Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
PLoS Biol ; 22(3): e3002552, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38502677

ABSTRACT

Impediments in replication fork progression cause genomic instability, mutagenesis, and severe pathologies. At stalled forks, RPA-coated single-stranded DNA (ssDNA) activates the ATR kinase and directs fork remodeling, 2 key early events of the replication stress response. RFWD3, a recently described Fanconi anemia (FA) ubiquitin ligase, associates with RPA and promotes its ubiquitylation, facilitating late steps of homologous recombination (HR). Intriguingly, RFWD3 also regulates fork progression, restart and stability via poorly understood mechanisms. Here, we used proteomics to identify putative RFWD3 substrates during replication stress in human cells. We show that RFWD3 interacts with and ubiquitylates the SMARCAL1 DNA translocase directly in vitro and following DNA damage in vivo. SMARCAL1 ubiquitylation does not trigger its subsequent proteasomal degradation but instead disengages it from RPA thereby regulating its function at replication forks. Proper regulation of SMARCAL1 by RFWD3 at stalled forks protects them from excessive MUS81-mediated cleavage in response to UV irradiation, thereby limiting DNA replication stress. Collectively, our results identify RFWD3-mediated SMARCAL1 ubiquitylation as a novel mechanism that modulates fork remodeling to avoid genome instability triggered by aberrant fork processing.


Subject(s)
DNA Replication , DNA, Single-Stranded , Humans , DNA, Single-Stranded/genetics , DNA Replication/genetics , Replication Protein A/genetics , Replication Protein A/metabolism , Protein Binding , Ubiquitination , DNA Damage , Genomic Instability , DNA Helicases/genetics , DNA Helicases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
Nucleic Acids Res ; 49(22): 12836-12854, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34871413

ABSTRACT

Cockayne syndrome group B (CSB) protein has been implicated in the repair of a variety of DNA lesions that induce replication stress. However, little is known about its role at stalled replication forks. Here, we report that CSB is recruited to stalled forks in a manner dependent upon its T1031 phosphorylation by CDK. While dispensable for MRE11 association with stalled forks in wild-type cells, CSB is required for further accumulation of MRE11 at stalled forks in BRCA1/2-deficient cells. CSB promotes MRE11-mediated fork degradation in BRCA1/2-deficient cells. CSB possesses an intrinsic ATP-dependent fork reversal activity in vitro, which is activated upon removal of its N-terminal region that is known to autoinhibit CSB's ATPase domain. CSB functions similarly to fork reversal factors SMARCAL1, ZRANB3 and HLTF to regulate slowdown in fork progression upon exposure to replication stress, indicative of a role of CSB in fork reversal in vivo. Furthermore, CSB not only acts epistatically with MRE11 to facilitate fork restart but also promotes RAD52-mediated break-induced replication repair of double-strand breaks arising from cleavage of stalled forks by MUS81 in BRCA1/2-deficient cells. Loss of CSB exacerbates chemosensitivity in BRCA1/2-deficient cells, underscoring an important role of CSB in the treatment of cancer lacking functional BRCA1/2.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , DNA Repair , DNA/genetics , MRE11 Homologue Protein/genetics , Poly-ADP-Ribose Binding Proteins/genetics , BRCA1 Protein/deficiency , BRCA1 Protein/metabolism , BRCA2 Protein/deficiency , BRCA2 Protein/metabolism , Cell Line , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA Replication/genetics , HCT116 Cells , HEK293 Cells , Humans , MRE11 Homologue Protein/metabolism , Mutation , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Interference
3.
DNA Repair (Amst) ; 104: 103140, 2021 08.
Article in English | MEDLINE | ID: mdl-34087728

ABSTRACT

A broad spectrum of spontaneous and genotoxin-induced DNA lesions impede replication fork progression. The DNA damage response that acts to promote completion of DNA replication is associated with dynamic changes in chromatin structure that include two distinct processes which operate genome-wide during S-phase. The first, often referred to as histone recycling or parental histone segregation, is characterized by the transfer of parental histones located ahead of replication forks onto nascent DNA. The second, known as de novo chromatin assembly, consists of the deposition of new histone molecules onto nascent DNA. Because these two processes occur at all replication forks, their potential to influence a multitude of DNA repair and DNA damage tolerance mechanisms is considerable. The purpose of this review is to provide a description of parental histone segregation and de novo chromatin assembly, and to illustrate how these processes influence cellular responses to DNA replication roadblocks.


