Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nucleic Acids Res ; 51(14): 7342-7356, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37326017

ABSTRACT

Efficient repair of oxidized DNA is critical for genome-integrity maintenance. Cockayne syndrome protein B (CSB) is an ATP-dependent chromatin remodeler that collaborates with Poly(ADP-ribose) polymerase I (PARP1) in the repair of oxidative DNA lesions. How these proteins integrate during DNA repair remains largely unknown. Here, using chromatin co-fractionation studies, we demonstrate that PARP1 and PARP2 promote recruitment of CSB to oxidatively-damaged DNA. CSB, in turn, contributes to the recruitment of XRCC1, and histone PARylation factor 1 (HPF1), and promotes histone PARylation. Using alkaline comet assays to monitor DNA repair, we found that CSB regulates single-strand break repair (SSBR) mediated by PARP1 and PARP2. Strikingly, CSB's function in SSBR is largely bypassed when transcription is inhibited, suggesting CSB-mediated SSBR occurs primarily at actively transcribed DNA regions. While PARP1 repairs SSBs at sites regardless of the transcription status, we found that PARP2 predominantly functions in actively transcribed DNA regions. Therefore, our study raises the hypothesis that SSBR is executed by different mechanisms based on the transcription status.


Subject(s)
Chromatin , Humans , Carrier Proteins/genetics , Chromatin/genetics , DNA/genetics , DNA/metabolism , DNA Repair , Histones/metabolism , Nuclear Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , X-ray Repair Cross Complementing Protein 1/genetics , X-ray Repair Cross Complementing Protein 1/metabolism
2.
Methods Mol Biol ; 2472: 95-108, 2022.
Article in English | MEDLINE | ID: mdl-35674895

ABSTRACT

The sequence-specific transcription factor RBPJ, also known as CSL (CBF1, Su(H), Lag1), is an evolutionarily conserved protein that mediates Notch signaling to guide cell fates. When cells enter mitosis, DNA is condensed and most transcription factors dissociate from chromatin; however, a few, select transcription factors, termed bookmarking factors, remain associated. These mitotic chromatin-bound factors are believed to play important roles in maintaining cell fates through cell division. RBPJ is one such factor that remains mitotic chromatin associated and therefore could function as a bookmarking factor. Here, we describe how to obtain highly purified mitotic cells from the mouse embryonal carcinoma cell line F9, perform chromatin immunoprecipitation with mitotic cells, and measure the first run of RNA synthesis upon mitotic exit. These methods serve as basis to understand the roles of mitotic bookmarking by RBPJ in propagating Notch signals through cell division.


Subject(s)
Chromatin , Chromosomes , Animals , Chromatin/genetics , Chromosomes/metabolism , Gene Expression Regulation , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Mitosis , Transcription Factors/metabolism
3.
Biomedicines ; 10(2)2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35203571

ABSTRACT

Oxidative stress contributes to numerous diseases, including cancer. CSB is an ATP-dependent chromatin remodeler critical for oxidative stress relief. PARP1 is the major sensor for DNA breaks and fundamental for efficient single-strand break repair. DNA breaks activate PARP1, leading to the synthesis of poly(ADP-ribose) (PAR) on itself and neighboring proteins, which is crucial for the recruitment of DNA repair machinery. CSB and PARP1 interact; however, how CSB mechanistically participates in oxidative DNA damage repair mediated by PARP1 remains unclear. Using chromatin immunoprecipitation followed by quantitative PCR, we found that CSB and PARP1 facilitate each other's chromatin association during the onset of oxidative stress, and that CSB facilitates PARP1 removal when the level of chromatin-bound CSB increases. Furthermore, by monitoring chromatin PAR levels using Western blot analysis, we found that CSB sustains the DNA damage signal initiated by PARP1, and may prevent PARP1 overactivation by facilitating DNA repair. By assaying cell viability in response to oxidative stress, we further demonstrate that PARP1 regulation by CSB is a major CSB function in oxidatively-stressed cells. Together, our study uncovers a dynamic interplay between CSB and PARP1 that is critical for oxidative stress relief.

