89 results on '"Oliver AW"'
Search Results
2. The ASCIZ-DYNLL1 axis promotes 53BP1-dependent non-homologous end joining and PARP inhibitor sensitivity
- Author
-
Becker, JR, Cuella-Martin, R, Barazas, M, Liu, R, Oliveira, C, Oliver, AW, Bilham, K, Holt, AB, Blackford, AN, Heierhorst, J, Jonkers, J, Rottenberg, S, Chapman, JR, Becker, JR, Cuella-Martin, R, Barazas, M, Liu, R, Oliveira, C, Oliver, AW, Bilham, K, Holt, AB, Blackford, AN, Heierhorst, J, Jonkers, J, Rottenberg, S, and Chapman, JR
- Abstract
53BP1 controls a specialized non-homologous end joining (NHEJ) pathway that is essential for adaptive immunity, yet oncogenic in BRCA1 mutant cancers. Intra-chromosomal DNA double-strand break (DSB) joining events during immunoglobulin class switch recombination (CSR) require 53BP1. However, in BRCA1 mutant cells, 53BP1 blocks homologous recombination (HR) and promotes toxic NHEJ, resulting in genomic instability. Here, we identify the protein dimerization hub-DYNLL1-as an organizer of multimeric 53BP1 complexes. DYNLL1 binding stimulates 53BP1 oligomerization, and promotes 53BP1's recruitment to, and interaction with, DSB-associated chromatin. Consequently, DYNLL1 regulates 53BP1-dependent NHEJ: CSR is compromised upon deletion of Dynll1 or its transcriptional regulator Asciz, or by mutation of DYNLL1 binding motifs in 53BP1; furthermore, Brca1 mutant cells and tumours are rendered resistant to poly-ADP ribose polymerase (PARP) inhibitor treatments upon deletion of Dynll1 or Asciz. Thus, our results reveal a mechanism that regulates 53BP1-dependent NHEJ and the therapeutic response of BRCA1-deficient cancers.
- Published
- 2018
3. Targeting activated Rho proteins: a new approach for treatment of HPV and other virus-related cancers?
- Author
-
Hampson IN, Oliver AW, and Hampson L
- Published
- 2011
- Full Text
- View/download PDF
4. Update on Effects of the Prophylactic HPV Vaccines on HPV Type Prevalence and Cervical Pathology.
- Author
-
Hampson IN and Oliver AW
- Subjects
- Humans, Female, Prevalence, Cervix Uteri virology, Cervix Uteri pathology, Vaccination, Papillomaviridae immunology, Papillomaviridae classification, Papillomaviridae genetics, Adolescent, Vaccine Efficacy, Papillomavirus Infections prevention & control, Papillomavirus Infections epidemiology, Papillomavirus Infections virology, Papillomavirus Vaccines immunology, Papillomavirus Vaccines administration & dosage, Uterine Cervical Neoplasms prevention & control, Uterine Cervical Neoplasms virology, Uterine Cervical Neoplasms epidemiology
- Abstract
Most national prophylactic HPV vaccination programs started in approximately 2008, with either the bivalent Cervarix HPV16/18 or quadrivalent Gardasil (HPV6/11/16/18) vaccines, which were then followed by introduction of the nonavalent Gardasil 9 (HPV6/11/16/18/ 31/33/45/52/58) vaccine from 2015. Since that time, these products have demonstrated their ability to prevent infection with vaccine-covered HPV types and subsequent development of HPV-related cervical and genital pathologies. The data indicate that vaccination of young girls prior to sexual debut is more effective than vaccination of older HPV+ve women. Although some studies have shown a decline in the prevalence of vaccine-covered HPV types, there are national and regional differences in overall vaccine efficacy. Furthermore, several recently published studies show an increase in the prevalence of non-vaccine-covered HPV types in vaccinated populations, which is indicative of HPV type-replacement. It is also notable that vaccine-related changes in HPV type prevalence spread between vaccinated and unvaccinated women at the same geographical location-presumably via sexual transmission. In conclusion, it is not yet clear what effect dissemination of vaccine-associated changes in HPV type prevalence will have on vaccine efficacy and cervical pathology, particularly in mixed populations of vaccinated and unvaccinated women. However, it is very clear these observations do underscore the need for long-term continuation of cervical screening combined with regular reassessment of testing practices.
- Published
- 2024
- Full Text
- View/download PDF
5. Binding of the TRF2 iDDR motif to RAD50 highlights a convergent evolutionary strategy to inactivate MRN at telomeres.
- Author
-
Khayat F, Alshmery M, Pal M, Oliver AW, and Bianchi A
- Subjects
- Humans, MRE11 Homologue Protein metabolism, MRE11 Homologue Protein genetics, Evolution, Molecular, DNA Breaks, Double-Stranded, Amino Acid Motifs, Nuclear Proteins metabolism, Nuclear Proteins genetics, Cell Cycle Proteins metabolism, Cell Cycle Proteins genetics, Binding Sites, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins genetics, Carrier Proteins metabolism, Carrier Proteins genetics, Telomeric Repeat Binding Protein 2 metabolism, Telomeric Repeat Binding Protein 2 genetics, Telomere metabolism, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, Acid Anhydride Hydrolases metabolism, DNA Repair Enzymes metabolism, DNA Repair Enzymes genetics, Telomere-Binding Proteins metabolism, Telomere-Binding Proteins genetics, Protein Binding
- Abstract
Telomeres protect chromosome ends from unscheduled DNA repair, including from the MRN (MRE11, RAD50, NBS1) complex, which processes double-stranded DNA breaks (DSBs) via activation of the ATM kinase, promotes DNA end-tethering aiding the non-homologous end-joining (NHEJ) pathway, and initiates DSB resection through the MRE11 nuclease. A protein motif (MIN, for MRN inhibitor) inhibits MRN at budding yeast telomeres by binding to RAD50 and evolved at least twice, in unrelated telomeric proteins Rif2 and Taz1. We identify the iDDR motif of human shelterin protein TRF2 as a third example of convergent evolution for this telomeric mechanism for binding MRN, despite the iDDR lacking sequence homology to the MIN motif. CtIP is required for activation of MRE11 nuclease action, and we provide evidence for binding of a short C-terminal region of CtIP to a RAD50 interface that partly overlaps with the iDDR binding site, indicating that the interaction is mutually exclusive. In addition, we show that the iDDR impairs the DNA binding activity of RAD50. These results highlight direct inhibition of MRN action as a crucial role of telomeric proteins across organisms and point to multiple mechanisms enforced by the iDDR to disable the many activities of the MRN complex., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
6. Design and synthesis of quinazolin-4-one derivatives as potential anticancer agents and investigation of their interaction with RecQ helicases.
- Author
-
Haggag HS, Aboukhatwa SM, Nafie MS, Paul A, Sharafeldin N, Oliver AW, and El-Hamamsy MH
- Subjects
- Molecular Docking Simulation, Quinazolinones pharmacology, Adenosine Triphosphate, RecQ Helicases metabolism, Antineoplastic Agents pharmacology
- Abstract
The upregulation of RecQ helicases has been associated with cancer cell survival and resistance to chemotherapy, making them appealing targets for therapeutic intervention. In this study, twenty-nine novel quinazolinone derivatives were designed and synthesized. The anti-proliferative activity of all compounds was evaluated against 60 cancer cell lines at the National Cancer Institute Developmental Therapeutic Program, with six compounds (11f, 11g, 11k, 11n, 11p, and 11q) being promoted to a five-dose screen. Compound 11g demonstrated high cytotoxic activity against all examined cell lines. The compounds were further assayed for Bloom syndrome (BLM) helicase inhibition, where 11g, 11q, and 11u showed moderate activity. These compounds were counter-screened against WRN and RECQ1 helicases, where 11g moderately inhibited both enzymes. An ATP competition assay confirmed that the compounds bound to the ATP site of RecQ helicases, and molecular docking simulations were used to study the binding mode within the active site of BLM, WRN, and RECQ1 helicases. Compound 11g induced apoptosis in both HCT-116 and MDA-MB-231 cell lines, but also caused an G2/M phase cell cycle arrest in HCT-116 cells. This data revealed the potential of 11g as a modulator of cell cycle dynamics and supports its interaction with RecQ helicases. In addition, compound 11g displayed non-significant toxicity against FCH normal colon cells at doses up to 100 µM, which confirming its high safety margin and selectivity on cancer cells. Overall, these findings suggest compound 11g as a potential pan RecQ helicase inhibitor with high anticancer potency and a favorable safety margin and selectivity., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. TopBP1 utilises a bipartite GINS binding mode to support genome replication.
- Author
-
Day M, Tetik B, Parlak M, Almeida-Hernández Y, Räschle M, Kaschani F, Siegert H, Marko A, Sanchez-Garcia E, Kaiser M, Barker IA, Pearl LH, Oliver AW, and Boos D
- Subjects
- Animals, DNA Polymerase II metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Minichromosome Maintenance Proteins metabolism, Virus Replication, DNA Replication, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism
- Abstract
Activation of the replicative Mcm2-7 helicase by loading GINS and Cdc45 is crucial for replication origin firing, and as such for faithful genetic inheritance. Our biochemical and structural studies demonstrate that the helicase activator GINS interacts with TopBP1 through two separate binding surfaces, the first involving a stretch of highly conserved amino acids in the TopBP1-GINI region, the second a surface on TopBP1-BRCT4. The two surfaces bind to opposite ends of the A domain of the GINS subunit Psf1. Mutation analysis reveals that either surface is individually able to support TopBP1-GINS interaction, albeit with reduced affinity. Consistently, either surface is sufficient for replication origin firing in Xenopus egg extracts and becomes essential in the absence of the other. The TopBP1-GINS interaction appears sterically incompatible with simultaneous binding of DNA polymerase epsilon (Polε) to GINS when bound to Mcm2-7-Cdc45, although TopBP1-BRCT4 and the Polε subunit PolE2 show only partial competitivity in binding to Psf1. Our TopBP1-GINS model improves the understanding of the recently characterised metazoan pre-loading complex. It further predicts the coordination of three molecular origin firing processes, DNA polymerase epsilon arrival, TopBP1 ejection and GINS integration into Mcm2-7-Cdc45., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