Subject(s)
Chromatin/metabolism , DNA Damage , DNA Repair , DNA Replication , Chromatin Assembly and Disassembly , DNA/metabolism , Eukaryota/genetics , Eukaryota/metabolism , Histone Code , Humans
4.
Mol Cancer Ther ; 20(1): 37-49, 2021 01.
Article in English | MEDLINE | ID: mdl-33087510

ABSTRACT

We report the discovery, via a unique high-throughput screening strategy, of a novel bioactive anticancer compound: Thiol Alkylating Compound Inducing Massive Apoptosis (TACIMA)-218. We demonstrate that this molecule engenders apoptotic cell death in genetically diverse murine and human cancer cell lines, irrespective of their p53 status, while sparing normal cells. TACIMA-218 causes oxidative stress in the absence of protective antioxidants normally induced by Nuclear factor erythroid 2-related factor 2 activation. As such, TACIMA-218 represses RNA translation and triggers cell signaling cascade alterations in AKT, p38, and JNK pathways. In addition, TACIMA-218 manifests thiol-alkylating properties resulting in the disruption of redox homeostasis along with key metabolic pathways. When administered to immunocompetent animals as a monotherapy, TACIMA-218 has no apparent toxicity and induces complete regression of pre-established lymphoma and melanoma tumors. In sum, TACIMA-218 is a potent oxidative stress inducer capable of selective cancer cell targeting.


Subject(s)
Antineoplastic Agents/pharmacology , Oxidants/pharmacology , Alkylation , Animals , Cell Death/drug effects , Cell Line, Tumor , Chromatin/metabolism , Cysteine/metabolism , Endoplasmic Reticulum Stress/drug effects , Glycolysis/drug effects , Heme Oxygenase-1/metabolism , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Phenotype , Protein Processing, Post-Translational/drug effects , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism
5.
Front Pharmacol ; 11: 237, 2020.
Article in English | MEDLINE | ID: mdl-32231565

ABSTRACT

Phenotypic screening is an ideal strategy for the discovery of novel bioactive molecules. Using a customized high-throughput screening (HTS) assay employing primary T lymphocytes, we screened a small library of 4,398 compounds with unknown biological function/target to identify compounds eliciting immunomodulatory properties and discovered a sulfonyl-containing hit, we named InhiTinib. This compound inhibited interferon (IFN)-gamma production and proliferation of primary CD3+ T cells without inducing cell death. In contrast, InhiTinib triggered apoptosis in several murine and human cancer cell lines. Besides, the compound was well tolerated by immunocompetent mice, triggered tumor regression in animals with pre-established EL4 T-cell lymphomas, and prolonged the overall survival of mice harboring advanced tumors. Altogether, our data demonstrate the anti-cancer properties of InhiTinib, which can henceforth bridge to wider-scale biochemical and clinical tests following further in-depth pharmacodynamic studies.

6.
Methods Mol Biol ; 1528: 149-164, 2017.
Article in English | MEDLINE | ID: mdl-27854020

ABSTRACT

Antibodies that recognize specific histone modifications are invaluable tools to study chromatin structure and function. There are numerous commercially available antibodies that recognize a remarkable diversity of histone modifications. Unfortunately, many of them fail to work in certain applications or lack the high degree of specificity required of these reagents. The production of affinity-purified polyclonal antibodies against histone modifications demands a little effort but, in return, provides extremely valuable tools that overcome many of the concerns and limitations of commercial antibodies. We present a series of protocols and guidelines for the production and use of large amounts of polyclonal antibodies that recognize modifications of canonical histones. Our protocols can be applied to obtain antibodies that occur in histone variants and proteins other than histones. In addition, some of our protocols are compatible with the production of monoclonal or recombinant antibodies.


Subject(s)
Histones/metabolism , Acetylation , Animals , Chromatin/metabolism , Humans , Methylation , Nucleosomes/metabolism , Protein Processing, Post-Translational
7.
Sci Rep ; 6: 36013, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27782169

ABSTRACT

The mechanism of action of valproate (VPA), a widely prescribed short chain fatty acid with anticonvulsant and anticancer properties, remains poorly understood. Here, the yeast Saccharomyces cerevisiae was used as model to investigate the biological consequences of VPA exposure. We found that low pH strongly potentiates VPA-induced growth inhibition. Transcriptional profiling revealed that under these conditions, VPA modulates the expression of genes involved in diverse cellular processes including protein folding, cell wall organisation, sexual reproduction, and cell cycle progression. We further investigated the impact of VPA on selected processes and found that this drug: i) activates markers of the unfolded protein stress response such as Hac1 mRNA splicing; ii) modulates the cell wall integrity pathway by inhibiting the activation of the Slt2 MAP kinase, and synergizes with cell wall stressors such as micafungin and calcofluor white in preventing yeast growth; iii) prevents activation of the Kss1 and Fus3 MAP kinases of the mating pheromone pathway, which in turn abolishes cellular responses to alpha factor; and iv) blocks cell cycle progression and DNA replication. Overall, our data identify heretofore unknown biological responses to VPA in budding yeast, and highlight the broad spectrum of cellular pathways influenced by this chemical in eukaryotes.


Subject(s)
Cell Cycle/drug effects , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/enzymology , Valproic Acid/pharmacology , Cell Cycle/genetics , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
8.
Nucleic Acids Res ; 44(6): 2706-26, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26748095

ABSTRACT

The Saccharomyces cerevisiae genome encodes five sirtuins (Sir2 and Hst1-4), which constitute a conserved family of NAD-dependent histone deacetylases. Cells lacking any individual sirtuin display mild growth and gene silencing defects. However, hst3Δ hst4Δ double mutants are exquisitely sensitive to genotoxins, and hst3Δ hst4Δ sir2Δmutants are inviable. Our published data also indicate that pharmacological inhibition of sirtuins prevents growth of several fungal pathogens, although the biological basis is unclear. Here, we present genome-wide fitness assays conducted with nicotinamide (NAM), a pan-sirtuin inhibitor. Our data indicate that NAM treatment causes yeast to solicit specific DNA damage response pathways for survival, and that NAM-induced growth defects are mainly attributable to inhibition of Hst3 and Hst4 and consequent elevation of histone H3 lysine 56 acetylation (H3K56ac). Our results further reveal that in the presence of constitutive H3K56ac, the Slx4 scaffolding protein and PP4 phosphatase complex play essential roles in preventing hyperactivation of the DNA damage-response kinase Rad53 in response to spontaneous DNA damage caused by reactive oxygen species. Overall, our data support the concept that chromosome-wide histone deacetylation by sirtuins is critical to mitigate growth defects caused by endogenous genotoxins.


Subject(s)
Chromatin/enzymology , Gene Expression Regulation, Fungal , Genome, Fungal , Histones/genetics , Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics , Sirtuin 2/genetics , Acetylation/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Chromatin/chemistry , Chromatin/drug effects , DNA Damage , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Niacinamide/pharmacology , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Sirtuin 2/metabolism , Stress, Physiological
9.
J Biol Chem ; 291(2): 522-37, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26578521

ABSTRACT

Nucleotide excision repair (NER) is a highly conserved pathway that removes helix-distorting DNA lesions induced by a plethora of mutagens, including UV light. Our laboratory previously demonstrated that human cells deficient in either ATM and Rad3-related (ATR) kinase or translesion DNA polymerase η (i.e. key proteins that promote the completion of DNA replication in response to UV-induced replicative stress) are characterized by profound inhibition of NER exclusively during S phase. Toward elucidating the mechanistic basis of this phenomenon, we developed a novel assay to quantify NER kinetics as a function of cell cycle in the model organism Saccharomyces cerevisiae. Using this assay, we demonstrate that in yeast, deficiency of the ATR homologue Mec1 or of any among several other proteins involved in the cellular response to replicative stress significantly abrogates NER uniquely during S phase. Moreover, initiation of DNA replication is required for manifestation of this defect, and S phase NER proficiency is correlated with the capacity of individual mutants to respond to replicative stress. Importantly, we demonstrate that partial depletion of Rfa1 recapitulates defective S phase-specific NER in wild type yeast; moreover, ectopic RPA1-3 overexpression rescues such deficiency in either ATR- or polymerase η-deficient human cells. Our results strongly suggest that reduction of NER capacity during periods of enhanced replicative stress, ostensibly caused by inordinate sequestration of RPA at stalled DNA replication forks, represents a conserved feature of the multifaceted eukaryotic DNA damage response.


Subject(s)
DNA Repair/genetics , Mutation/genetics , S Phase/genetics , Stress, Physiological/genetics , Cell Line, Tumor , DNA Repair/drug effects , Humans , Mutagens/toxicity , Phosphorylation/drug effects , Pyrimidine Dimers/metabolism , Replication Protein A/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Stress, Physiological/drug effects
10.
J Biol Chem ; 290(48): 28643-63, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26416890

ABSTRACT

The deubiquitinase (DUB) and tumor suppressor BAP1 catalyzes ubiquitin removal from histone H2A Lys-119 and coordinates cell proliferation, but how BAP1 partners modulate its function remains poorly understood. Here, we report that BAP1 forms two mutually exclusive complexes with the transcriptional regulators ASXL1 and ASXL2, which are necessary for maintaining proper protein levels of this DUB. Conversely, BAP1 is essential for maintaining ASXL2, but not ASXL1, protein stability. Notably, cancer-associated loss of BAP1 expression results in ASXL2 destabilization and hence loss of its function. ASXL1 and ASXL2 use their ASXM domains to interact with the C-terminal domain (CTD) of BAP1, and these interactions are required for ubiquitin binding and H2A deubiquitination. The deubiquitination-promoting effect of ASXM requires intramolecular interactions between catalytic and non-catalytic domains of BAP1, which generate a composite ubiquitin-binding interface (CUBI). Notably, the CUBI engages multiple interactions with ubiquitin involving (i) the ubiquitin carboxyl hydrolase catalytic domain of BAP1, which interacts with the hydrophobic patch of ubiquitin, and (ii) the CTD domain, which interacts with a charged patch of ubiquitin. Significantly, we identified cancer-associated mutations of BAP1 that disrupt the CUBI and notably an in-frame deletion in the CTD that inhibits its interaction with ASXL1/2 and DUB activity and deregulates cell proliferation. Moreover, we demonstrated that BAP1 interaction with ASXL2 regulates cell senescence and that ASXL2 cancer-associated mutations disrupt BAP1 DUB activity. Thus, inactivation of the BAP1/ASXL2 axis might contribute to cancer development.


Subject(s)
Cell Proliferation , Neoplasms/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Proteases/metabolism , HEK293 Cells , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neoplasms/genetics , Neoplasms/pathology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Proteases/genetics
11.
Epigenetics ; 10(8): 677-91, 2015.
Article in English | MEDLINE | ID: mdl-26075789