4.
SLAS Discov ; 25(8): 895-905, 2020 09.
Article in English | MEDLINE | ID: mdl-32567455

ABSTRACT

Notch signaling is often involved in cancer cell initiation and proliferation. Aberrant Notch activation underlies more than 50% of T-cell acute lymphoblastic leukemia (T-ALL); accordingly, chemicals disrupting Notch signaling are of potential to treat Notch-dependent cancer. Here, we developed a flow cytometry-based high-throughput assay to identify compounds that disrupt the interactions of DNA and RBPJ, the major downstream effector of Notch signaling. From 1492 compounds, we identified 18 compounds that disrupt RBPJ-DNA interactions in a dose-dependent manner. Cell-based assays further revealed that auranofin downregulates Notch-dependent transcription and decreases RBPJ-chromatin interactions in cells. Most strikingly, T-ALL cells that depend on Notch signaling for proliferation are more sensitive to auranofin treatment, supporting the notion that auranofin downregulates Notch signaling by disrupting RBPJ-DNA interaction. These results validate the feasibility of our assay scheme to screen for additional Notch inhibitors and provide a rationale to further test the use of auranofin in treating Notch-dependent cancer.


Subject(s)
Auranofin/pharmacology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Notch/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/drug effects , DNA/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Flow Cytometry , High-Throughput Screening Assays/methods , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/drug effects , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptors, Notch/genetics , Signal Transduction/drug effects
5.
Nucleic Acids Res ; 47(9): 4521-4538, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30916347

ABSTRACT

Faithful propagation of transcription programs through cell division underlies cell-identity maintenance. Transcriptional regulators selectively bound on mitotic chromatin are emerging critical elements for mitotic transcriptional memory; however, mechanisms governing their site-selective binding remain elusive. By studying how protein-protein interactions impact mitotic chromatin binding of RBPJ, the major downstream effector of the Notch signaling pathway, we found that histone modifying enzymes HDAC1 and KDM5A play critical, regulatory roles in this process. We found that HDAC1 knockdown or inactivation leads to increased RBPJ occupancy on mitotic chromatin in a site-specific manner, with a concomitant increase of KDM5A occupancy at these sites. Strikingly, the presence of KDM5A is essential for increased RBPJ occupancy. Our results uncover a regulatory mechanism in which HDAC1 negatively regulates RBPJ binding on mitotic chromatin in a KDM5A-dependent manner. We propose that relative chromatin affinity of a minimal regulatory complex, reflecting a specific transcription program, renders selective RBPJ binding on mitotic chromatin.


Subject(s)
Chromatin/genetics , Histone Deacetylase 1/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Retinoblastoma-Binding Protein 2/genetics , Animals , Binding Sites , CRISPR-Cas Systems/genetics , Cell Cycle/genetics , Cell Division/genetics , Gene Expression Regulation , Gene Knockout Techniques , Humans , Mice , Mitosis/genetics , Promoter Regions, Genetic , Protein Binding/genetics , Signal Transduction/genetics
6.
J Biol Chem ; 293(46): 17863-17874, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30266807

ABSTRACT

Cockayne syndrome protein B (CSB) is an ATP-dependent chromatin remodeler that relieves oxidative stress by regulating DNA repair and transcription. CSB is proposed to participate in base-excision repair (BER), the primary pathway for repairing oxidative DNA damage, but exactly how CSB participates in this process is unknown. It is also unclear whether CSB contributes to other repair pathways during oxidative stress. Here, using a patient-derived CS1AN-sv cell line, we examined how CSB is targeted to chromatin in response to menadione-induced oxidative stress, both globally and locus-specifically. We found that menadione-induced, global CSB-chromatin association does not require CSB's ATPase activity and is, therefore, mechanistically distinct from UV-induced CSB-chromatin association. Importantly, poly(ADP-ribose) polymerase 1 (PARP1) enhanced the kinetics of global menadione-induced CSB-chromatin association. We found that the major BER enzymes, 8-oxoguanine DNA glycosylase (OGG1) and apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1), do not influence this association. Additionally, the level of γ-H2A histone family member X (γ-H2AX), a marker for dsDNA breaks, was not increased in menadione-treated cells. Therefore, our results support a model whereby PARP1 localizes to ssDNA breaks and recruits CSB to participate in DNA repair. Furthermore, this global CSB-chromatin association occurred independently of RNA polymerase II-mediated transcription elongation. However, unlike global CSB-chromatin association, both PARP1 knockdown and inhibition of transcription elongation interfered with menadione-induced CSB recruitment to specific genomic regions. This observation supports the hypothesis that CSB is also targeted to specific genomic loci to participate in transcriptional regulation in response to oxidative stress.