8. CC-seq: Nucleotide-Resolution Mapping of Spo11 DNA Double-Strand Breaks in S. cerevisiae Cells.
- Author
-
Brown GGB, Gittens WH, Allison RM, Oliver AW, and Neale MJ
- Subjects
- Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, DNA, Fungal genetics, DNA, Fungal metabolism, Sequence Analysis, DNA methods, DNA Repair, Saccharomyces cerevisiae genetics, DNA Breaks, Double-Stranded, Endodeoxyribonucleases metabolism, Endodeoxyribonucleases genetics, Meiosis genetics
- Abstract
During meiosis, Spo11 generates DNA double-strand breaks to induce recombination, becoming covalently attached to the 5' ends on both sides of the break during this process. Such Spo11 "covalent complexes" are transient in wild-type cells, but accumulate in nuclease mutants unable to initiate repair. The CC-seq method presented here details how to map the location of these Spo11 complexes genome-wide with strand-specific nucleotide-resolution accuracy in synchronized Saccharomyces cerevisiae meiotic cells., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
9. Schizosaccharomyces pombe Rtf2 is important for replication fork barrier activity of RTS1 via splicing of Rtf1 .
- Author
-
Budden AM, Eravci M, Watson AT, Campillo-Funollet E, Oliver AW, Naiman K, and Carr AM
- Subjects
- Humans, RNA Splicing, RNA Processing, Post-Transcriptional, Introns, DNA Replication genetics, Schizosaccharomyces genetics
- Abstract
Arrested replication forks, when restarted by homologous recombination, result in error-prone DNA syntheses and non-allelic homologous recombination. Fission yeast RTS1 is a model fork barrier used to probe mechanisms of recombination-dependent restart. RTS1 barrier activity is entirely dependent on the DNA binding protein Rtf1 and partially dependent on a second protein, Rtf2. Human RTF2 was recently implicated in fork restart, leading us to examine fission yeast Rtf2's role in more detail. In agreement with previous studies, we observe reduced barrier activity upon rtf2 deletion. However, we identified Rtf2 to be physically associated with mRNA processing and splicing factors and rtf2 deletion to cause increased intron retention. One of the most affected introns resided in the rtf1 transcript. Using an intronless rtf1, we observed no reduction in RFB activity in the absence of Rtf2. Thus, Rtf2 is essential for correct rtf1 splicing to allow optimal RTS1 barrier activity., Competing Interests: AB, ME, AW, EC, AO, KN, AC No competing interests declared, (© 2023, Budden et al.)
- Published
- 2023
- Full Text
- View/download PDF
10. Structural basis for the inactivation of cytosolic DNA sensing by the vaccinia virus.
- Author
-
Rivera-Calzada A, Arribas-Bosacoma R, Ruiz-Ramos A, Escudero-Bravo P, Boskovic J, Fernandez-Leiro R, Oliver AW, Pearl LH, and Llorca O
- Subjects
- Ku Autoantigen metabolism, DNA metabolism, DNA-Activated Protein Kinase metabolism, Vaccinia virus genetics, DNA-Binding Proteins metabolism
- Abstract
Detection of cytosolic DNA is a central element of the innate immunity system against viral infection. The Ku heterodimer, a component of the NHEJ pathway of DNA repair in the nucleus, functions as DNA sensor that detects dsDNA of viruses that replicate in the cytoplasm. Vaccinia virus expresses two proteins, C4 and C16, that inactivate DNA sensing and enhance virulence. The structural basis for this is unknown. Here we determine the structure of the C16 - Ku complex using cryoEM. Ku binds dsDNA by a preformed ring but C16 sterically blocks this access route, abrogating binding to a dsDNA end and its insertion into DNA-PK, thereby averting signalling into the downstream innate immunity system. C4 replicates these activities using a domain with 54% identity to C16. Our results reveal how vaccinia virus subverts the capacity of Ku to recognize viral DNA., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
11. Pathogenic variants in SLF2 and SMC5 cause segmented chromosomes and mosaic variegated hyperploidy.
- Author
-
Grange LJ, Reynolds JJ, Ullah F, Isidor B, Shearer RF, Latypova X, Baxley RM, Oliver AW, Ganesh A, Cooke SL, Jhujh SS, McNee GS, Hollingworth R, Higgs MR, Natsume T, Khan T, Martos-Moreno GÁ, Chupp S, Mathew CG, Parry D, Simpson MA, Nahavandi N, Yüksel Z, Drasdo M, Kron A, Vogt P, Jonasson A, Seth SA, Gonzaga-Jauregui C, Brigatti KW, Stegmann APA, Kanemaki M, Josifova D, Uchiyama Y, Oh Y, Morimoto A, Osaka H, Ammous Z, Argente J, Matsumoto N, Stumpel CTRM, Taylor AMR, Jackson AP, Bielinsky AK, Mailand N, Le Caignec C, Davis EE, and Stewart GS
- Subjects
- Humans, DNA Repair genetics, Chromosomes metabolism, Genomic Instability, DNA-Binding Proteins metabolism, Ubiquitin-Protein Ligases metabolism, Chromosomal Proteins, Non-Histone metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Microcephaly genetics
- Abstract
Embryonic development is dictated by tight regulation of DNA replication, cell division and differentiation. Mutations in DNA repair and replication genes disrupt this equilibrium, giving rise to neurodevelopmental disease characterized by microcephaly, short stature and chromosomal breakage. Here, we identify biallelic variants in two components of the RAD18-SLF1/2-SMC5/6 genome stability pathway, SLF2 and SMC5, in 11 patients with microcephaly, short stature, cardiac abnormalities and anemia. Patient-derived cells exhibit a unique chromosomal instability phenotype consisting of segmented and dicentric chromosomes with mosaic variegated hyperploidy. To signify the importance of these segmented chromosomes, we have named this disorder Atelís (meaning - incomplete) Syndrome. Analysis of Atelís Syndrome cells reveals elevated levels of replication stress, partly due to a reduced ability to replicate through G-quadruplex DNA structures, and also loss of sister chromatid cohesion. Together, these data strengthen the functional link between SLF2 and the SMC5/6 complex, highlighting a distinct role for this pathway in maintaining genome stability., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
12. Cryo-EM structure of the Smc5/6 holo-complex.
- Author
-
Hallett ST, Campbell Harry I, Schellenberger P, Zhou L, Cronin NB, Baxter J, Etheridge TJ, Murray JM, and Oliver AW
- Subjects
- Nuclear Proteins metabolism, Cryoelectron Microscopy, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Cell Cycle Proteins metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
The Smc5/6 complex plays an essential role in the resolution of recombination intermediates formed during mitosis or meiosis, or as a result of the cellular response to replication stress. It also functions as a restriction factor preventing viral replication. Here, we report the cryogenic EM (cryo-EM) structure of the six-subunit budding yeast Smc5/6 holo-complex, reconstituted from recombinant proteins expressed in insect cells - providing both an architectural overview of the entire complex and an understanding of how the Nse1/3/4 subcomplex binds to the hetero-dimeric SMC protein core. In addition, we demonstrate that a region within the head domain of Smc5, equivalent to the 'W-loop' of Smc4 or 'F-loop' of Smc1, mediates an important interaction with Nse1. Notably, mutations that alter the surface-charge profile of the region of Nse1 which accepts the Smc5-loop, lead to a slow-growth phenotype and a global reduction in the chromatin-associated fraction of the Smc5/6 complex, as judged by single molecule localisation microscopy experiments in live yeast. Moreover, when taken together, our data indicates functional equivalence between the structurally unrelated KITE and HAWK accessory subunits associated with SMC complexes., (© The Author(s) 2022. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
13. Structure of the human RAD17-RFC clamp loader and 9-1-1 checkpoint clamp bound to a dsDNA-ssDNA junction.
- Author
-
Day M, Oliver AW, and Pearl LH
- Subjects
- DNA chemistry, DNA Damage, Humans, Replication Protein C metabolism, Cell Cycle Proteins metabolism, DNA, Single-Stranded genetics
- Abstract
The RAD9-RAD1-HUS1 (9-1-1) clamp forms one half of the DNA damage checkpoint system that signals the presence of substantial regions of single-stranded DNA arising from replication fork collapse or resection of DNA double strand breaks. Loaded at the 5'-recessed end of a dsDNA-ssDNA junction by the RAD17-RFC clamp loader complex, the phosphorylated C-terminal tail of the RAD9 subunit of 9-1-1 engages with the mediator scaffold TOPBP1 which in turn activates the ATR kinase, localised through the interaction of its constitutive partner ATRIP with RPA-coated ssDNA. Using cryogenic electron microscopy (cryoEM) we have determined the structure of a complex of the human RAD17-RFC clamp loader bound to human 9-1-1, engaged with a dsDNA-ssDNA junction. The structure answers the key questions of how RAD17 confers specificity for 9-1-1 over PCNA, and how the clamp loader specifically recognises the recessed 5' DNA end and fixes the orientation of 9-1-1 on the ssDNA., (© The Author(s) 2022. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
14. Phosphorylation-dependent assembly of DNA damage response systems and the central roles of TOPBP1.
- Author
-
Day M, Oliver AW, and Pearl LH
- Subjects
- Carrier Proteins metabolism, Cell Cycle Proteins metabolism, DNA Damage, DNA Repair, Phosphorylation, DNA-Binding Proteins metabolism, Nuclear Proteins metabolism
- Abstract
The cellular response to DNA damage (DDR) that causes replication collapse and/or DNA double strand breaks, is characterised by a massive change in the post-translational modifications (PTM) of hundreds of proteins involved in the detection and repair of DNA damage, and the communication of the state of damage to the cellular systems that regulate replication and cell division. A substantial proportion of these PTMs involve targeted phosphorylation, which among other effects, promotes the formation of multiprotein complexes through the specific binding of phosphorylated motifs on one protein, by specialised domains on other proteins. Understanding the nature of these phosphorylation mediated interactions allows definition of the pathways and networks that coordinate the DDR, and helps identify new targets for therapeutic intervention that may be of benefit in the treatment of cancer, where DDR plays a key role. In this review we summarise the present understanding of how phosphorylated motifs are recognised by BRCT domains, which occur in many DDR proteins. We particularly focus on TOPBP1 - a multi-BRCT domain scaffold protein with essential roles in replication and the repair and signalling of DNA damage., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
15. CK2 Phosphorylation of Human Papillomavirus 16 E2 on Serine 23 Promotes Interaction with TopBP1 and Is Critical for E2 Interaction with Mitotic Chromatin and the Viral Life Cycle.
