancer (GC) patient tissues, and associated with poor prognosis and metastasis. However, its specific role in GC remains unclear. This study aimed to annotate the role of KAT7 in GC cells. The results showed that the overexpression of KAT7 promoted cell growth, migration, and invasion, while KAT7 inhibition has the opposite effect. Besides, KAT7 participated in cell cycle phase distribution and epithelial-mesenchymal transition (EMT) process of GC cells. In addition, KAT7 promoted the transcription and nuclear translocation of Yes-associated protein 1 (YAP1) in MKN45 cells. Silence of YAP1 partly reversed the promoting effect of KAT7 on GC cells progression. In summary, this study indicates that KAT7 promoted GC cells progression through promoting YAP1 activation, contributes to understand the specific role of KAT7 in GC.


Subject(s)
Lysine Acetyltransferases , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , YAP-Signaling Proteins , Cell Movement , Gene Expression Regulation, Neoplastic/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Cell Proliferation , Lysine Acetyltransferases/metabolism , Histone Acetyltransferases
17.
Environ Technol ; 43(4): 514-523, 2022 Jan.
Article in English | MEDLINE | ID: mdl-32660381

ABSTRACT

In this paper, a method of ball milling and sieving is proposed for recovery of high-grade copper from waste printed circuit boards (WPCBs). The effects of the milling time on the metals grade and recovery of the Cu, Sn and Pb during mechanical treatment were investigated. The results showed that, after 3 cycles of ball milling and sieving, the content of Cu was enriched to 94.72 wt.% from the initial 74.22 wt.% with a high recovery rate of 86.78%. Moreover, the contents of Sn and Pb were enriched to 28.27 wt.% and 18.86 wt.% from 10.13 wt.% and 6.63 wt.% in the by-products, respectively. However, excessive grinding occurred when the milling time was longer than 3 h and led to a sharp decrease in Cu recovery. The X-ray diffraction (XRD) patterns indicated that the metal phases mainly comprised pure Cu, Sn, Pb in the WPCB particles, while a Cu-Sn alloy was formed during the milling process, and the Cu-Sn alloy was also enriched in the tailings. The results presented here establish that ball milling and sieving is an alternative approach to recovering high-grade copper from WPCBs.


Subject(s)
Copper , Electronic Waste , Metals , Recycling , X-Ray Diffraction
18.
Structure ; 30(12): 1565-1572.e4, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36459975

ABSTRACT

Telomerase is crucial for telomere maintenance and genome integrity. The most salient feature of Tetrahymena telomerase is that its CST subcomplex (p75-p45-p19) is tethered to the telomerase catalytic core by interacting with the hub p50. Although the cryoelectron microscopy (cryo-EM) structures of Tetrahymena telomerase have recently been reported, the mechanisms of how and why p50 bridges the CST subcomplex to the telomerase catalytic core remain unclear. Here, we present the nuclear magnetic resonance (NMR) structure of the p75OB1-p50PBM complex. Loss of the interaction between p75 and p50 detaches the CST subcomplex from the telomerase catalytic core in Tetrahymena. The tethering of the CST subcomplex to telomerase is required for telomere homeostasis. However, the detached CST subcomplex is still capable of facilitating the telomeric complementary-strand (C-strand) fill in similar to the non-tethered CST complexes in other organisms. These results expand our understanding of telomere synthesis in Tetrahymena.


Subject(s)
Telomerase , Tetrahymena thermophila , Cryoelectron Microscopy , Telomere , Catalytic Domain
19.
Mol Cell Biochem ; 354(1-2): 11-20, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21461611

ABSTRACT

The epigenetic dysregulation of tumor suppressor genes plays an important role in many cancers, including hepatocellular carcinoma (HCC). In this study, we identified a new gene, family with sequence similarity 43, member B (FAM43B), based on a previous genome-wide approach. FAM43B was significantly downregulated in 60% (24/40) HCC specimens as compared to non-HCC livers. Enforced FAM43B overexpression could suppress cell growth and colony formation in vitro, and induce cell cycle delay, whereas FAM43B knockdown enhanced cell growth. The expression level of FAM43B was found related to the methylation level of FAM43B promoter in HCC cell lines and HCC specimens. The collective data suggest that the expression of FAM43B was regulated by methylation and the epigenetic silencing of FAM43B could contribute to HCC tumorigenesis by regulating cell proliferation.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Genes, Tumor Suppressor , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Organ Specificity , Phylogeny , Promoter Regions, Genetic , RNA Interference
20.
Mol Genet Genomics ; 283(5): 485-92, 2010 May.
Article in English | MEDLINE | ID: mdl-20358383

ABSTRACT

Proliferation of liver cells can be observed in hepatocarcinogenesis, at different stages of liver development, and during liver regeneration after an injury. Does it imply that they share similar molecular mechanisms? Here, the transcriptional profiles of hepatocellular carcinoma (HCC), liver development, and liver regeneration were systematically compared as a preliminary attempt to answer this question. From the comparison, we found that advanced HCC mimics early development in terms of deprived normal liver functions and activated cellular proliferation, but advanced HCC and early development differ in expressions of cancer-related genes and their transcriptional controls. HCC and liver regeneration demonstrate different expression patterns as a whole, but regeneration is similar to dysplasia (pre-stage of HCC) in terms of their proximity to the normal state. In summary, of these three important processes, the carcinogenic progress carries the highest variance in expression; HCC pre-stage shares some resemblance with liver regeneration; and advanced HCC stage displays similarity with early development.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Liver Regeneration/genetics , Liver/embryology , Liver/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cluster Analysis , Gene Expression Profiling , Liver/pathology , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Staging , Principal Component Analysis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL