Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Res Sq ; 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39070640

ABSTRACT

DNA Double-strand breaks (DSBs) are harmful lesions and major sources of genomic instability. Studies have suggested that DSBs induce local transcriptional silencing that consequently promotes genomic stability. Several factors have been proposed to actively participate in this process, including ATM and Polycomb repressive complex 1 (PRC1). Here we found that disrupting PRC1 clustering disrupts DSB-induced gene silencing. Interactome analysis of PHC2, a PRC1 subunit that promotes the formation of the Polycomb body, found several nucleoporins that constitute the Nuclear Pore Complex (NPC). Similar to PHC2, depleting the nucleoporins also disrupted the DSB-induced gene silencing. We found that some of these nucleoporins, such as NUP107 and NUP43, which are members of the Y-complex of NPC, localize to DSB sites. These nucleoporin-enriched DSBs were distant from the nuclear periphery. The presence of nucleoporins and PHC2 at DSB regions were inter-dependent, suggesting that they act cooperatively in the DSB-induced gene silencing. We further found two structural components within NUP107 to be necessary for the transcriptional repression at DSBs: ATM/ATR-mediated phosphorylation at Serine37 residue within the N-terminal disordered tail, and the NUP133-binding surface at the C-terminus. These results provide a new functional interplay among nucleoporins, ATM and the Polycomb proteins in the DSB metabolism, and underscore their emerging roles in genome stability maintenance. *Hongseon Song, Yubin Bae, Sangin Kim, and Dante Deascanis contributed equally to this work.

2.
Nucleic Acids Res ; 51(19): 10467-10483, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37713620

ABSTRACT

Proper regulation of replication fork progression is important for genomic maintenance. Subverting the transcription-induced conflicts is crucial in preserving the integrity of replication forks. Various chromatin remodelers, such as histone chaperone and histone deacetylases are known to modulate replication stress, but how these factors are organized or collaborate are not well understood. Here we found a new role of the OTUD5 deubiquitinase in limiting replication stress. We found that OTUD5 is recruited to replication forks, and its depletion causes replication fork stress. Through its C-terminal disordered tail, OTUD5 assembles a complex containing FACT, HDAC1 and HDAC2 at replication forks. A cell line engineered to specifically uncouple FACT interaction with OTUD5 exhibits increases in FACT loading onto chromatin, R-loop formation, and replication fork stress. OTUD5 mediates these processes by recruiting and stabilizing HDAC1 and HDAC2, which decreases H4K16 acetylation and FACT recruitment. Finally, proteomic analysis revealed that the cells with deficient OTUD5-FACT interaction activates the Fanconi Anemia pathway for survival. Altogether, this study identified a new interaction network among OTUD5-FACT-HDAC1/2 that limits transcription-induced replication stress.


Subject(s)
Chromatin , DNA Replication , Humans , Cell Line , Chromatin/genetics , Genomic Instability , Proteomics
3.
Cell Death Dis ; 13(11): 933, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36344491

ABSTRACT

Serine/arginine-rich splicing factor 3 (SRSF3) is an RNA binding protein that most often regulates gene expression at the splicing level. Although the role of SRSF3 in mRNA splicing in the nucleus is well known, its splicing-independent role outside of the nucleus is poorly understood. Here, we found that SRSF3 exerts a translational control of p21 mRNA. Depletion of SRSF3 induces cellular senescence and increases the expression of p21 independent of p53. Consistent with the expression patterns of SRSF3 and p21 mRNA in the TCGA database, SRSF3 knockdown increases the p21 mRNA level and its translation efficiency as well. SRSF3 physically associates with the 3'UTR region of p21 mRNA and the translational initiation factor, eIF4A1. Our study proposes a model in which SRSF3 regulates translation by interacting with eIF4A1 at the 3'UTR region of p21 mRNA. We also found that SRSF3 localizes to the cytoplasmic RNA granule along with eIF4A1, which may assist in translational repression therein. Thus, our results provide a new mode of regulation for p21 expression, a crucial regulator of the cell cycle and senescence, which occurs at the translational level and involves SRSF3.