ABSTRACT

O-GlcNAcylation is a posttranslational modification catalyzed by the O-Linked N-acetylglucosamine (O-GlcNAc) transferase (OGT) and reversed by O-GlcNAcase (OGA). Numerous transcriptional regulators, including chromatin modifying enzymes, transcription factors, and co-factors, are targeted by O-GlcNAcylation, indicating that this modification is central for chromatin-associated processes. Recently, OGT-mediated O-GlcNAcylation was reported to be a novel histone modification, suggesting a potential role in directly coordinating chromatin structure and function. In contrast, using multiple biochemical approaches, we report here that histone O-GlcNAcylation is undetectable in mammalian cells. Conversely, O-GlcNAcylation of the transcription regulators Host Cell Factor-1 (HCF-1) and Ten-Eleven Translocation protein 2 (TET2) could be readily observed. Our study raises questions on the occurrence and abundance of O-GlcNAcylation as a histone modification in mammalian cells and reveals technical complications regarding the detection of genuine protein O-GlcNAcylation. Therefore, the identification of the specific contexts in which histone O-GlcNAcylation might occur is still to be established.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Histones/genetics , Host Cell Factor C1/genetics , Proto-Oncogene Proteins/genetics , beta-N-Acetylhexosaminidases/genetics , Acylation , Animals , Chromatin/genetics , Dioxygenases , Glycosylation , HEK293 Cells , Histones/metabolism , Humans , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational/genetics , beta-N-Acetylhexosaminidases/metabolism
12.
Mol Cell ; 54(3): 392-406, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24703950

ABSTRACT

The tumor suppressor BAP1 interacts with chromatin-associated proteins and regulates cell proliferation, but its mechanism of action and regulation remain poorly defined. We show that the ubiquitin-conjugating enzyme UBE2O multi-monoubiquitinates the nuclear localization signal of BAP1, thereby inducing its cytoplasmic sequestration. This activity is counteracted by BAP1 autodeubiquitination through intramolecular interactions. Significantly, we identified cancer-derived BAP1 mutations that abrogate autodeubiquitination and promote its cytoplasmic retention, indicating that BAP1 autodeubiquitination ensures tumor suppression. The antagonistic relationship between UBE2O and BAP1 is also observed during adipogenesis, whereby UBE2O promotes differentiation and cytoplasmic localization of BAP1. Finally, we established a putative targeting consensus sequence of UBE2O and identified numerous chromatin remodeling factors as potential targets, several of which tested positive for UBE2O-mediated ubiquitination. Thus, UBE2O defines an atypical ubiquitin-signaling pathway that coordinates the function of BAP1 and establishes a paradigm for regulation of nuclear trafficking of chromatin-associated proteins.


Subject(s)
Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , 3T3-L1 Cells , Adipocytes/physiology , Amino Acid Sequence , Animals , Cell Differentiation , Consensus Sequence , Cytoplasm/metabolism , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Mutation, Missense , Neoplasms/genetics , Nuclear Localization Signals , Protein Transport , Signal Transduction , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/genetics
13.
Proc Natl Acad Sci U S A ; 111(1): 285-90, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24347639

ABSTRACT

The cellular response to highly genotoxic DNA double-strand breaks (DSBs) involves the exquisite coordination of multiple signaling and repair factors. Here, we conducted a functional RNAi screen and identified BAP1 as a deubiquitinase required for efficient assembly of the homologous recombination (HR) factors BRCA1 and RAD51 at ionizing radiation (IR) -induced foci. BAP1 is a chromatin-associated protein frequently inactivated in cancers of various tissues. To further investigate the role of BAP1 in DSB repair, we used a gene targeting approach to knockout (KO) this deubiquitinase in chicken DT40 cells. We show that BAP1-deficient cells are (i) sensitive to IR and other agents that induce DSBs, (ii) defective in HR-mediated immunoglobulin gene conversion, and (iii) exhibit an increased frequency of chromosomal breaks after IR treatment. We also show that BAP1 is recruited to chromatin in the proximity of a single site-specific I-SceI-induced DSB. Finally, we identified six IR-induced phosphorylation sites in BAP1 and showed that mutation of these residues inhibits BAP1 recruitment to DSB sites. We also found that both BAP1 catalytic activity and its phosphorylation are critical for promoting DNA repair and cellular recovery from DNA damage. Our data reveal an important role for BAP1 in DSB repair by HR, thereby providing a possible molecular basis for its tumor suppressor function.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Gene Expression Regulation, Neoplastic , Homologous Recombination , Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , BRCA1 Protein/metabolism , Cell Line , Cell Line, Tumor , Chickens , DNA Damage , HEK293 Cells , HeLa Cells , Humans , Immunoglobulins/genetics , MCF-7 Cells , Microscopy, Fluorescence , Mutation , Neoplasms/genetics , Phenotype , Phosphorylation , Rad51 Recombinase , Radiation, Ionizing
14.
Cell Signal ; 24(2): 410-421, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22033037