Subject(s)
Chromatin/metabolism , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA/metabolism , Oxidative Stress/physiology , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , CCCTC-Binding Factor/metabolism , DNA/radiation effects , DNA Damage , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Genetic Loci , Humans , Mutation , Poly-ADP-Ribose Binding Proteins/genetics , Protein Binding , Ultraviolet Rays , Vitamin K 3/pharmacology
7.
Nucleic Acids Res ; 46(15): 7471-7479, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30032309

ABSTRACT

Cockayne syndrome protein B (CSB) is a member of the SNF2/SWI2 ATPase family and is essential for transcription-coupled nucleotide excision DNA repair (TC-NER). CSB also plays critical roles in transcription regulation. CSB can hydrolyze ATP in a DNA-dependent manner, alter protein-DNA contacts and anneal DNA strands. How the different biochemical activities of CSB are utilized in these cellular processes have only begun to become clear in recent years. Mutations in the gene encoding CSB account for majority of the Cockayne syndrome cases, which result in extreme sun sensitivity, premature aging features and/or abnormalities in neurology and development. Here, we summarize and integrate recent biochemical, structural, single-molecule and somatic cell genetic studies that have advanced our understanding of CSB. First, we review studies on the mechanisms that regulate the different biochemical activities of CSB. Next, we summarize how CSB is targeted to regulate transcription under different growth conditions. We then discuss recent advances in our understanding of how CSB regulates transcription mechanistically. Lastly, we summarize the various roles that CSB plays in the different steps of TC-NER, integrating the results of different studies and proposing a model as to how CSB facilitates TC-NER.


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , DNA Repair/genetics , Gene Expression Regulation/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Adenosine Triphosphate/metabolism , DNA/genetics , Humans , Mutation/genetics , Schizosaccharomyces/genetics , Transcription, Genetic/genetics
8.
Nucleic Acids Res ; 45(8): 4696-4707, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28369616

ABSTRACT

Cockayne syndrome protein B (CSB) belongs to the SWI2/SNF2 ATP-dependent chromatin remodeler family, and CSB is the only ATP-dependent chromatin remodeler essential for transcription-coupled nucleotide excision DNA repair. CSB alone remodels nucleosomes ∼10-fold slower than the ACF remodeling complex. Strikingly, NAP1-like histone chaperones interact with CSB and greatly enhance CSB-mediated chromatin remodeling. While chromatin remodeling by CSB and NAP1-like proteins is crucial for efficient transcription-coupled DNA repair, the mechanism by which NAP1-like proteins enhance chromatin remodeling by CSB remains unknown. Here we studied CSB's DNA-binding and nucleosome-remodeling activities at the single molecule level in real time. We also determined how the NAP1L1 chaperone modulates these activities. We found that CSB interacts with DNA in two principle ways: by simple binding and a more complex association that involves gross DNA distortion. Remarkably, NAP1L1 suppresses both these interactions. Additionally, we demonstrate that nucleosome remodeling by CSB consists of three distinct phases: activation, translocation and pausing, similar to ACF. Importantly, we found that NAP1L1 promotes CSB-mediated remodeling by accelerating both activation and translocation. Additionally, NAP1L1 increases CSB processivity by decreasing the pausing probability during translocation. Our study, therefore, uncovers the different steps of CSB-mediated chromatin remodeling that can be regulated by NAP1L1.