Subject(s)
RNA Splicing , RNA-Binding Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , 3' Untranslated Regions/genetics , RNA Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , RNA-Binding Proteins/metabolism
4.
Nucleic Acids Res ; 50(18): 10469-10486, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36155803

ABSTRACT

Human CtIP maintains genomic integrity primarily by promoting 5' DNA end resection, an initial step of the homologous recombination (HR). A few mechanisms have been suggested as to how CtIP recruitment to damage sites is controlled, but it is likely that we do not yet have full understanding of the process. Here, we provide evidence that CtIP recruitment and functioning are controlled by the SIAH2 E3 ubiquitin ligase. We found that SIAH2 interacts and ubiquitinates CtIP at its N-terminal lysine residues. Mutating the key CtIP lysine residues impaired CtIP recruitment to DSBs and stalled replication forks, DSB end resection, overall HR repair capacity of cells, and recovery of stalled replication forks, suggesting that the SIAH2-induced ubiquitination is important for relocating CtIP to sites of damage. Depleting SIAH2 consistently phenocopied these results. Overall, our work suggests that SIAH2 is a new regulator of CtIP and HR repair, and emphasizes that SIAH2-mediated recruitment of the CtIP is an important step for CtIP's function during HR repair.


Subject(s)
DNA Repair , DNA Replication , Endodeoxyribonucleases/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , Endodeoxyribonucleases/genetics , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitination
5.
Mol Cell Oncol ; 9(1): 2054263, 2022.
Article in English | MEDLINE | ID: mdl-35372672

ABSTRACT

RAD51 loading onto chromatin is a key step during the homologous recombination (HR) repair. We recently reported a new mode of RAD51 regulation, which is mediated by TOPORS E3 SUMO ligase and RAD51 SUMOylation. ATM/ATR-induced phosphorylation of TOPORS is necessary for this event, revealing a new role of these master DNA damage response kinases in HR repair.

6.
Mar Drugs ; 20(1)2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35049906

ABSTRACT

Sponges are at the forefront of marine natural product research. In the deep sea, extreme conditions have driven secondary metabolite pathway evolution such that we might expect deep-sea sponges to yield a broad range of unique natural products. Here, we investigate the chemodiversity of a deep-sea tetractinellid sponge, Characella pachastrelloides, collected from ~800 m depth in Irish waters. First, we analyzed the MS/MS data obtained from fractions of this sponge on the GNPS public online platform to guide our exploration of its chemodiversity. Novel glycolipopeptides named characellides were previously isolated from the sponge and herein cyanocobalamin, a manufactured form of vitamin B12, not previously found in nature, was isolated in a large amount. We also identified several poecillastrins from the molecular network, a class of polyketide known to exhibit cytotoxicity. Light sensitivity prevented the isolation and characterization of these polyketides, but their presence was confirmed by characteristic NMR and MS signals. Finally, we isolated the new betaine 6-methylhercynine, which contains a unique methylation at C-2 of the imidazole ring. This compound showed potent cytotoxicity towards against HeLa (cervical cancer) cells.


Subject(s)
Antineoplastic Agents/pharmacology , Porifera , Vitamin B 12/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Aquatic Organisms , Female , HeLa Cells/drug effects , Humans , Uterine Cervical Neoplasms/pathology , Vitamin B 12/chemistry , Vitamin B 12/therapeutic use
7.
Nucleic Acids Res ; 50(3): 1501-1516, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35061896

ABSTRACT

Homologous recombination (HR) is critical for error-free repair of DNA double-strand breaks. Chromatin loading of RAD51, a key protein that mediates the recombination, is a crucial step in the execution of the HR repair. Here, we present evidence that SUMOylation of RAD51 is crucial for the RAD51 recruitment to chromatin and HR repair. We found that topoisomerase 1-binding arginine/serine-rich protein (TOPORS) induces the SUMOylation of RAD51 at lysine residues 57 and 70 in response to DNA damaging agents. The SUMOylation was facilitated by an ATM-induced phosphorylation of TOPORS at threonine 515 upon DNA damage. Knockdown of TOPORS or expression of SUMOylation-deficient RAD51 mutants caused reduction in supporting normal RAD51 functions during the HR repair, suggesting the physiological importance of the modification. We found that the SUMOylation-deficient RAD51 reduces the association with its crucial binding partner BRCA2, explaining its deficiency in supporting the HR repair. These findings altogether demonstrate a crucial role for TOPORS-mediated RAD51 SUMOylation in promoting HR repair and genomic maintenance.


Subject(s)
Rad51 Recombinase , Recombinational DNA Repair , Chromatin , DNA/metabolism , DNA Damage , DNA Repair/genetics , Homologous Recombination , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Sumoylation
8.
Int J Mol Sci ; 22(15)2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34360546

ABSTRACT

Promyelocytic leukemia (PML) protein is the core component of subnuclear structures called PML nuclear bodies that are known to play important roles in cell survival, DNA damage responses, and DNA repair. Fanconi anemia (FA) proteins are required for repairing interstrand DNA crosslinks (ICLs). Here we report a novel role of PML proteins, regulating the ICL repair pathway. We found that depletion of the PML protein led to the significant reduction of damage-induced FANCD2 mono-ubiquitination and FANCD2 foci formation. Consistently, the cells treated with siRNA against PML showed enhanced sensitivity to a crosslinking agent, mitomycin C. Further studies showed that depletion of PML reduced the protein expression of FANCA, FANCG, and FANCD2 via reduced transcriptional activity. Interestingly, we observed that damage-induced CHK1 phosphorylation was severely impaired in cells with depleted PML, and we demonstrated that CHK1 regulates FANCA, FANCG, and FANCD2 transcription. Finally, we showed that inhibition of CHK1 phosphorylation further sensitized cancer cells to mitomycin C. Taken together, these findings suggest that the PML is critical for damage-induced CHK1 phosphorylation, which is important for FA gene expression and for repairing ICLs.


Subject(s)
Checkpoint Kinase 1/metabolism , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group G Protein/metabolism , Fanconi Anemia/pathology , Gene Expression Regulation , Checkpoint Kinase 1/genetics , DNA Damage , DNA Repair , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group G Protein/genetics , HeLa Cells , Humans , Phosphorylation , Ubiquitination
9.
Oncotarget ; 11(21): 2024-2025, 2020 May 26.
Article in English | MEDLINE | ID: mdl-32523656

ABSTRACT

[This corrects the article DOI: 10.18632/oncotarget.10275.].

10.
PLoS Genet ; 16(3): e1008524, 2020 03.
Article in English | MEDLINE | ID: mdl-32142505

ABSTRACT

Common fragile sites (CFSs) are breakage-prone genomic loci, and are considered to be hotspots for genomic rearrangements frequently observed in cancers. Understanding the underlying mechanisms for CFS instability will lead to better insight on cancer etiology. Here we show that Polycomb group proteins BMI1 and RNF2 are suppressors of transcription-replication conflicts (TRCs) and CFS instability. Cells depleted of BMI1 or RNF2 showed slower replication forks and elevated fork stalling. These phenotypes are associated with increase occupancy of RNA Pol II (RNAPII) at CFSs, suggesting that the BMI1-RNF2 complex regulate RNAPII elongation at these fragile regions. Using proximity ligase assays, we showed that depleting BMI1 or RNF2 causes increased associations between RNAPII with EdU-labeled nascent forks and replisomes, suggesting increased TRC incidences. Increased occupancy of a fork protective factor FANCD2 and R-loop resolvase RNH1 at CFSs are observed in RNF2 CRISPR-KO cells, which are consistent with increased transcription-associated replication stress in RNF2-deficient cells. Depleting FANCD2 or FANCI proteins further increased genomic instability and cell death of the RNF2-deficient cells, suggesting that in the absence of RNF2, cells depend on these fork-protective factors for survival. These data suggest that the Polycomb proteins have non-canonical roles in suppressing TRC and preserving genomic integrity.


Subject(s)
Chromosome Fragile Sites/genetics , DNA Replication/genetics , Polycomb Repressive Complex 1/genetics , Transcription, Genetic/genetics , Cell Line , Cell Line, Tumor , Genomic Instability/genetics , HEK293 Cells , HeLa Cells , Humans
11.
Mar Drugs ; 17(9)2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31480497

ABSTRACT

: An Antarctic coral belonging to the order Pennatulacea, collected during the 2013 austral autumn by trawl from 662 to 944 m depth, has yielded three new briarane diterpenes, bathyptilone A-C (1-3) along with a trinorditerpene, enbepeanone A (4), which bears a new carbon skeleton. Structure elucidation was facilitated by one- and two-dimensional NMR spectroscopy, mass spectrometry and confirmed by X-ray crystallography. The three compounds were screened in four cancer cell lines. Bathyptilone A displayed selective nanomolar cytotoxicity against the neurogenic mammalian cell line Ntera-2.


Subject(s)
Anthozoa/chemistry , Terpenes/chemistry , Animals , Antarctic Regions , Crystallography, X-Ray/methods , Diterpenes/chemistry , HeLa Cells , Humans , Magnetic Resonance Spectroscopy/methods , Mass Spectrometry/methods
12.
Oncogenesis ; 8(9): 46, 2019 Aug 21.
Article in English | MEDLINE | ID: mdl-31434871

ABSTRACT

Mechanisms underlying the pathogenesis of high-grade serous epithelial ovarian cancers (HGSOC) are not yet well defined although key precursor cells have been identified (including fimbriated fallopian tube epithelium, FTSECs). Since iron is elevated in endometriotic cysts and the pelvic cavity, it is suggested that this source of redox-active iron may contribute to ovarian cancer pathogenesis. Specifically, sources of nontransferrin-bound iron (NTBI) within the pelvic cavity could arise from ovulation, retrograde menstruation, follicular fluid, or iron overload conditions (i.e., hemochromatosis). Herein, we investigated the cellular response of p53-inactivated and telomerase-expressing (immortalized) FTSECs (Pax8+/FoxJ1-) to NTBI (presented as ferric ammonium citrate (FAC), supplemented in media for >2 months) in order to assess its ability to promote the transition to a tumor-like phenotype; this cellular response was compared with immortalized FTSECs transformed with H-RasV12A and c-MycT58A. Both approaches resulted in increased cell numbers and expression of the oncogenic transcriptional regulator, ecotropic virus integration site 1 (EVI1, a gene most frequently amplified at 3q26.2 in HGSOC, represented by multiple variants), along with other oncogenic gene products. In contrast to the transformed cells, FAC-exposed FTSECs elicited elevated migratory capacity (and epithelial-mesenchymal transition mRNA profile) along with increased expression of DNA damage response proteins (i.e., FANCD2) and hTERT mRNA relative to controls. Interestingly, in FAC-exposed FTSECs, EVI1 siRNA attenuated hTERT mRNA expression, whereas siRNAs targeting ß-catenin and BMI1 (both elevated with chronic iron exposure) reduced Myc and Cyclin D1 proteins. Collectively, our novel findings provide strong foundational evidence for potential iron-induced initiation events, including EVI1 alterations, in the pathogenesis of HGSOC, warranting further in depth investigations. Thus, these findings will substantially advance our understanding of the contribution of iron enriched within the pelvic cavity, which may identify patients at risk of developing this deadly disease.

13.
Mar Drugs ; 17(4)2019 Apr 17.
Article in English | MEDLINE | ID: mdl-30999651

ABSTRACT

The subtidal red alga Plocamium cartilagineum was collected from the Western Antarctic Peninsula during the 2011 and 2017 austral summers. Bulk collections from specific sites corresponded to chemogroups identified by Young et al. in 2013. One of the chemogroups yielded several known acyclic halogenated monoterpenes (2-5) as well as undescribed compounds of the same class, anverenes B-D (6-8). Examination of another chemogroup yielded an undescribed cyclic halogenated monoterpene anverene E (9) as its major secondary metabolite. Elucidation of structures was achieved through one-dimensional (1D) and 2D nuclear magnetic resonance (NMR) spectroscopy and negative chemical ionization mass spectrometry. Compounds 1-9 show moderate cytotoxicity against cervical cancer (HeLa) cells.


Subject(s)
Monoterpenes/chemistry , Monoterpenes/pharmacology , Plocamium/chemistry , Antarctic Regions , Cell Line, Tumor , Cell Survival/drug effects , HeLa Cells , Humans , Hydrocarbons, Halogenated/chemistry , Hydrocarbons, Halogenated/isolation & purification , Hydrocarbons, Halogenated/pharmacology , Inhibitory Concentration 50 , Molecular Conformation , Monoterpenes/isolation & purification , Nuclear Magnetic Resonance, Biomolecular
14.
Nucleic Acids Res ; 47(2): 729-746, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30508113

ABSTRACT

Timely stalling and resumption of RNA polymerases at damaged chromatin are actively regulated processes. Prior work showed an importance of FACT histone chaperone in such process. Here we provide a new role of OTUD5 deubiquitinase in the FACT-dependent process. Through a DUB RNAi screen, we found OTUD5 as a specific stabilizer of the UBR5 E3 ligase. OTUD5 localizes to DNA double strand breaks (DSBs), interacts with UBR5 and represses the RNA Pol II elongation and RNA synthesis. OTUD5 co-localizes and interacts with the FACT component SPT16 and antagonizes the histone H2A deposition at DSB lesions. OTUD5 interacts with UBR5 and SPT16 independently through two distinct regions, and both interactions are necessary for arresting the Pol II elongation at lesions. These analyses suggested that the catalytic (through UBR5 stabilization) as well as scaffolding (through FACT binding) activities of OTUD5 are involved in the FACT-dependent transcription. We found that a cancer-associated missense mutation within the OTUD5 Ubiquitin Interacting Motif (UIM) abrogates the FACT association and the Pol II arrest, providing a possible link between the transcriptional regulation and tumor suppression. Our work establishes OTUD5 as a new regulator of the DNA damage response, and provides an insight into the FACT-dependent transcription at damaged chromatin.


Subject(s)
Chromatin/metabolism , DNA Damage , Endopeptidases/metabolism , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , DNA Breaks, Double-Stranded , Endopeptidases/chemistry , Endopeptidases/genetics , Gene Expression Regulation , Genomic Instability , Histones/metabolism , Humans , Mutation , Neoplasms/genetics , Protein Interaction Domains and Motifs , RNA Polymerase II/metabolism , Transcription Elongation, Genetic , Transcription Factors/metabolism
15.
Proc Natl Acad Sci U S A ; 113(40): 11243-11248, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27647897

ABSTRACT

BMI1 is a component of the Polycomb Repressive Complex 1 (PRC1), which plays a key role in maintaining epigenetic silencing during development. BMI1 also participates in gene silencing during DNA damage response, but the precise downstream function of BMI1 in gene silencing is unclear. Here we identified the UBR5 E3 ligase as a downstream factor of BMI1. We found that UBR5 forms damage-inducible nuclear foci in a manner dependent on the PRC1 components BMI1, RNF1 (RING1a), and RNF2 (RING1b). Whereas transcription is repressed at UV-induced lesions on chromatin, depletion of the PRC1 members or UBR5 alone derepressed transcription elongation at these sites, suggesting that UBR5 functions in a linear pathway with PRC1 in inducing gene silencing at lesions. Mass spectrometry (MS) analysis revealed that UBR5 associates with BMI1 as well as FACT components SPT16 and SSRP1. We found that UBR5 localizes to the UV-induced lesions along with SPT16. We show that UBR5 ubiquitinates SPT16, and depletion of UBR5 or BMI1 leads to an enlargement of SPT16 foci size at UV lesions, suggesting that UBR5 and BMI1 repress SPT16 enrichment at the damaged sites. Consistently, depletion of the FACT components effectively reversed the transcriptional derepression incurred in the UBR5 and BMI1 KO cells. Finally, UBR5 and BMI1 KO cells are hypersensitive to UV, which supports the notion that faulty RNA synthesis at damaged sites is harmful to the cell fitness. Altogether, these results suggest that BMI1 and UBR5 repress the polymerase II (Pol II)-mediated transcription at damaged sites, by negatively regulating the FACT-dependent Pol II elongation.


Subject(s)
Chromatin/metabolism , Polycomb Repressive Complex 1/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism , HCT116 Cells , HeLa Cells , Humans , Protein Binding , Signal Transduction , Transcription Elongation, Genetic , Ultraviolet Rays
16.
Nat Commun ; 7: 12125, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27381497

ABSTRACT

Stress granules (SGs) harbour translationally stalled messenger ribonucleoproteins and play important roles in regulating gene expression and cell fate. Here we show that neddylation promotes SG assembly in response to arsenite-induced oxidative stress. Inhibition or depletion of key components of the neddylation machinery concomitantly inhibits stress-induced polysome disassembly and SG assembly. Affinity purification and subsequent mass-spectrometric analysis of Nedd8-conjugated proteins from translationally stalled ribosomal fractions identified ribosomal proteins, translation factors and RNA-binding proteins (RBPs), including SRSF3, a previously known SG regulator. We show that SRSF3 is selectively neddylated at Lys85 in response to arsenite. A non-neddylatable SRSF3 (K85R) mutant do not prevent arsenite-induced polysome disassembly, but fails to support the SG assembly, suggesting that the neddylation pathway plays an important role in SG assembly.


Subject(s)
Arsenites/toxicity , Cytoplasmic Granules/drug effects , NEDD8 Protein/genetics , Protein Biosynthesis/drug effects , Serine-Arginine Splicing Factors/genetics , Cell Line, Tumor , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , HEK293 Cells , HeLa Cells , Humans , Lysine/metabolism , NEDD8 Protein/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Oxidative Stress , Polyribosomes/drug effects , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
17.
Cell Cycle ; 15(19): 2636-2646, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27463890

ABSTRACT

USP1 deubiquitinating enzyme and its stoichiometric binding partner UAF1 play an essential role in promoting DNA homologous recombination (HR) repair in response to various types of DNA damaging agents. Deubiquitination of FANCD2 may be attributed to the key role of USP1-UAF1 complex in regulating HR repair, however whether USP1-UAF1 promotes HR repair independently of FANCD2 deubiquitination is not known. Here we show evidence that the USP1-UAF1 complex has a FANCD2-independent function in promoting HR repair. Proteomic search of UAF1-interacting proteins revealed that UAF1 associates with RAD51AP1, a RAD51-interacting protein implicated in HR repair. We show that UAF1 mediates the interaction between USP1 and RAD51AP1, and that depletion of USP1 or UAF1 led to a decreased stability of RAD51AP1. Protein interaction mapping analysis identified some key residues within RAD51AP1 required for interacting with the USP1-UAF1 complex. Cells expressing the UAF1 interaction-deficient mutant of RAD51AP1 show increased chromosomal aberrations in response to Mitomycin C treatment. Moreover, similar to the RAD51AP1 depleted cells, the cells expressing UAF1-interaction deficient RAD51AP1 display persistent RAD51 foci following DNA damage exposure, indicating that these factors regulate a later step during the HR repair. These data altogether suggest that the USP1-UAF1 complex promotes HR repair via multiple mechanisms: through FANCD2 deubiquitination, as well as by interacting with RAD51AP1.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Recombinational DNA Repair , Ubiquitin-Specific Proteases/metabolism , Gene Deletion , Gene Knockdown Techniques , HeLa Cells , Humans , Mutant Proteins/metabolism , Phenotype , Protein Binding , Protein Interaction Mapping , Protein Stability , RNA-Binding Proteins
18.
Oncotarget ; 7(29): 45976-45994, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27351285