ABSTRACT

Rivaling or cooperating with other post-translational modifications, ubiquitination plays central roles in regulating numerous cellular processes. Not surprisingly, gain- or loss-of-function mutations in several components of the ubiquitin system are causally linked to human pathologies including cancer. The covalent attachment of ubiquitin to target proteins occurs in sequential steps and involves ubiquitin ligases (E3s) which are the most abundant enzymes of the ubiquitin system. Although often associated with proteasomal degradation, ubiquitination is also involved in regulatory events in a proteasome-independent manner. Moreover, ubiquitination is reversible and specific proteases, termed deubiquitinases (DUBs), remove ubiquitin from protein substrates. While we now appreciate the importance of ubiquitin signaling in coordinating a plethora of physio-pathological processes, the molecular mechanisms are not fully understood. This review summarizes current findings on the critical functions exerted by E3s and DUBs in transcriptional control, particularly chromatin remodeling and transcription initiation/elongation.


Subject(s)
Endopeptidases/metabolism , Gene Expression Regulation , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Ubiquitination/genetics , Animals , Cell Communication , Chromatin Assembly and Disassembly/genetics , Endopeptidases/genetics , Humans , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics
15.
Proc Natl Acad Sci U S A ; 108(7): 2747-52, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21285374

ABSTRACT

Host Cell Factor 1 (HCF-1) plays critical roles in regulating gene expression in a plethora of physiological processes. HCF-1 is first synthesized as a precursor, and subsequently specifically proteolytically cleaved within a large middle region termed the proteolytic processing domain (PPD). Although the underlying mechanism remains enigmatic, proteolysis of HCF-1 regulates its transcriptional activity and is important for cell cycle progression. Here we report that HCF-1 proteolysis is a regulated process. We demonstrate that a large proportion of the signaling enzyme O-linked-N-acetylglucosaminyl transferase (OGT) is complexed with HCF-1 and this interaction is essential for HCF-1 cleavage. Moreover, HCF-1 is, in turn, required for stabilizing OGT in the nucleus. We provide evidence indicating that OGT regulates HCF-1 cleavage via interaction with and O-GlcNAcylation of the HCF-1 PPD. In contrast, although OGT also interacts with the basic domain in the HCF-1 amino-terminal subunit, neither the interaction nor the O-GlcNAcylation of this region are required for proteolysis. Moreover, we show that OGT-mediated modulation of HCF-1 impacts the expression of the herpes simplex virus immediate-early genes, targets of HCF-1 during the initiation of viral infection. Together the data indicate that O-GlcNAcylation of HCF-1 is a signal for its proteolytic processing and reveal a unique crosstalk between these posttranslational modifications. Additionally, interactions of OGT with multiple HCF-1 domains may indicate that OGT has several functions in association with HCF-1.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Host Cell Factor C1/metabolism , N-Acetylglucosaminyltransferases/metabolism , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Fluorescent Antibody Technique , Gene Expression Regulation/genetics , Humans , Immediate-Early Proteins/metabolism , Immunoprecipitation , Mutagenesis , Reverse Transcriptase Polymerase Chain Reaction , Simplexvirus/metabolism
16.
PLoS One ; 5(11): e14027, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-21103343