- Author
-
Prabhakar AT, James CD, Das D, Otoa R, Day M, Burgner J, Fontan CT, Wang X, Glass SH, Wieland A, Donaldson MM, Bristol ML, Li R, Oliver AW, Pearl LH, Smith BO, and Morgan IM
- Subjects
- Carrier Proteins genetics, Casein Kinase II genetics, Casein Kinase II metabolism, DNA-Binding Proteins genetics, Human papillomavirus 16 pathogenicity, Humans, Keratinocytes virology, Life Cycle Stages, Nuclear Proteins genetics, Oncogene Proteins, Viral genetics, Phosphorylation, Serine metabolism, Virus Replication, Carrier Proteins metabolism, Chromatin, DNA-Binding Proteins metabolism, Host-Pathogen Interactions genetics, Human papillomavirus 16 genetics, Mitosis, Nuclear Proteins metabolism, Oncogene Proteins, Viral metabolism, Serine genetics
- Abstract
During the human papillomavirus 16 (HPV16) life cycle, the E2 protein interacts with host factors to regulate viral transcription, replication, and genome segregation/retention. Our understanding of host partner proteins and their roles in E2 functions remains incomplete. Here we demonstrate that CK2 phosphorylation of E2 on serine 23 promotes interaction with TopBP1 in vitro and in vivo and that E2 is phosphorylated on this residue during the HPV16 life cycle. We investigated the consequences of mutating serine 23 on E2 functions. E2-S23A (E2 with serine 23 mutated to alanine) activates and represses transcription identically to E2-WT (wild-type E2), and E2-S23A is as efficient as E2-WT in transient replication assays. However, E2-S23A has compromised interaction with mitotic chromatin compared with E2-WT. In E2-WT cells, both E2 and TopBP1 levels increase during mitosis compared with vector control cells. In E2-S23A cells, neither E2 nor TopBP1 levels increase during mitosis. Introduction of the S23A mutation into the HPV16 genome resulted in delayed immortalization of human foreskin keratinocytes (HFK) and higher episomal viral genome copy number in resulting established HFK. Remarkably, S23A cells had a disrupted viral life cycle in organotypic raft cultures, with a loss of E2 expression and a failure of viral replication. Overall, our results demonstrate that CK2 phosphorylation of E2 on serine 23 promotes interaction with TopBP1 and that this interaction is critical for the viral life cycle. IMPORTANCE Human papillomaviruses are causative agents in around 5% of all cancers, with no specific antiviral therapeutics available for treating infections or resultant cancers. In this report, we demonstrate that phosphorylation of HPV16 E2 by CK2 promotes formation of a complex with the cellular protein TopBP1 in vitro and in vivo . This complex results in stabilization of E2 during mitosis. We demonstrate that CK2 phosphorylates E2 on serine 23 in vivo and that CK2 inhibitors disrupt the E2-TopBP1 complex. Mutation of E2 serine 23 to alanine disrupts the HPV16 life cycle, hindering immortalization and disrupting the viral life cycle, demonstrating a critical function for this residue.
- Published
- 2021
- Full Text
- View/download PDF
16. Inhibition of MRN activity by a telomere protein motif.
- Author
-
Khayat F, Cannavo E, Alshmery M, Foster WR, Chahwan C, Maddalena M, Smith C, Oliver AW, Watson AT, Carr AM, Cejka P, and Bianchi A
- Subjects
- Amino Acid Sequence, DNA Breaks, Double-Stranded, DNA End-Joining Repair, DNA Helicases genetics, DNA, Fungal genetics, DNA, Fungal metabolism, Endodeoxyribonucleases genetics, Exodeoxyribonucleases genetics, Genomic Instability, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Origin Recognition Complex genetics, Origin Recognition Complex metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Sequence Homology, Amino Acid, Telomere genetics, Telomere-Binding Proteins genetics, Telomere-Binding Proteins metabolism, Amino Acid Motifs, DNA Helicases metabolism, Endodeoxyribonucleases metabolism, Exodeoxyribonucleases metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism, Telomere metabolism
- Abstract
The MRN complex (MRX in Saccharomyces cerevisiae, made of Mre11, Rad50 and Nbs1/Xrs2) initiates double-stranded DNA break repair and activates the Tel1/ATM kinase in the DNA damage response. Telomeres counter both outcomes at chromosome ends, partly by keeping MRN-ATM in check. We show that MRX is disabled by telomeric protein Rif2 through an N-terminal motif (MIN, MRN/X-inhibitory motif). MIN executes suppression of Tel1, DNA end-resection and non-homologous end joining by binding the Rad50 N-terminal region. Our data suggest that MIN promotes a transition within MRX that is not conductive for endonuclease activity, DNA-end tethering or Tel1 kinase activation, highlighting an Achilles' heel in MRN, which we propose is also exploited by the RIF2 paralog ORC4 (Origin Recognition Complex 4) in Kluyveromyces lactis and the Schizosaccharomyces pombe telomeric factor Taz1, which is evolutionarily unrelated to Orc4/Rif2. This raises the possibility that analogous mechanisms might be deployed in other eukaryotes as well.
- Published
- 2021
- Full Text
- View/download PDF
17. Structural basis for recruitment of the CHK1 DNA damage kinase by the CLASPIN scaffold protein.
- Author
-
Day M, Parry-Morris S, Houghton-Gisby J, Oliver AW, and Pearl LH
- Subjects
- Animals, Binding Sites, Checkpoint Kinase 1 genetics, Crystallography, X-Ray, Humans, Models, Molecular, Mutation, Phosphorylation, Protein Binding, Protein Conformation, Protein Domains, Sf9 Cells, Adaptor Proteins, Signal Transducing chemistry, Adaptor Proteins, Signal Transducing metabolism, Checkpoint Kinase 1 chemistry, Checkpoint Kinase 1 metabolism
- Abstract
CHK1 is a protein kinase that functions downstream of activated ATR to phosphorylate multiple targets as part of intra-S and G2/M DNA damage checkpoints. Its role in allowing cells to survive replicative stress has made it an important target for anti-cancer drug discovery. Activation of CHK1 by ATR depends on their mutual interaction with CLASPIN, a natively unstructured protein that interacts with CHK1 through a cluster of phosphorylation sites in its C-terminal half. We have now determined the crystal structure of the kinase domain of CHK1 bound to a high-affinity motif from CLASPIN. Our data show that CLASPIN engages a conserved site on CHK1 adjacent to the substrate-binding cleft, involved in phosphate sensing in other kinases. The CLASPIN motif is not phosphorylated by CHK1, nor does it affect phosphorylation of a CDC25 substrate peptide, suggesting that it functions purely as a scaffold for CHK1 activation by ATR., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. Nse5/6 is a negative regulator of the ATPase activity of the Smc5/6 complex.
- Author
-
Hallett ST, Schellenberger P, Zhou L, Beuron F, Morris E, Murray JM, and Oliver AW
- Subjects
- Adenosine Triphosphatases chemistry, Adenosine Triphosphatases genetics, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Cell Cycle Proteins ultrastructure, Chromosomal Proteins, Non-Histone chemistry, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone ultrastructure, DNA metabolism, Escherichia coli metabolism, Microscopy, Electron, Transmission, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins ultrastructure, Adenosine Triphosphatases metabolism, Cell Cycle Proteins metabolism, Chromosomal Proteins, Non-Histone metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
The multi-component Smc5/6 complex plays a critical role in the resolution of recombination intermediates formed during mitosis and meiosis, and in the cellular response to replication stress. Using recombinant proteins, we have reconstituted a series of defined Saccharomyces cerevisiae Smc5/6 complexes, visualised them by negative stain electron microscopy, and tested their ability to function as an ATPase. We find that only the six protein 'holo-complex' is capable of turning over ATP and that its activity is significantly increased by the addition of double-stranded DNA to reaction mixes. Furthermore, stimulation is wholly dependent on functional ATP-binding pockets in both Smc5 and Smc6. Importantly, we demonstrate that budding yeast Nse5/6 acts as a negative regulator of Smc5/6 ATPase activity, binding to the head-end of the complex to suppress turnover, irrespective of the DNA-bound status of the complex., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
19. Live-cell single-molecule tracking highlights requirements for stable Smc5/6 chromatin association in vivo.
- Author
-
Etheridge TJ, Villahermosa D, Campillo-Funollet E, Herbert AD, Irmisch A, Watson AT, Dang HQ, Osborne MA, Oliver AW, Carr AM, and Murray JM
- Subjects
- Cell Cycle Proteins metabolism, Chromatin metabolism, Schizosaccharomyces metabolism, Schizosaccharomyces pombe Proteins metabolism, Single Molecule Imaging
- Abstract
The essential Smc5/6 complex is required in response to replication stress and is best known for ensuring the fidelity of homologous recombination. Using single-molecule tracking in live fission yeast to investigate Smc5/6 chromatin association, we show that Smc5/6 is chromatin associated in unchallenged cells and this depends on the non-SMC protein Nse6. We define a minimum of two Nse6-dependent sub-pathways, one of which requires the BRCT-domain protein Brc1. Using defined mutants in genes encoding the core Smc5/6 complex subunits, we show that the Nse3 double-stranded DNA binding activity and the arginine fingers of the two Smc5/6 ATPase binding sites are critical for chromatin association. Interestingly, disrupting the single-stranded DNA (ssDNA) binding activity at the hinge region does not prevent chromatin association but leads to elevated levels of gross chromosomal rearrangements during replication restart. This is consistent with a downstream function for ssDNA binding in regulating homologous recombination., Competing Interests: TE, DV, EC, AH, AI, AW, HD, MO, AO, AC, JM No competing interests declared, (© 2021, Etheridge et al.)
- Published
- 2021
- Full Text
- View/download PDF
20. Uncovering an allosteric mode of action for a selective inhibitor of human Bloom syndrome protein.
- Author
-
Chen X, Ali YI, Fisher CE, Arribas-Bosacoma R, Rajasekaran MB, Williams G, Walker S, Booth JR, Hudson JJ, Roe SM, Pearl LH, Ward SE, Pearl FM, and Oliver AW
- Subjects
- DNA metabolism, DNA, Cruciform, DNA, Single-Stranded, Drug Discovery, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Escherichia coli, High-Throughput Screening Assays, Humans, RecQ Helicases metabolism, RecQ Helicases antagonists & inhibitors, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology
- Abstract
BLM (Bloom syndrome protein) is a RECQ-family helicase involved in the dissolution of complex DNA structures and repair intermediates. Synthetic lethality analysis implicates BLM as a promising target in a range of cancers with defects in the DNA damage response; however, selective small molecule inhibitors of defined mechanism are currently lacking. Here, we identify and characterise a specific inhibitor of BLM's ATPase-coupled DNA helicase activity, by allosteric trapping of a DNA-bound translocation intermediate. Crystallographic structures of BLM-DNA-ADP-inhibitor complexes identify a hitherto unknown interdomain interface, whose opening and closing are integral to translocation of ssDNA, and which provides a highly selective pocket for drug discovery. Comparison with structures of other RECQ helicases provides a model for branch migration of Holliday junctions by BLM., Competing Interests: XC, YA, CF, RA, MR, GW, SW, JB, JH, SR, LP, SW, FP, AO No competing interests declared, (© 2021, Chen et al.)
- Published
- 2021
- Full Text
- View/download PDF
21. Potential Effects of Human Papillomavirus Type Substitution, Superinfection Exclusion and Latency on the Efficacy of the Current L1 Prophylactic Vaccines.
- Author
-
Hampson IN, Oliver AW, and Hampson L
- Subjects
- Female, Humans, Incidence, Neoplasms etiology, Papillomaviridae classification, Papillomavirus Infections complications, Papillomavirus Infections pathology, Papillomavirus Infections virology, Prevalence, Squamous Intraepithelial Lesions of the Cervix etiology, Squamous Intraepithelial Lesions of the Cervix pathology, Vaccination, Virus Replication, Capsid Proteins immunology, Oncogene Proteins, Viral immunology, Papillomaviridae physiology, Papillomavirus Infections prevention & control, Papillomavirus Vaccines immunology, Superinfection virology, Virus Latency immunology
- Abstract
There are >200 different types of human papilloma virus (HPV) of which >51 infect genital epithelium, with the ~14 of these classed as high-risk being more commonly associated with cervical cancer. During development of the disease, high-risk types have an increased tendency to develop a truncated non-replicative life cycle, whereas low-risk, non-cancer-associated HPV types are either asymptomatic or cause benign lesions completing their full replicative life cycle. HPVs can also be present as non-replicative so-called "latent" infections and they can also show superinfection exclusion, where cells with pre-existing infections with one type cannot be infected with a different HPV type. Thus, the HPV repertoire and replication status present in an individual can form a complex dynamic meta-community which changes with respect to both time and exposure to different HPV types. In light of these considerations, it is not clear how current prophylactic HPV vaccines will affect this system and the potential for iatrogenic outcomes is discussed in light of recent outcome data.
- Published
- 2020
- Full Text
- View/download PDF
22. A role of the Nse4 kleisin and Nse1/Nse3 KITE subunits in the ATPase cycle of SMC5/6.
- Author
-
Vondrova L, Kolesar P, Adamus M, Nociar M, Oliver AW, and Palecek JJ
- Subjects
- Carrier Proteins genetics, Cell Cycle Proteins genetics, Macromolecular Substances metabolism, Mutagenesis, Site-Directed, Nuclear Proteins genetics, Protein Binding genetics, Schizosaccharomyces genetics, Schizosaccharomyces metabolism, Schizosaccharomyces pombe Proteins genetics, Sequence Alignment, Two-Hybrid System Techniques, Adenosine Triphosphatases metabolism, Carrier Proteins metabolism, Cell Cycle Proteins metabolism, Nuclear Proteins metabolism, Schizosaccharomyces pombe Proteins metabolism
- Abstract
The SMC (Structural Maintenance of Chromosomes) complexes are composed of SMC dimers, kleisin and kleisin-interacting (HAWK or KITE) subunits. Mutual interactions of these subunits constitute the basal architecture of the SMC complexes. In addition, binding of ATP molecules to the SMC subunits and their hydrolysis drive dynamics of these complexes. Here, we developed new systems to follow the interactions between SMC5/6 subunits and the relative stability of the complex. First, we show that the N-terminal domain of the Nse4 kleisin molecule binds to the SMC6 neck and bridges it to the SMC5 head. Second, binding of the Nse1 and Nse3 KITE proteins to the Nse4 linker increased stability of the ATP-free SMC5/6 complex. In contrast, binding of ATP to SMC5/6 containing KITE subunits significantly decreased its stability. Elongation of the Nse4 linker partially suppressed instability of the ATP-bound complex, suggesting that the binding of the KITE proteins to the Nse4 linker constrains its limited size. Our data suggest that the KITE proteins may shape the Nse4 linker to fit the ATP-free complex optimally and to facilitate opening of the complex upon ATP binding. This mechanism suggests an important role of the KITE subunits in the dynamics of the SMC5/6 complexes.
- Published
- 2020
- Full Text
- View/download PDF
23. Phosphorylation-mediated interactions with TOPBP1 couple 53BP1 and 9-1-1 to control the G1 DNA damage checkpoint.
- Author
-
Bigot N, Day M, Baldock RA, Watts FZ, Oliver AW, and Pearl LH
- Subjects
- Ataxia Telangiectasia Mutated Proteins chemistry, Ataxia Telangiectasia Mutated Proteins genetics, Carrier Proteins genetics, Cell Cycle Checkpoints genetics, Checkpoint Kinase 1 chemistry, Checkpoint Kinase 1 genetics, DNA Replication genetics, DNA-Binding Proteins genetics, HeLa Cells, Humans, Methylation, Multiprotein Complexes genetics, Nuclear Proteins genetics, Phosphorylation, Protein Binding genetics, Protein Conformation, Protein Domains genetics, Protein Processing, Post-Translational genetics, S Phase genetics, Tumor Suppressor p53-Binding Protein 1 genetics, Ubiquitination genetics, Carrier Proteins chemistry, DNA Damage genetics, DNA-Binding Proteins chemistry, Multiprotein Complexes chemistry, Nuclear Proteins chemistry, Tumor Suppressor p53-Binding Protein 1 chemistry
- Abstract
Coordination of the cellular response to DNA damage is organised by multi-domain 'scaffold' proteins, including 53BP1 and TOPBP1, which recognise post-translational modifications such as phosphorylation, methylation and ubiquitylation on other proteins, and are themselves carriers of such regulatory signals. Here we show that the DNA damage checkpoint regulating S-phase entry is controlled by a phosphorylation-dependent interaction of 53BP1 and TOPBP1. BRCT domains of TOPBP1 selectively bind conserved phosphorylation sites in the N-terminus of 53BP1. Mutation of these sites does not affect formation of 53BP1 or ATM foci following DNA damage, but abolishes recruitment of TOPBP1, ATR and CHK1 to 53BP1 damage foci, abrogating cell cycle arrest and permitting progression into S-phase. TOPBP1 interaction with 53BP1 is structurally complimentary to its interaction with RAD9-RAD1-HUS1, allowing these damage recognition factors to bind simultaneously to the same TOPBP1 molecule and cooperate in ATR activation in the G1 DNA damage checkpoint., Competing Interests: NB, MD, RB, FW, AO, LP No competing interests declared, (© 2019, Bigot et al.)
- Published
- 2019
- Full Text
- View/download PDF
24. MDC1 Interacts with TOPBP1 to Maintain Chromosomal Stability during Mitosis.
- Author
-
Leimbacher PA, Jones SE, Shorrocks AK, de Marco Zompit M, Day M, Blaauwendraad J, Bundschuh D, Bonham S, Fischer R, Fink D, Kessler BM, Oliver AW, Pearl LH, Blackford AN, and Stucki M
- Subjects
- Adaptor Proteins, Signal Transducing, Cell Cycle Proteins, DNA Breaks, Double-Stranded, DNA Damage genetics, DNA Repair genetics, G1 Phase genetics, Genome, Human genetics, Genomic Instability genetics, Histones, Humans, Phosphorylation, Signal Transduction genetics, Carrier Proteins genetics, Chromosomal Instability genetics, DNA-Binding Proteins genetics, Mitosis genetics, Nuclear Proteins genetics, Trans-Activators genetics
- Abstract
In mitosis, cells inactivate DNA double-strand break (DSB) repair pathways to preserve genome stability. However, some early signaling events still occur, such as recruitment of the scaffold protein MDC1 to phosphorylated histone H2AX at DSBs. Yet, it remains unclear whether these events are important for maintaining genome stability during mitosis. Here, we identify a highly conserved protein-interaction surface in MDC1 that is phosphorylated by CK2 and recognized by the DNA-damage response mediator protein TOPBP1. Disruption of MDC1-TOPBP1 binding causes a specific loss of TOPBP1 recruitment to DSBs in mitotic but not interphase cells, accompanied by mitotic radiosensitivity, increased micronuclei, and chromosomal instability. Mechanistically, we find that TOPBP1 forms filamentous structures capable of bridging MDC1 foci in mitosis, indicating that MDC1-TOPBP1 complexes tether DSBs until repair is reactivated in the following G1 phase. Thus, we reveal an important, hitherto-unnoticed cooperation between MDC1 and TOPBP1 in maintaining genome stability during cell division., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
25. Efficient Single-Strand Break Repair Requires Binding to Both Poly(ADP-Ribose) and DNA by the Central BRCT Domain of XRCC1.
- Author
-
Polo LM, Xu Y, Hornyak P, Garces F, Zeng Z, Hailstone R, Matthews SJ, Caldecott KW, Oliver AW, and Pearl LH
- Subjects
- Humans, Poly Adenosine Diphosphate Ribose genetics, X-ray Repair Cross Complementing Protein 1 genetics, DNA Breaks, Single-Stranded, Poly Adenosine Diphosphate Ribose metabolism, X-ray Repair Cross Complementing Protein 1 metabolism
- Abstract
XRCC1 accelerates repair of DNA single-strand breaks by acting as a scaffold protein for the recruitment of Polβ, LigIIIα, and end-processing factors, such as PNKP and APTX. XRCC1 itself is recruited to DNA damage through interaction of its central BRCT domain with poly(ADP-ribose) chains generated by PARP1 or PARP2. XRCC1 is believed to interact directly with DNA at sites of damage, but the molecular basis for this interaction within XRCC1 remains unclear. We now show that the central BRCT domain simultaneously mediates interaction of XRCC1 with poly(ADP-ribose) and DNA, through separate and non-overlapping binding sites on opposite faces of the domain. Mutation of residues within the DNA binding site, which includes the site of a common disease-associated human polymorphism, affects DNA binding of this XRCC1 domain in vitro and impairs XRCC1 recruitment and retention at DNA damage and repair of single-strand breaks in vivo., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
26. Are SMC Complexes Loop Extruding Factors? Linking Theory With Fact.
- Author
-
Baxter J, Oliver AW, and Schalbetter SA
- Subjects
- Animals, Archaea metabolism, Bacteria metabolism, Bacterial Proteins metabolism, Cell Cycle Proteins metabolism, Chromatin ultrastructure, DNA metabolism, Eukaryota metabolism, Humans, Chromatin metabolism, Chromosomal Proteins, Non-Histone metabolism
- Abstract
The extreme length of chromosomal DNA requires organizing mechanisms to both promote functional genetic interactions and ensure faithful chromosome segregation when cells divide. Microscopy and genome-wide contact frequency analyses indicate that intra-chromosomal looping of DNA is a primary pathway of chromosomal organization during all stages of the cell cycle. DNA loop extrusion has emerged as a unifying model for how chromosome loops are formed in cis in different genomic contexts and cell cycle stages. The highly conserved family of SMC complexes have been found to be required for DNA cis-looping and have been suggested to be the enzymatic core of loop extruding machines. Here, the current body of evidence available for the in vivo and in vitro action of SMC complexes is discussed and compared to the predictions made by the loop extrusion model. How SMC complexes may differentially act on chromatin to generate DNA loops and how they could work to generate the dynamic and functionally appropriate organization of DNA in cells is explored., (© 2018 The Authors. BioEssays Published by Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
27. The ASCIZ-DYNLL1 axis promotes 53BP1-dependent non-homologous end joining and PARP inhibitor sensitivity.
- Author
-
Becker JR, Cuella-Martin R, Barazas M, Liu R, Oliveira C, Oliver AW, Bilham K, Holt AB, Blackford AN, Heierhorst J, Jonkers J, Rottenberg S, and Chapman JR
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms pathology, CRISPR-Cas Systems, Cell Line, Tumor, DNA Breaks, Double-Stranded, Female, Genomic Instability genetics, HEK293 Cells, Humans, MCF-7 Cells, Mice, Mice, Inbred C57BL, Mice, Knockout, BRCA1 Protein genetics, Cytoplasmic Dyneins metabolism, DNA End-Joining Repair genetics, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Transcription Factors metabolism, Tumor Suppressor p53-Binding Protein 1 genetics
- Abstract
53BP1 controls a specialized non-homologous end joining (NHEJ) pathway that is essential for adaptive immunity, yet oncogenic in BRCA1 mutant cancers. Intra-chromosomal DNA double-strand break (DSB) joining events during immunoglobulin class switch recombination (CSR) require 53BP1. However, in BRCA1 mutant cells, 53BP1 blocks homologous recombination (HR) and promotes toxic NHEJ, resulting in genomic instability. Here, we identify the protein dimerization hub-DYNLL1-as an organizer of multimeric 53BP1 complexes. DYNLL1 binding stimulates 53BP1 oligomerization, and promotes 53BP1's recruitment to, and interaction with, DSB-associated chromatin. Consequently, DYNLL1 regulates 53BP1-dependent NHEJ: CSR is compromised upon deletion of Dynll1 or its transcriptional regulator Asciz, or by mutation of DYNLL1 binding motifs in 53BP1; furthermore, Brca1 mutant cells and tumours are rendered resistant to poly-ADP ribose polymerase (PARP) inhibitor treatments upon deletion of Dynll1 or Asciz. Thus, our results reveal a mechanism that regulates 53BP1-dependent NHEJ and the therapeutic response of BRCA1-deficient cancers.
- Published
- 2018
- Full Text
- View/download PDF
28. BRCT domains of the DNA damage checkpoint proteins TOPBP1/Rad4 display distinct specificities for phosphopeptide ligands.
- Author
-
Day M, Rappas M, Ptasinska K, Boos D, Oliver AW, and Pearl LH
- Subjects
- Amino Acid Sequence, Carrier Proteins chemistry, Carrier Proteins genetics, Carrier Proteins metabolism, DNA Damage, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Humans, Ligands, Models, Molecular, Nuclear Proteins chemistry, Nuclear Proteins genetics, Nuclear Proteins metabolism, Phosphopeptides genetics, Phosphopeptides metabolism, Phosphorylation, Protein Binding, Schizosaccharomyces genetics, Schizosaccharomyces metabolism, Schizosaccharomyces pombe Proteins genetics, Schizosaccharomyces pombe Proteins metabolism, Transglutaminases genetics, Transglutaminases metabolism, DNA-Binding Proteins chemistry, Phosphopeptides chemistry, Protein Domains, Schizosaccharomyces pombe Proteins chemistry, Transglutaminases chemistry
- Abstract
TOPBP1 and its fission yeast homologueRad4, are critical players in a range of DNA replication, repair and damage signalling processes. They are composed of multiple BRCT domains, some of which bind phosphorylated motifs in other proteins. They thus act as multi-point adaptors bringing proteins together into functional combinations, dependent on post-translational modifications downstream of cell cycle and DNA damage signals. We have now structurally and/or biochemically characterised a sufficient number of high-affinity complexes for the conserved N-terminal region of TOPBP1 and Rad4 with diverse phospho-ligands, including human RAD9 and Treslin, and Schizosaccharomyces pombe Crb2 and Sld3, to define the determinants of BRCT domain specificity. We use this to identify and characterise previously unknown phosphorylation-dependent TOPBP1/Rad4-binding motifs in human RHNO1 and the fission yeast homologue of MDC1, Mdb1. These results provide important insights into how multiple BRCT domains within TOPBP1/Rad4 achieve selective and combinatorial binding of their multiple partner proteins., Competing Interests: MD, MR, KP, DB, AO, LP No competing interests declared, (© 2018, Day et al.)
- Published
- 2018
- Full Text
- View/download PDF
29. DIS3 isoforms vary in their endoribonuclease activity and are differentially expressed within haematological cancers.
- Author
-
Robinson SR, Viegas SC, Matos RG, Domingues S, Bedir M, Stewart HJS, Chevassut TJ, Oliver AW, Arraiano CM, and Newbury SF
- Subjects
- Exosome Multienzyme Ribonuclease Complex genetics, HEK293 Cells, HeLa Cells, Hematologic Neoplasms genetics, Hematologic Neoplasms pathology, Humans, Isoenzymes biosynthesis, Isoenzymes genetics, Neoplasm Proteins genetics, THP-1 Cells, Alternative Splicing, Exosome Multienzyme Ribonuclease Complex biosynthesis, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Hematologic Neoplasms enzymology, Neoplasm Proteins biosynthesis
- Abstract
DIS3 (defective in sister chromatid joining) is the catalytic subunit of the exosome, a protein complex involved in the 3'-5' degradation of RNAs. DIS3 is a highly conserved exoribonuclease, also known as Rrp44. Global sequencing studies have identified DIS3 as being mutated in a range of cancers, with a considerable incidence in multiple myeloma. In this work, we have identified two protein-coding isoforms of DIS3. Both isoforms are functionally relevant and result from alternative splicing. They differ from each other in the size of their N-terminal PIN (PilT N-terminal) domain, which has been shown to have endoribonuclease activity and tether DIS3 to the exosome. Isoform 1 encodes a full-length PIN domain, whereas the PIN domain of isoform 2 is shorter and is missing a segment with conserved amino acids. We have carried out biochemical activity assays on both isoforms of full-length DIS3 and the isolated PIN domains. We find that isoform 2, despite missing part of the PIN domain, has greater endonuclease activity compared with isoform 1. Examination of the available structural information allows us to provide a hypothesis to explain this altered behaviour. Our results also show that multiple myeloma patient cells and all cancer cell lines tested have higher levels of isoform 1 compared with isoform 2, whereas acute myeloid leukaemia and chronic myelomonocytic leukaemia patient cells and samples from healthy donors have similar levels of isoforms 1 and 2. Taken together, our data indicate that significant changes in the ratios of the two isoforms could be symptomatic of haematological cancers., (© 2018 The Author(s).)
- Published
- 2018
- Full Text
- View/download PDF
30. CCRK is a novel signalling hub exploitable in cancer immunotherapy.
- Author
-
Mok MT, Zhou J, Tang W, Zeng X, Oliver AW, Ward SE, and Cheng AS
- Subjects
- Animals, Apoptosis drug effects, Cyclin-Dependent Kinases genetics, Gene Expression Regulation, Neoplastic drug effects, Humans, Molecular Targeted Therapy, Neoplasms enzymology, Neoplasms genetics, Protein Kinase Inhibitors therapeutic use, Small Molecule Libraries therapeutic use, Tumor Microenvironment immunology, Cyclin-Dependent Kinase-Activating Kinase, Cyclin-Dependent Kinases antagonists & inhibitors, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy, Signal Transduction drug effects, Tumor Microenvironment drug effects
- Abstract
Cyclin-dependent kinase 20 (CDK20), or more commonly referred to as cell cycle-related kinase (CCRK), is the latest member of CDK family with strong linkage to human cancers. Accumulating studies have reported the consistent overexpression of CCRK in cancers arising from brain, colon, liver, lung and ovary. Such aberrant up-regulation of CCRK is clinically significant as it correlates with tumor staging, shorter patient survival and poor prognosis. Intriguingly, the signalling molecules perturbed by CCRK are divergent and cancer-specific, including the cell cycle regulators CDK2, cyclin D1, cyclin E and RB in glioblastoma, ovarian carcinoma and colorectal cancer, and KEAP1-NRF2 cytoprotective pathway in lung cancer. In hepatocellular carcinoma (HCC), CCRK mediates virus-host interaction to promote hepatitis B virus-associated tumorigenesis. Further mechanistic analyses reveal that CCRK orchestrates a self-reinforcing circuitry comprising of AR, GSK3β, β-catenin, AKT, EZH2, and NF-κB signalling for transcriptional and epigenetic regulation of oncogenes and tumor suppressor genes. Notably, EZH2 and NF-κB in this circuit have been recently shown to induce IL-6 production to facilitate tumor immune evasion. Concordantly, in a hepatoma preclinical model, ablation of Ccrk disrupts the immunosuppressive tumor microenvironment and enhances the therapeutic efficacy of immune checkpoint blockade via potentiation of anti-tumor T cell responses. In this review, we summarized the multifaceted tumor-intrinsic and -extrinsic functions of CCRK, which represents a novel signalling hub exploitable in cancer immunotherapy., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
31. Structural basis for recruitment of BRCA2 by PALB2.
- Author
-
Oliver AW, Swift S, Lord CJ, Ashworth A, and Pearl LH
- Published
- 2017
- Full Text
- View/download PDF
32. Specialized interfaces of Smc5/6 control hinge stability and DNA association.
- Author
-
Alt A, Dang HQ, Wells OS, Polo LM, Smith MA, McGregor GA, Welte T, Lehmann AR, Pearl LH, Murray JM, and Oliver AW
- Subjects
- Adenosine Triphosphatases chemistry, Binding Sites, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Survival physiology, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Crystallography, X-Ray, DNA Damage, DNA-Binding Proteins chemistry, Humans, Models, Molecular, Multiprotein Complexes chemistry, Mutagenesis, Site-Directed, Mutation, Phenotype, Protein Binding, Protein Domains physiology, Protein Multimerization physiology, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Schizosaccharomyces physiology, Schizosaccharomyces pombe Proteins genetics, Schizosaccharomyces pombe Proteins metabolism, Cohesins, Cell Cycle Proteins chemistry, Chromosomal Proteins, Non-Histone chemistry, DNA Repair physiology, DNA, Single-Stranded metabolism, Schizosaccharomyces pombe Proteins chemistry
- Abstract
The Structural Maintenance of Chromosomes (SMC) complexes: cohesin, condensin and Smc5/6 are involved in the organization of higher-order chromosome structure-which is essential for accurate chromosome duplication and segregation. Each complex is scaffolded by a specific SMC protein dimer (heterodimer in eukaryotes) held together via their hinge domains. Here we show that the Smc5/6-hinge, like those of cohesin and condensin, also forms a toroidal structure but with distinctive subunit interfaces absent from the other SMC complexes; an unusual 'molecular latch' and a functional 'hub'. Defined mutations in these interfaces cause severe phenotypic effects with sensitivity to DNA-damaging agents in fission yeast and reduced viability in human cells. We show that the Smc5/6-hinge complex binds preferentially to ssDNA and that this interaction is affected by both 'latch' and 'hub' mutations, suggesting a key role for these unique features in controlling DNA association by the Smc5/6 complex.
- Published
- 2017
- Full Text
- View/download PDF
33. Silver nanoparticles exhibit size-dependent differential toxicity and induce expression of syncytin-1 in FA-AML1 and MOLT-4 leukaemia cell lines.
- Author
-
Alqahtani S, Promtong P, Oliver AW, He XT, Walker TD, Povey A, Hampson L, and Hampson IN
- Subjects
- Cell Proliferation, Endogenous Retroviruses metabolism, Fanconi Anemia complications, Gene Expression Regulation, Leukemic, Gene Products, env genetics, Humans, Leukemia, Myeloid, Acute etiology, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute physiopathology, Leukemia, T-Cell metabolism, Leukemia, T-Cell physiopathology, Metal Nanoparticles chemistry, Pregnancy Proteins genetics, RNA, Messenger, Silver pharmacology, Gene Products, env drug effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, T-Cell drug therapy, Metal Nanoparticles toxicity, Pregnancy Proteins drug effects, Silver toxicity, Up-Regulation
- Abstract
Human endogenous retrovirus (HERV) sequences make up ~8% of the human genome and increased expression of some HERV proteins has been observed in various pathologies including leukaemia and multiple sclerosis. However, little is known about the function of these HERV proteins or environmental factors which regulate their expression. Silver nanoparticles (AgNPs) are used very extensively as antimicrobials and antivirals in numerous consumer products although their effect on the expression of HERV gene products is unknown. Cell proliferation and cell toxicity assays were carried out on human acute T lymphoblastic leukaemia (MOLT-4) and Fanconi anaemia associated acute myeloid leukaemia (FA-AML1) cells treated with two different sizes of AgNPs (7nm and 50nm diameter). Reverse-transcriptase polymerase chain reaction and western blotting were then used to the assess expression of HERV-W syncytin-1 mRNA and protein in these cells. FA-AML1 cells were more sensitive overall than MOLT-4 to treatment with the smaller 7nm sized AgNp's being the most toxic in these cells. MOLT-4 cell were more resistant and showed no evidence of differential toxicity to the different sized particles. Syncytin-1 mRNA and protein were induced by both 7 and 50nm AgNPs in both cell types yet with different kinetics. In summary, the observation that AgNPs induce expression of syncytin-1 in FA-AML1 and MOLT-4 cells at doses as little as 5 µg/ml is grounds for concern since this protein is up-regulated in both malignant and neurodegenerative diseases. Considering the widespread use of AgNPs in the environment it is clear that their ability to induce syncytin-1 should be investigated further in other cell types., (© The Author 2016. Published by Oxford University Press on behalf of the UK Environmental Mutagen Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
34. A first generation inhibitor of human Greatwall kinase, enabled by structural and functional characterisation of a minimal kinase domain construct.
- Author
-
Ocasio CA, Rajasekaran MB, Walker S, Le Grand D, Spencer J, Pearl FM, Ward SE, Savic V, Pearl LH, Hochegger H, and Oliver AW
- Subjects
- Crystallization, HeLa Cells, Humans, Microtubule-Associated Proteins chemistry, Phosphorylation, Protein Domains, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases chemistry, Structure-Activity Relationship, Microtubule-Associated Proteins antagonists & inhibitors, Protein Kinase Inhibitors chemical synthesis, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
MASTL (microtubule-associated serine/threonine kinase-like), more commonly known as Greatwall (GWL), has been proposed as a novel cancer therapy target. GWL plays a crucial role in mitotic progression, via its known substrates ENSA/ARPP19, which when phosphorylated inactivate PP2A/B55 phosphatase. When over-expressed in breast cancer, GWL induces oncogenic properties such as transformation and invasiveness. Conversely, down-regulation of GWL selectively sensitises tumour cells to chemotherapy. Here we describe the first structure of the GWL minimal kinase domain and development of a small-molecule inhibitor GKI-1 (Greatwall Kinase Inhibitor-1). In vitro, GKI-1 inhibits full-length human GWL, and shows cellular efficacy. Treatment of HeLa cells with GKI-1 reduces ENSA/ARPP19 phosphorylation levels, such that they are comparable to those obtained by siRNA depletion of GWL; resulting in a decrease in mitotic events, mitotic arrest/cell death and cytokinesis failure. Furthermore, GKI-1 will be a useful starting point for the development of more potent and selective GWL inhibitors.
- Published
- 2016
- Full Text
- View/download PDF
35. Analysis of the Prevalence of HTLV-1 Proviral DNA in Cervical Smears and Carcinomas from HIV Positive and Negative Kenyan Women.
- Author
-
He X, Maranga IO, Oliver AW, Gichangi P, Hampson L, and Hampson IN
- Subjects
- Adult, Cross-Sectional Studies, DNA, Viral genetics, Female, HIV Infections complications, Humans, Kenya epidemiology, Middle Aged, Proviruses genetics, Vaginal Smears, Young Adult, Carcinoma complications, DNA, Viral isolation & purification, HTLV-I Infections epidemiology, Proviruses isolation & purification, Uterine Cervical Neoplasms complications
- Abstract
The oncogenic retrovirus human T-cell lymphotropic virus type 1 (HTLV-1) is endemic in some countries although its prevalence and relationship with other sexually transmitted infections in Sub-Saharan Africa is largely unknown. A novel endpoint PCR method was used to analyse the prevalence of HTLV-1 proviral DNA in genomic DNA extracted from liquid based cytology (LBC) cervical smears and invasive cervical carcinomas (ICCs) obtained from human immunodeficiency virus-positive (HIV+ve) and HIV-negative (HIV-ve) Kenyan women. Patient sociodemographic details were recorded by structured questionnaire and these data analysed with respect to HIV status, human papillomavirus (HPV) type (Papilocheck(®)) and cytology. This showed 22/113 (19.5%) of LBC's from HIV+ve patients were positive for HTLV-1 compared to 4/111 (3.6%) of those from HIV-ve women (p = 0.0002; odds ratio (OR) = 6.42 (2.07-26.56)). Only 1/37 (2.7%) of HIV+ve and none of the 44 HIV-ve ICC samples were positive for HTLV-1. There was also a significant correlation between HTLV-1 infection, numbers of sexual partners (p < 0.05) and smoking (p < 0.01). Using this unique method, these data suggest an unexpectedly high prevalence of HTLV-1 DNA in HIV+ve women in this geographical location. However, the low level of HTLV-1 detected in HIV+ve ICC samples was unexpected and the reasons for this are unclear., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
36. PARP3 is a sensor of nicked nucleosomes and monoribosylates histone H2B(Glu2).
- Author
-
Grundy GJ, Polo LM, Zeng Z, Rulten SL, Hoch NC, Paomephan P, Xu Y, Sweet SM, Thorne AW, Oliver AW, Matthews SJ, Pearl LH, and Caldecott KW
- Subjects
- Animals, Cell Line, Chickens, Chromatin metabolism, Chromosomes metabolism, DNA metabolism, DNA Repair, Humans, Models, Molecular, Poly(ADP-ribose) Polymerases chemistry, Protein Domains, DNA Breaks, Single-Stranded, Histones metabolism, Nucleosomes metabolism, Poly(ADP-ribose) Polymerases metabolism, Ribose metabolism
- Abstract
PARP3 is a member of the ADP-ribosyl transferase superfamily that we show accelerates the repair of chromosomal DNA single-strand breaks in avian DT40 cells. Two-dimensional nuclear magnetic resonance experiments reveal that PARP3 employs a conserved DNA-binding interface to detect and stably bind DNA breaks and to accumulate at sites of chromosome damage. PARP3 preferentially binds to and is activated by mononucleosomes containing nicked DNA and which target PARP3 trans-ribosylation activity to a single-histone substrate. Although nicks in naked DNA stimulate PARP3 autoribosylation, nicks in mononucleosomes promote the trans-ribosylation of histone H2B specifically at Glu2. These data identify PARP3 as a molecular sensor of nicked nucleosomes and demonstrate, for the first time, the ribosylation of chromatin at a site-specific DNA single-strand break.
- Published
- 2016
- Full Text
- View/download PDF
37. Destabilized SMC5/6 complex leads to chromosome breakage syndrome with severe lung disease.
- Author
-
van der Crabben SN, Hennus MP, McGregor GA, Ritter DI, Nagamani SC, Wells OS, Harakalova M, Chinn IK, Alt A, Vondrova L, Hochstenbach R, van Montfrans JM, Terheggen-Lagro SW, van Lieshout S, van Roosmalen MJ, Renkens I, Duran K, Nijman IJ, Kloosterman WP, Hennekam E, Orange JS, van Hasselt PM, Wheeler DA, Palecek JJ, Lehmann AR, Oliver AW, Pearl LH, Plon SE, Murray JM, and van Haaften G
- Subjects
- Alleles, B-Lymphocytes cytology, Cell Proliferation, Child, Child, Preschool, Chromosomal Proteins, Non-Histone, Chromosome Segregation, Chromosomes ultrastructure, DNA Damage, DNA Repair, DNA Replication, Family Health, Female, Fibroblasts metabolism, Homozygote, Humans, Infant, Male, Meiosis, Mitosis, Mutation, Missense, Pedigree, Recombination, Genetic, Syndrome, T-Lymphocytes cytology, Abnormalities, Multiple genetics, Cell Cycle Proteins genetics, Chromosome Breakage, Intracellular Signaling Peptides and Proteins genetics, Lung Diseases genetics
- Abstract
The structural maintenance of chromosomes (SMC) family of proteins supports mitotic proliferation, meiosis, and DNA repair to control genomic stability. Impairments in chromosome maintenance are linked to rare chromosome breakage disorders. Here, we have identified a chromosome breakage syndrome associated with severe lung disease in early childhood. Four children from two unrelated kindreds died of severe pulmonary disease during infancy following viral pneumonia with evidence of combined T and B cell immunodeficiency. Whole exome sequencing revealed biallelic missense mutations in the NSMCE3 (also known as NDNL2) gene, which encodes a subunit of the SMC5/6 complex that is essential for DNA damage response and chromosome segregation. The NSMCE3 mutations disrupted interactions within the SMC5/6 complex, leading to destabilization of the complex. Patient cells showed chromosome rearrangements, micronuclei, sensitivity to replication stress and DNA damage, and defective homologous recombination. This work associates missense mutations in NSMCE3 with an autosomal recessive chromosome breakage syndrome that leads to defective T and B cell function and acute respiratory distress syndrome in early childhood.