Subject(s)
DNA Helicases/genetics , DNA Repair Enzymes/genetics , Histone Chaperones/genetics , Nucleosome Assembly Protein 1/genetics , Transcription, Genetic , Adenosine Triphosphate/metabolism , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , DNA Repair/genetics , Humans , Nucleosomes/genetics , Poly-ADP-Ribose Binding Proteins
9.
Nucleic Acids Res ; 44(5): 2125-35, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26578602

ABSTRACT

Cockayne syndrome is a premature aging disease associated with numerous developmental and neurological abnormalities, and elevated levels of reactive oxygen species have been found in cells derived from Cockayne syndrome patients. The majority of Cockayne syndrome cases contain mutations in the ATP-dependent chromatin remodeler CSB; however, how CSB protects cells from oxidative stress remains largely unclear. Here, we demonstrate that oxidative stress alters the genomic occupancy of the CSB protein and increases CSB occupancy at promoters. Additionally, we found that the long-range chromatin-structure regulator CTCF plays a pivotal role in regulating sites of genomic CSB occupancy upon oxidative stress. We show that CSB directly interacts with CTCF in vitro and that oxidative stress enhances the CSB-CTCF interaction in cells. Reciprocally, we demonstrate that CSB facilitates CTCF-DNA interactions in vitro and regulates CTCF-chromatin interactions in oxidatively stressed cells. Together, our results indicate that CSB and CTCF can regulate each other's chromatin association, thereby modulating chromatin structure and coordinating gene expression in response to oxidative stress.


Subject(s)
Chromatin/chemistry , DNA Helicases/genetics , DNA Repair Enzymes/genetics , DNA/genetics , Fibroblasts/metabolism , Repressor Proteins/genetics , Base Sequence , Binding Sites , CCCTC-Binding Factor , Cell Line, Transformed , Chromatin/drug effects , Chromatin/metabolism , Chromatin Assembly and Disassembly/drug effects , DNA/metabolism , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation , Humans , Molecular Sequence Data , Oxidative Stress , Poly-ADP-Ribose Binding Proteins , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Signal Transduction , Vitamin K 3/pharmacology
10.
RNA ; 21(12): 2053-66, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26437669

ABSTRACT

HnRNP L is a ubiquitous splicing-regulatory protein that is critical for the development and function of mammalian T cells. Previous work has identified a few targets of hnRNP L-dependent alternative splicing in T cells and has described transcriptome-wide association of hnRNP L with RNA. However, a comprehensive analysis of the impact of hnRNP L on mRNA expression remains lacking. Here we use next-generation sequencing to identify transcriptome changes upon depletion of hnRNP L in a model T-cell line. We demonstrate that hnRNP L primarily regulates cassette-type alternative splicing, with minimal impact of hnRNP L depletion on transcript abundance, intron retention, or other modes of alternative splicing. Strikingly, we find that binding of hnRNP L within or flanking an exon largely correlates with exon repression by hnRNP L. In contrast, exons that are enhanced by hnRNP L generally lack proximal hnRNP L binding. Notably, these hnRNP L-enhanced exons share sequence and context features that correlate with poor nucleosome positioning, suggesting that hnRNP may enhance inclusion of a subset of exons via a cotranscriptional or epigenetic mechanism. Our data demonstrate that hnRNP L controls inclusion of a broad spectrum of alternative cassette exons in T cells and suggest both direct RNA regulation as well as indirect mechanisms sensitive to the epigenetic landscape.


Subject(s)
Epigenesis, Genetic , RNA, Messenger/metabolism , Ribonucleoproteins/physiology , Alternative Splicing , Exons , Humans , Jurkat Cells , Nucleosomes/metabolism , RNA, Messenger/genetics , Transcriptome
12.
PLoS Genet ; 10(4): e1004284, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24743307