ABSTRACT

BACKGROUND: The function of BRCA1 in response to ionizing radiation, which directly generates DNA double strand breaks, has been extensively characterized. However previous investigations have produced conflicting data on mutagens that initially induce other classes of DNA adducts. Because of the fundamental and clinical importance of understanding BRCA1 function, we sought to rigorously evaluate the role of this tumor suppressor in response to diverse forms of genotoxic stress. METHODOLOGY/PRINCIPAL FINDINGS: We investigated BRCA1 stability and localization in various human cells treated with model mutagens that trigger different DNA damage signaling pathways. We established that, unlike ionizing radiation, either UVC or methylmethanesulfonate (MMS) (generating bulky DNA adducts or alkylated bases respectively) induces a transient downregulation of BRCA1 protein which is neither prevented nor enhanced by inhibition of PIKKs. Moreover, we found that the proteasome mediates early degradation of BRCA1, BARD1, BACH1, and Rad52 implying that critical components of the homologous recombination machinery need to be functionally abrogated as part of the early response to UV or MMS. Significantly, we found that inhibition of BRCA1/BARD1 downregulation is accompanied by the unscheduled recruitment of both proteins to chromatin along with Rad51. Consistently, treatment of cells with MMS engendered complete disassembly of Rad51 from pre-formed ionizing radiation-induced foci. Following the initial phase of BRCA1/BARD1 downregulation, we found that the recovery of these proteins in foci coincides with the formation of RPA and Rad51 foci. This indicates that homologous recombination is reactivated at later stage of the cellular response to MMS, most likely to repair DSBs generated by replication blocks. CONCLUSION/SIGNIFICANCE: Taken together our results demonstrate that (i) the stabilities of BRCA1/BARD1 complexes are regulated in a mutagen-specific manner, and (ii) indicate the existence of mechanisms that may be required to prevent the simultaneous recruitment of conflicting signaling pathways to sites of DNA damage.


Subject(s)
BRCA1 Protein/metabolism , DNA Damage , Protein Serine-Threonine Kinases/metabolism , Rad51 Recombinase/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , Chromatin/metabolism , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Down-Regulation/radiation effects , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Male , Methyl Methanesulfonate/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ultraviolet Rays
17.
Mol Cell Biol ; 30(21): 5071-85, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20805357

ABSTRACT

The candidate tumor suppressor BAP1 is a deubiquitinating enzyme (DUB) involved in the regulation of cell proliferation, although the molecular mechanisms governing its function remain poorly defined. BAP1 was recently shown to interact with and deubiquitinate the transcriptional regulator host cell factor 1 (HCF-1). Here we show that BAP1 assembles multiprotein complexes containing numerous transcription factors and cofactors, including HCF-1 and the transcription factor Yin Yang 1 (YY1). Through its coiled-coil motif, BAP1 directly interacts with the zinc fingers of YY1. Moreover, HCF-1 interacts with the middle region of YY1 encompassing the glycine-lysine-rich domain and is essential for the formation of a ternary complex with YY1 and BAP1 in vivo. BAP1 activates transcription in an enzymatic-activity-dependent manner and regulates the expression of a variety of genes involved in numerous cellular processes. We further show that BAP1 and HCF-1 are recruited by YY1 to the promoter of the cox7c gene, which encodes a mitochondrial protein used here as a model of BAP1-activated gene expression. Our findings (i) establish a direct link between BAP1 and the transcriptional control of genes regulating cell growth and proliferation and (ii) shed light on a novel mechanism of transcription regulation involving ubiquitin signaling.


Subject(s)
Host Cell Factor C1/chemistry , Host Cell Factor C1/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , YY1 Transcription Factor/chemistry , YY1 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cattle , Cell Line , Cell Proliferation , DNA/genetics , DNA/metabolism , Electron Transport Complex IV/genetics , HeLa Cells , Host Cell Factor C1/antagonists & inhibitors , Host Cell Factor C1/genetics , Humans , In Vitro Techniques , Mice , Models, Biological , Molecular Sequence Data , Multiprotein Complexes , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA Interference , Sequence Homology, Nucleic Acid , Signal Transduction , Transcriptional Activation , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitination , YY1 Transcription Factor/antagonists & inhibitors , YY1 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...