- Published
- 2016
- Full Text
- View/download PDF
38. Mode of action of DNA-competitive small molecule inhibitors of tyrosyl DNA phosphodiesterase 2.
- Author
-
Hornyak P, Askwith T, Walker S, Komulainen E, Paradowski M, Pennicott LE, Bartlett EJ, Brissett NC, Raoof A, Watson M, Jordan AM, Ogilvie DJ, Ward SE, Atack JR, Pearl LH, Caldecott KW, and Oliver AW
- Subjects
- Animals, Binding Sites, Crystallography, X-Ray, Enzyme Activation drug effects, Humans, Mice, Phosphoric Diester Hydrolases chemistry, Protein Binding, Protein Denaturation, Protein Structure, Secondary, Protein Structure, Tertiary, Riboflavin analogs & derivatives, Riboflavin pharmacology, Temperature, Phosphoric Diester Hydrolases metabolism, Riboflavin chemistry
- Abstract
Tyrosyl-DNA phosphodiesterase 2 (TDP2) is a 5'-tyrosyl DNA phosphodiesterase important for the repair of DNA adducts generated by non-productive (abortive) activity of topoisomerase II (TOP2). TDP2 facilitates therapeutic resistance to topoisomerase poisons, which are widely used in the treatment of a range of cancer types. Consequently, TDP2 is an interesting target for the development of small molecule inhibitors that could restore sensitivity to topoisomerase-directed therapies. Previous studies identified a class of deazaflavin-based molecules that showed inhibitory activity against TDP2 at therapeutically useful concentrations, but their mode of action was uncertain. We have confirmed that the deazaflavin series inhibits TDP2 enzyme activity in a fluorescence-based assay, suitable for high-throughput screen (HTS)-screening. We have gone on to determine crystal structures of these compounds bound to a 'humanized' form of murine TDP2. The structures reveal their novel mode of action as competitive ligands for the binding site of an incoming DNA substrate, and point the way to generating novel and potent inhibitors of TDP2., (© 2016 The Author(s).)
- Published
- 2016
- Full Text
- View/download PDF
39. The Ku-binding motif is a conserved module for recruitment and stimulation of non-homologous end-joining proteins.
- Author
-
Grundy GJ, Rulten SL, Arribas-Bosacoma R, Davidson K, Kozik Z, Oliver AW, Pearl LH, and Caldecott KW
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, DNA Breaks, Double-Stranded, DNA Damage, Exodeoxyribonucleases chemistry, Exodeoxyribonucleases metabolism, Humans, Ku Autoantigen, Models, Biological, Molecular Sequence Data, Protein Binding, Protein Structure, Tertiary, RecQ Helicases chemistry, RecQ Helicases metabolism, Werner Syndrome Helicase, Antigens, Nuclear metabolism, Conserved Sequence, DNA End-Joining Repair, DNA-Binding Proteins metabolism
- Abstract
The Ku-binding motif (KBM) is a short peptide module first identified in APLF that we now show is also present in Werner syndrome protein (WRN) and in Modulator of retrovirus infection homologue (MRI). We also identify a related but functionally distinct motif in XLF, WRN, MRI and PAXX, which we denote the XLF-like motif. We show that WRN possesses two KBMs; one at the N terminus next to the exonuclease domain and one at the C terminus next to an XLF-like motif. We reveal that the WRN C-terminal KBM and XLF-like motif function cooperatively to bind Ku complexes and that the N-terminal KBM mediates Ku-dependent stimulation of WRN exonuclease activity. We also show that WRN accelerates DSB repair by a mechanism requiring both KBMs, demonstrating the importance of WRN interaction with Ku. These data define a conserved family of KBMs that function as molecular tethers to recruit and/or stimulate enzymes during NHEJ.
- Published
- 2016
- Full Text
- View/download PDF
40. A Hypomorphic PALB2 Allele Gives Rise to an Unusual Form of FA-N Associated with Lymphoid Tumour Development.
- Author
-
Byrd PJ, Stewart GS, Smith A, Eaton C, Taylor AJ, Guy C, Eringyte I, Fooks P, Last JI, Horsley R, Oliver AW, Janic D, Dokmanovic L, Stankovic T, and Taylor AM
- Subjects
- Alleles, BRCA2 Protein genetics, BRCA2 Protein metabolism, Chromosomes genetics, DNA Damage genetics, Fanconi Anemia pathology, Fanconi Anemia Complementation Group N Protein, Humans, Lymphocytes metabolism, Lymphocytes pathology, Lymphoma, Non-Hodgkin pathology, Mutation, Fanconi Anemia genetics, Lymphoma, Non-Hodgkin genetics, Nuclear Proteins genetics, Rad51 Recombinase genetics, Tumor Suppressor Proteins genetics
- Abstract
Patients with biallelic truncating mutations in PALB2 have a severe form of Fanconi anaemia (FA-N), with a predisposition for developing embryonal-type tumours in infancy. Here we describe two unusual patients from a single family, carrying biallelic PALB2 mutations, one truncating, c.1676_1677delAAinsG;(p.Gln559ArgfsTer2), and the second, c.2586+1G>A; p.Thr839_Lys862del resulting in an in frame skip of exon 6 (24 amino acids). Strikingly, the affected individuals did not exhibit the severe developmental defects typical of FA-N patients and initially presented with B cell non-Hodgkin lymphoma. The expressed p.Thr839_Lys862del mutant PALB2 protein retained the ability to interact with BRCA2, previously unreported in FA-N patients. There was also a large increased chromosomal radiosensitivity following irradiation in G2 and increased sensitivity to mitomycin C. Although patient cells were unable to form Rad51 foci following exposure to either DNA damaging agent, U2OS cells, in which the mutant PALB2 with in frame skip of exon 6 was induced, did show recruitment of Rad51 to foci following damage. We conclude that a very mild form of FA-N exists arising from a hypomorphic PALB2 allele.
- Published
- 2016
- Full Text
- View/download PDF
41. Chromatin association of the SMC5/6 complex is dependent on binding of its NSE3 subunit to DNA.
- Author
-
Zabrady K, Adamus M, Vondrova L, Liao C, Skoupilova H, Novakova M, Jurcisinova L, Alt A, Oliver AW, Lehmann AR, and Palecek JJ
- Subjects
- Amino Acid Sequence, Cell Cycle Proteins chemistry, DNA Replication, Humans, Molecular Sequence Data, Protein Binding, Recombination, Genetic, Schizosaccharomyces genetics, Schizosaccharomyces metabolism, Sequence Homology, Amino Acid, Cell Cycle Proteins metabolism, Chromatin metabolism, DNA metabolism, Nuclear Proteins metabolism, Schizosaccharomyces pombe Proteins metabolism
- Abstract
SMC5/6 is a highly conserved protein complex related to cohesin and condensin, which are the key components of higher-order chromatin structures. The SMC5/6 complex is essential for proliferation in yeast and is involved in replication fork stability and processing. However, the precise mechanism of action of SMC5/6 is not known. Here we present evidence that the NSE1/NSE3/NSE4 sub-complex of SMC5/6 binds to double-stranded DNA without any preference for DNA-replication/recombination intermediates. Mutations of key basic residues within the NSE1/NSE3/NSE4 DNA-binding surface reduce binding to DNA in vitro. Their introduction into the Schizosaccharomyces pombe genome results in cell death or hypersensitivity to DNA damaging agents. Chromatin immunoprecipitation analysis of the hypomorphic nse3 DNA-binding mutant shows a reduced association of fission yeast SMC5/6 with chromatin. Based on our results, we propose a model for loading of the SMC5/6 complex onto the chromatin., (© The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2016
- Full Text
- View/download PDF
42. A Single-Arm, Proof-Of-Concept Trial of Lopimune (Lopinavir/Ritonavir) as a Treatment for HPV-Related Pre-Invasive Cervical Disease.
- Author
-
Hampson L, Maranga IO, Masinde MS, Oliver AW, Batman G, He X, Desai M, Okemwa PM, Stringfellow H, Martin-Hirsch P, Mwaniki AM, Gichangi P, and Hampson IN
- Subjects
- Administration, Intravaginal, Adult, Cervix Uteri pathology, Cervix Uteri virology, Colposcopy, Drug Administration Schedule, Drug Combinations, Female, Genotype, Humans, Kenya, Molecular Typing, Papillomaviridae drug effects, Papillomaviridae genetics, Papillomaviridae growth & development, Papillomavirus Infections pathology, Papillomavirus Infections psychology, Papillomavirus Infections virology, Patient Acceptance of Health Care psychology, Self Administration, Severity of Illness Index, Squamous Intraepithelial Lesions of the Cervix pathology, Squamous Intraepithelial Lesions of the Cervix psychology, Squamous Intraepithelial Lesions of the Cervix virology, Treatment Outcome, Antiviral Agents therapeutic use, Cervix Uteri drug effects, Lopinavir therapeutic use, Papillomavirus Infections drug therapy, Ritonavir therapeutic use, Squamous Intraepithelial Lesions of the Cervix drug therapy
- Abstract
Background: Cervical cancer is the most common female malignancy in the developing nations and the third most common cancer in women globally. An effective, inexpensive and self-applied topical treatment would be an ideal solution for treatment of screen-detected, pre-invasive cervical disease in low resource settings., Methods: Between 01/03/2013 and 01/08/2013, women attending Kenyatta National Hospital's Family Planning and Gynaecology Outpatients clinics were tested for HIV, HPV (Cervista®) and liquid based cervical cytology (LBC-ThinPrep®). HIV negative women diagnosed as high-risk HPV positive with high grade squamous intraepithelial lesions (HSIL) were examined by colposcopy and given a 2 week course of 1 capsule of Lopimune (CIPLA) twice daily, to be self-applied as a vaginal pessary. Colposcopy, HPV testing and LBC were repeated at 4 and 12 weeks post-start of treatment with a final punch biopsy at 3 months for histology. Primary outcome measures were acceptability of treatment with efficacy as a secondary consideration., Results: A total of 23 women with HSIL were treated with Lopimune during which time no adverse reactions were reported. A maximum concentration of 10 ng/ml of lopinavir was detected in patient plasma 1 week after starting treatment. HPV was no longer detected in 12/23 (52.2%, 95%CI: 30.6-73.2%). Post-treatment cytology at 12 weeks on women with HSIL, showed 14/22 (63.6%, 95%CI: 40.6-82.8%) had no dysplasia and 4/22 (18.2%, 95%CI: 9.9-65.1%) were now low grade demonstrating a combined positive response in 81.8% of women of which 77.8% was confirmed by histology. These data are supported by colposcopic images, which show regression of cervical lesions., Conclusions: These results demonstrate the potential of Lopimune as a self-applied therapy for HPV infection and related cervical lesions. Since there were no serious adverse events or detectable post-treatment morbidity, this study indicates that further trials are clearly justified to define optimal regimes and the overall benefit of this therapy., Trial Registration: ISRCTN Registry 48776874.