ABSTRACT

Cockayne syndrome is an inherited premature aging disease associated with numerous developmental and neurological defects, and mutations in the gene encoding the CSB protein account for the majority of Cockayne syndrome cases. Accumulating evidence suggests that CSB functions in transcription regulation, in addition to its roles in DNA repair, and those defects in this transcriptional activity might contribute to the clinical features of Cockayne syndrome. Transcription profiling studies have so far uncovered CSB-dependent effects on gene expression; however, the direct targets of CSB's transcriptional activity remain largely unknown. In this paper, we report the first comprehensive analysis of CSB genomic occupancy during replicative cell growth. We found that CSB occupancy sites display a high correlation to regions with epigenetic features of promoters and enhancers. Furthermore, we found that CSB occupancy is enriched at sites containing the TPA-response element. Consistent with this binding site preference, we show that CSB and the transcription factor c-Jun can be found in the same protein-DNA complex, suggesting that c-Jun can target CSB to specific genomic regions. In support of this notion, we observed decreased CSB occupancy of TPA-response elements when c-Jun levels were diminished. By modulating CSB abundance, we found that CSB can influence the expression of nearby genes and impact nucleosome positioning in the vicinity of its binding site. These results indicate that CSB can be targeted to specific genomic loci by sequence-specific transcription factors to regulate transcription and local chromatin structure. Additionally, comparison of CSB occupancy sites with the MSigDB Pathways database suggests that CSB might function in peroxisome proliferation, EGF receptor transactivation, G protein signaling and NF-κB activation, shedding new light on the possible causes and mechanisms of Cockayne syndrome.


Subject(s)
Chromatin/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , Cell Line , Chromatin/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Poly-ADP-Ribose Binding Proteins , Promoter Regions, Genetic/genetics
13.
PLoS Genet ; 10(3): e1004204, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24603501

ABSTRACT

Mechanisms that maintain transcriptional memory through cell division are important to maintain cell identity, and sequence-specific transcription factors that remain associated with mitotic chromatin are emerging as key players in transcriptional memory propagation. Here, we show that the major transcriptional effector of Notch signaling, RBPJ, is retained on mitotic chromatin, and that this mitotic chromatin association is mediated through the direct association of RBPJ with DNA. We further demonstrate that RBPJ binds directly to nucleosomal DNA in vitro, with a preference for sites close to the entry/exit position of the nucleosomal DNA. Genome-wide analysis in the murine embryonal-carcinoma cell line F9 revealed that roughly 60% of the sites occupied by RBPJ in asynchronous cells were also occupied in mitotic cells. Among them, we found that a fraction of RBPJ occupancy sites shifted between interphase and mitosis, suggesting that RBPJ can be retained on mitotic chromatin by sliding on DNA rather than disengaging from chromatin during mitotic chromatin condensation. We propose that RBPJ can function as a mitotic bookmark, marking genes for efficient transcriptional activation or repression upon mitotic exit. Strikingly, we found that sites of RBPJ occupancy were enriched for CTCF-binding motifs in addition to RBPJ-binding motifs, and that RBPJ and CTCF interact. Given that CTCF regulates transcription and bridges long-range chromatin interactions, our results raise the intriguing hypothesis that by collaborating with CTCF, RBPJ may participate in establishing chromatin domains and/or long-range chromatin interactions that could be propagated through cell division to maintain gene expression programs.


Subject(s)
Chromatin/genetics , DNA/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mitosis/genetics , Animals , CCCTC-Binding Factor , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Interphase/genetics , Mice , Nucleosomes/genetics , Nucleosomes/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/genetics
14.
PLoS Genet ; 9(4): e1003407, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23637612

ABSTRACT

The Cockayne syndrome complementation group B (CSB) protein is essential for transcription-coupled DNA repair, and mutations in CSB are associated with Cockayne syndrome--a devastating disease with complex clinical features, including the appearance of premature aging, sun sensitivity, and numerous neurological and developmental defects. CSB belongs to the SWI2/SNF2 ATP-dependent chromatin remodeler family, but the extent to which CSB remodels chromatin and whether this activity is utilized in DNA repair is unknown. Here, we show that CSB repositions nucleosomes in an ATP-dependent manner in vitro and that this activity is greatly enhanced by the NAP1-like histone chaperones, which we identify as new CSB-binding partners. By mapping functional domains and analyzing CSB derivatives, we demonstrate that chromatin remodeling by the combined activities of CSB and the NAP1-like chaperones is required for efficient transcription-coupled DNA repair. Moreover, we show that chromatin remodeling and repair protein recruitment mediated by CSB are separable activities. The collaboration that we observed between CSB and the NAP1-like histone chaperones adds a new dimension to our understanding of the ways in which ATP-dependent chromatin remodelers and histone chaperones can regulate chromatin structure. Taken together, the results of this study offer new insights into the functions of chromatin remodeling by CSB in transcription-coupled DNA repair as well as the underlying mechanisms of Cockayne syndrome.