ABSTRACT

Inflammation is a potent inducer of tumorigenesis. Increased DNA damage or loss of genome integrity is thought to be one of the mechanisms linking inflammation and cancer development. It has been suggested that NF-κB-induced microRNA-146 (miR146a) may be a mediator of the inflammatory response. Based on our initial observation that miR146a overexpression strongly increases DNA damage, we investigated its potential role as a modulator of DNA repair. Here, we demonstrate that FANCM, a component in the Fanconi Anemia pathway, is a novel target of miR146a. miR146a suppressed FANCM expression by directly binding to the 3' untranslated region of the gene. miR146a-induced downregulation of FANCM was associated with inhibition of FANCD2 monoubiquitination, reduced DNA homologous recombination repair and checkpoint response, failed recovery from replication stress, and increased cellular sensitivity to cisplatin. These phenotypes were recapitulated when miR146a expression was induced by overexpressing the NF-κB subunit p65/RelA or Helicobacter pylori infection in a human gastric cell line; the phenotypes were effectively reversed with an anti-miR146a antagomir. These results suggest that undesired inflammation events caused by a pathogen or over-induction of miR146a can impair genome integrity via suppression of FANCM.


Subject(s)
DNA Helicases/biosynthesis , Gene Expression Regulation/genetics , MicroRNAs/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA Damage/physiology , DNA Helicases/genetics , DNA Repair/physiology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology
19.
BMB Rep ; 49(8): 449-54, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27345716

ABSTRACT

Stress Granules (SGs) are microscopically visible, phase dense aggregates of translationally stalled messenger ribonucleoprotein (mRNP) complexes formed in response to distinct stress conditions. It is generally considered that SG formation is induced to protect cells from conditions of stress. The precise constituents of SGs and the mechanism through which SGs are dynamically regulated in response to stress are not completely understood. Hence, it is important to identify proteins which regulate SG assembly and disassembly. In the present study, we report Neuregulin-2 (NRG2) as a novel component of SGs; furthermore, depletion of NRG2 potently inhibits SG formation. We also demonstrate that NRG2 specifically localizes to SGs under various stress conditions. Knockdown of NRG2 has no effect on stress-induced polysome disassembly, suggesting that the component does not influence early step of SG formation. It was also observed that reduced expression of NRG2 led to marginal increase in cell survival under arsenite-induced stress. [BMB Reports 2016; 49(8): 449-454].


Subject(s)
Cytoplasmic Granules/metabolism , Nerve Growth Factors/metabolism , Stress, Physiological , Arsenites/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cytoplasmic Granules/drug effects , Gene Deletion , Gene Knockdown Techniques , HEK293 Cells , Humans , Oxidative Stress/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , RNA, Small Interfering/metabolism , Stress, Physiological/drug effects
20.
Mol Cell Oncol ; 3(6): e1244513, 2016.
Article in English | MEDLINE | ID: mdl-28090582

ABSTRACT

One of the cellular responses to DNA damage is to monitor and execute temporary arrest of RNA synthesis at chromatin lesions. The Polycomb silencer BMI1 is a well-known contributor to this process. We recently described a new mode of BMI1-mediated transcription arrest at lesions that involves UBR5 E3 ligase and FACT histone chaperon.

SELECTION OF CITATIONS
SEARCH DETAIL