- Published
- 2016
- Full Text
- View/download PDF
43. ATM Localization and Heterochromatin Repair Depend on Direct Interaction of the 53BP1-BRCT2 Domain with γH2AX.
- Author
-
Baldock RA, Day M, Wilkinson OJ, Cloney R, Jeggo PA, Oliver AW, Watts FZ, and Pearl LH
- Subjects
- Animals, Chromosomal Proteins, Non-Histone metabolism, Crystallography, X-Ray, DNA Breaks, Double-Stranded, DNA-Binding Proteins metabolism, Fluorescent Antibody Technique, Gene Knockdown Techniques, Humans, Mice, Protein Processing, Post-Translational, Protein Structure, Quaternary, RNA, Small Interfering, Transfection, Tumor Suppressor p53-Binding Protein 1, Ataxia Telangiectasia Mutated Proteins metabolism, DNA Repair physiology, Heterochromatin metabolism, Histones metabolism, Intracellular Signaling Peptides and Proteins metabolism
- Abstract
53BP1 plays multiple roles in mammalian DNA damage repair, mediating pathway choice and facilitating DNA double-strand break repair in heterochromatin. Although it possesses a C-terminal BRCT2 domain, commonly involved in phospho-peptide binding in other proteins, initial recruitment of 53BP1 to sites of DNA damage depends on interaction with histone post-translational modifications--H4K20me2 and H2AK13/K15ub--downstream of the early γH2AX phosphorylation mark of DNA damage. We now show that, contrary to current models, the 53BP1-BRCT2 domain binds γH2AX directly, providing a third post-translational mark regulating 53BP1 function. We find that the interaction of 53BP1 with γH2AX is required for sustaining the 53BP1-dependent focal concentration of activated ATM that facilitates repair of DNA double-strand breaks in heterochromatin in G1., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
44. The XRCC1 phosphate-binding pocket binds poly (ADP-ribose) and is required for XRCC1 function.
- Author
-
Breslin C, Hornyak P, Ridley A, Rulten SL, Hanzlikova H, Oliver AW, and Caldecott KW
- Subjects
- Animals, Binding Sites, CHO Cells, Cell Line, Tumor, Cell Survival, Cricetulus, DNA Damage, Humans, Poly(ADP-ribose) Polymerases metabolism, Protein Structure, Tertiary, X-ray Repair Cross Complementing Protein 1, DNA Repair, DNA-Binding Proteins chemistry, DNA-Binding Proteins metabolism, Poly Adenosine Diphosphate Ribose metabolism
- Abstract
Poly (ADP-ribose) is synthesized at DNA single-strand breaks and can promote the recruitment of the scaffold protein, XRCC1. However, the mechanism and importance of this process has been challenged. To address this issue, we have characterized the mechanism of poly (ADP-ribose) binding by XRCC1 and examined its importance for XRCC1 function. We show that the phosphate-binding pocket in the central BRCT1 domain of XRCC1 is required for selective binding to poly (ADP-ribose) at low levels of ADP-ribosylation, and promotes interaction with cellular PARP1. We also show that the phosphate-binding pocket is required for EGFP-XRCC1 accumulation at DNA damage induced by UVA laser, H2O2, and at sites of sub-nuclear PCNA foci, suggesting that poly (ADP-ribose) promotes XRCC1 recruitment both at single-strand breaks globally across the genome and at sites of DNA replication stress. Finally, we show that the phosphate-binding pocket is required following DNA damage for XRCC1-dependent acceleration of DNA single-strand break repair, DNA base excision repair, and cell survival. These data support the hypothesis that poly (ADP-ribose) synthesis promotes XRCC1 recruitment at DNA damage sites and is important for XRCC1 function., (© The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2015
- Full Text
- View/download PDF
45. Corrigendum: The zinc-finger domains of PARP1 cooperate to recognize DNA strand breaks.
- Author
-
Ali AA, Timinszky G, Arribas-Bosacoma R, Kozlowski M, Hassa PO, Hassler M, Ladurner AG, Pearl LH, and Oliver AW
- Published
- 2015
- Full Text
- View/download PDF
46. The 3' to 5' Exoribonuclease DIS3: From Structure and Mechanisms to Biological Functions and Role in Human Disease.
- Author
-
Robinson SR, Oliver AW, Chevassut TJ, and Newbury SF
- Subjects
- Animals, Conserved Sequence, Humans, Protein Transport, Disease genetics, Exosome Multienzyme Ribonuclease Complex chemistry, Exosome Multienzyme Ribonuclease Complex metabolism
- Abstract
DIS3 is a conserved exoribonuclease and catalytic subunit of the exosome, a protein complex involved in the 3' to 5' degradation and processing of both nuclear and cytoplasmic RNA species. Recently, aberrant expression of DIS3 has been found to be implicated in a range of different cancers. Perhaps most striking is the finding that DIS3 is recurrently mutated in 11% of multiple myeloma patients. Much work has been done to elucidate the structural and biochemical characteristics of DIS3, including the mechanistic details of its role as an effector of RNA decay pathways. Nevertheless, we do not understand how DIS3 mutations can lead to cancer. There are a number of studies that pertain to the function of DIS3 at the organismal level. Mutant phenotypes in S. pombe, S. cerevisiae and Drosophila suggest DIS3 homologues have a common role in cell-cycle progression and microtubule assembly. DIS3 has also recently been implicated in antibody diversification of mouse B-cells. This article aims to review current knowledge of the structure, mechanisms and functions of DIS3 as well as highlighting the genetic patterns observed within myeloma patients, in order to yield insight into the putative role of DIS3 mutations in oncogenesis.
- Published
- 2015
- Full Text
- View/download PDF
47. High-sensitivity human papilloma virus genotyping reveals near universal positivity in anal squamous cell carcinoma: different implications for vaccine prevention and prognosis.
- Author
-
Baricevic I, He X, Chakrabarty B, Oliver AW, Bailey C, Summers J, Hampson L, Hampson I, Gilbert DC, and Renehan AG
- Subjects
- Adult, Aged, Aged, 80 and over, Anus Neoplasms pathology, Carcinoma, Squamous Cell pathology, Female, Genotype, Humans, Male, Middle Aged, Papillomaviridae genetics, Papillomaviridae immunology, Papillomavirus Infections virology, Prognosis, Randomized Controlled Trials as Topic, Anus Neoplasms prevention & control, Anus Neoplasms virology, Carcinoma, Squamous Cell prevention & control, Carcinoma, Squamous Cell virology, Papillomaviridae isolation & purification, Papillomavirus Infections prevention & control, Papillomavirus Vaccines administration & dosage
- Abstract
Background: Characterisation of human papilloma virus (HPV) infection in anal squamous cell carcinoma (ASCC) may have dual importance: first, aetiological; second, prognostic, informing outcome after chemo-radiotherapy (CRT). We undertook HPV genotyping, and allelic characterisations, to evaluate the aetiological role of HPV while simultaneously evaluating the impact of HPV genotyping on relapse-free (RFS) and overall survival (OS)., Method: Dual-primer HPV genotyping (subtypes 6, 11, 16, 18, 31, 33, 45, 52, 58) and DNA sequencing of HPV 16 positive tumours were analysed in 151 consecutively referred ASCCs, previously characterised by immunohistochemistry for p16 expression. In 110 patients treated with CRT, factors influencing RFS and OS were evaluated using univariate and multivariate models., Results: HPV positivity was observed in 95%. HPV 16 accounted for 89%; of these, 64% harboured the T350G E6 variant. HPV 16 positivity was significantly correlated with improved 5-year RFS (62% versus 40%; p = 0.027) and OS (59% versus 38%; p = 0.019). p16 expression was also significantly correlated with improved 5-year RFS (positive versus negative: 65% versus 16%; p < 0.0001) and OS (63% versus 13%; p < 0.0001). In multivariable models that included HPV 16 status, p16 status, sex, and age, p16 expression remained an independent prognostic factor for RFS (p < 0.0001) and OS (p = 0.002)., Conclusion: In ASCC, near-universal HPV detection rates were demonstrated, higher than generally reported in the literature, and supporting the development of multivalent HPV vaccinations for prevention. By contrast, p16 negatively, but not HPV 16 genotype, is an independent adverse prognosticator after chemo-radiotherapy in patients with ASCC., (Copyright © 2015. Published by Elsevier Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
48. Corrigendum to "The conserved Fanconi anemia nuclease Fan1 and the SUMO E3 ligase Pli1 act in two novel Pso2-independent pathways of DNA interstrand crosslink repair in yeast" [DNA Repair 12 (December (12)) (2013) 1011-1023].
- Author
-
Fontebasso Y, Etheridge TJ, Oliver AW, Murray JM, and Carr AM
- Published
- 2014
- Full Text
- View/download PDF
49. TopBP1: A BRCT-scaffold protein functioning in multiple cellular pathways.
- Author
-
Wardlaw CP, Carr AM, and Oliver AW
- Subjects
- Animals, Carrier Proteins chemistry, Carrier Proteins genetics, Cell Cycle Checkpoints, DNA Replication, DNA-Binding Proteins chemistry, DNA-Binding Proteins genetics, Humans, Nuclear Proteins chemistry, Nuclear Proteins genetics, Protein Structure, Tertiary, Carrier Proteins metabolism, DNA Repair, DNA-Binding Proteins metabolism, Nuclear Proteins metabolism
- Abstract
Human TopBP1 contains nine BRCT domains and functions in DNA replication initiation, checkpoint signalling, DNA repair and influences transcriptional control. TopBP1 and its homologues have been the subject of numerous scientific publications since the last comprehensive review in 2005, emerging as a key scaffold protein that links crucial components within these distinct cellular processes. This review focuses on recently published work, with particular emphasis on structural insights into TopBP1 function and the binding partners identified for DNA replication initiation, DNA-dependent checkpoints, DNA repair and transcription. We further summarise what is known about TopBP1 and links to human disease., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
50. Using HIV drugs to target human papilloma virus.
- Author
-
Hampson L, Oliver AW, and Hampson IN
- Subjects
- Alphapapillomavirus drug effects, Drug Repositioning, HIV Protease Inhibitors administration & dosage, Humans, Off-Label Use, Papillomavirus Infections virology, HIV Protease Inhibitors therapeutic use, Papillomavirus Infections drug therapy
- Abstract
Over the past decade it has been demonstrated that HIV protease inhibitors have various off-target activities that has enabled them to be repositioned as treatments for a range of other pathologies. Human papilloma virus and related malignancies have been shown to be susceptible to these agents and current progress with this indication is summarized here together with a discussion of the rationale for the off-target effects of these compounds.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.