Subject(s)
Chromatin Assembly and Disassembly , Cockayne Syndrome , Adenosine Triphosphate/metabolism , Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair , DNA Repair Enzymes/genetics , Histone Chaperones/genetics , Humans , Transcription, Genetic
15.
Mech Ageing Dev ; 134(5-6): 202-11, 2013.
Article in English | MEDLINE | ID: mdl-23422418

ABSTRACT

The Cockayne syndrome complementation group B protein, CSB, plays pivotal roles in transcription regulation and DNA repair. CSB belongs to the SNF2/SWI2 ATP-dependent chromatin remodeling protein family, and studies from many laboratories have revealed that CSB has multiple activities and modes of regulation. To understand the underlying mechanisms of Cockayne syndrome, it is necessary to understand how the biochemical activities of CSB are used to carry out its biological functions. In this review, we summarize our current knowledge of the structure, function and regulation of CSB, and discuss how these properties can impact the biological functions of this chromatin remodeler.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA Repair/physiology , Transcription, Genetic/physiology , Animals , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , Humans , Poly-ADP-Ribose Binding Proteins , Structure-Activity Relationship
16.
J Biol Chem ; 286(40): 34951-8, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21852235

ABSTRACT

The Cockayne syndrome complementation group B (CSB) protein is an ATP-dependent chromatin remodeler with an essential function in transcription-coupled DNA repair, and mutations in the CSB gene are associated with Cockayne syndrome. The p53 tumor suppressor has been known to interact with CSB, and both proteins have been implicated in overlapping biological processes, such as DNA repair and aging. The significance of the interaction between CSB and p53 has remained unclear, however. Here, we show that the chromatin association of CSB and p53 is inversely related. Using in vitro binding and chromatin immunoprecipitation approaches, we demonstrate that CSB facilitates the sequence-independent association of p53 with chromatin when p53 concentrations are low and that this is achieved by the interaction of CSB with the C-terminal region of p53. Remarkably, p53 prevents CSB from binding to nucleosomes when p53 concentrations are elevated. Examining the enzymatic properties of CSB revealed that p53 excludes CSB from nucleosomes by occluding a nucleosome interaction surface on CSB. Together, our results suggest that the reciprocal regulation of chromatin access by CSB and p53 could be part of a mechanism by which these two proteins coordinate their activities to regulate DNA repair, cell survival, and aging.


Subject(s)
DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Gene Expression Regulation , Tumor Suppressor Protein p53/metabolism , Adenosine Triphosphate/metabolism , Apoptosis , Cell Survival , Cellular Senescence , Chromatin/metabolism , Cockayne Syndrome/genetics , DNA Repair , Fibroblasts/metabolism , Genetic Complementation Test , Genome , Humans , Models, Biological , Models, Genetic , Nucleosomes/metabolism , Poly-ADP-Ribose Binding Proteins
17.
J Cell Sci ; 124(Pt 7): 1067-76, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21402876

ABSTRACT

Notch receptors and their ligands have crucial roles in development and tumorigenesis. We present evidence demonstrating the existence of an antagonistic relationship between Notch 4 and Trp53, which is controlled by the Mdm2-dependent ubiquitylation and degradation of the Notch receptor. We show that this signal-controlling mechanism is mediated by physical interactions between Mdm2 and Notch 4 and suggest the existence of a trimeric complex between Trp53, Notch 4 and Mdm2, which ultimately regulates Notch activity. Functional studies indicate that Trp53 can suppress NICD4-induced anchorage-independent growth in mammary epithelial cells and present evidence showing that Trp53 has a pivotal role in the suppression of Notch-associated tumorigenesis in the mammary gland.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Humans , Mice , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Receptor, Notch4 , Receptors, Notch/chemistry , Receptors, Notch/genetics , Tumor Suppressor Protein p53/genetics
18.
Vaccine ; 29(17): 3111-7, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21376119

ABSTRACT

OBJECTIVES: To investigate the views of students, support staff and academic staff at the University of Alberta in Edmonton, Canada on the allocation of scarce resources during an influenza pandemic to discover if there were any shared values. METHODS: A web-based questionnaire was circulated to students, support staff and academic staff asking them how they would rank the priority of eleven different groups for access to scarce resources. They were also asked to select one of seven priority access plans. RESULTS: The highest priority was given to health care workers by 89% of respondents, closely followed by emergency workers (85%). Only 12.7% of respondents gave politicians high priority. Respondents favored the "Save the most lives" priority access (39.9%) (N=5220). CONCLUSION: Current policies in place for the allocation of scarce resources during an influenza pandemic may not properly reflect the views of the general public. Further public consultation should be undertaken in order to uncover how they would allocate scarce resources.


Subject(s)
Health Care Rationing/ethics , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Pandemics/prevention & control , Adolescent , Adult , Aged , Aged, 80 and over , Alberta , Child , Child, Preschool , Disaster Planning , Female , Humans , Infant , Infant, Newborn , Influenza, Human/diagnosis , Influenza, Human/drug therapy , Male , Middle Aged , Surveys and Questionnaires , Universities , Young Adult
19.
BMC Dev Biol ; 10: 107, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20959007

ABSTRACT

BACKGROUND: The Notch signaling pathway regulates a diverse array of developmental processes, and aberrant Notch signaling can lead to diseases, including cancer. To obtain a more comprehensive understanding of the genetic network that integrates into Notch signaling, we performed a genome-wide RNAi screen in Drosophila cell culture to identify genes that modify Notch-dependent transcription. RESULTS: Employing complementary data analyses, we found 399 putative modifiers: 189 promoting and 210 antagonizing Notch activated transcription. These modifiers included several known Notch interactors, validating the robustness of the assay. Many novel modifiers were also identified, covering a range of cellular localizations from the extracellular matrix to the nucleus, as well as a large number of proteins with unknown function. Chromatin-modifying proteins represent a major class of genes identified, including histone deacetylase and demethylase complex components and other chromatin modifying, remodeling and replacement factors. A protein-protein interaction map of the Notch-dependent transcription modifiers revealed that a large number of the identified proteins interact physically with these core chromatin components. CONCLUSIONS: The genome-wide RNAi screen identified many genes that can modulate Notch transcriptional output. A protein interaction map of the identified genes highlighted a network of chromatin-modifying enzymes and remodelers that regulate Notch transcription. Our results open new avenues to explore the mechanisms of Notch signal regulation and the integration of this pathway into diverse cellular processes.


Subject(s)
Genome , RNA Interference , Receptors, Notch/metabolism , Signal Transduction/genetics , Transcription, Genetic , Animals , Cell Line , Chromatin/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epistasis, Genetic , Mutation , Protein Interaction Mapping , Receptors, Notch/genetics , Ribosomes/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
20.
PLoS One ; 5(5): e10532, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20479883

ABSTRACT

BACKGROUND: The Notch receptor links cell fate decisions of one cell to that of the immediate cellular neighbor. In humans, malfunction of Notch signaling results in diseases and congenital disorders. Structural information is essential for gaining insight into the mechanism of the receptor as well as for potentially interfering with its function for therapeutic purposes. METHODOLOGY/PRINCIPAL FINDINGS: We used the Affinity Grid approach to prepare specimens of the Notch extracellular domain (NECD) of the Drosophila Notch and human Notch1 receptors suitable for analysis by electron microscopy and three-dimensional (3D) image reconstruction. The resulting 3D density maps reveal that the NECD structure is conserved across species. We show that the NECD forms a dimer and adopts different yet defined conformations, and we identify the membrane-proximal region of the receptor and its ligand-binding site. CONCLUSIONS/SIGNIFICANCE: Our results provide direct and unambiguous evidence that the NECD forms a dimer. Our studies further show that the NECD adopts at least three distinct conformations that are likely related to different functional states of the receptor. These findings open the way to now correlate mutations in the NECD with its oligomeric state and conformation.


Subject(s)
Drosophila melanogaster/metabolism , Extracellular Space/chemistry , Protein Multimerization , Receptors, Notch/chemistry , Receptors, Notch/ultrastructure , Animals , Humans , Microscopy, Electron , Protein Structure, Tertiary , Receptors, Notch/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...