Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Susan M. Bailey is active.

Publication


Featured researches published by Susan M. Bailey.


The EMBO Journal | 2002

Functional interaction between DNA‐PKcs and telomerase in telomere length maintenance

Silvia Espejel; Sonia Franco; Antonella Sgura; Darren Gae; Susan M. Bailey; Guillermo E. Taccioli; Maria A. Blasco

DNA‐PKcs is the catalytic subunit of the DNA‐dependent protein kinase (DNA‐PK) complex that functions in the non‐homologous end‐joining of double‐strand breaks, and it has been shown previously to have a role in telomere capping. In particular, DNA‐PKcs deficiency leads to chromosome fusions involving telomeres produced by leading‐strand synthesis. Here, by generating mice doubly deficient in DNA‐PKcs and telomerase (Terc−/−/DNA‐PKcs−/−), we demonstrate that DNA‐PKcs also has a fundamental role in telomere length maintenance. In particular, Terc−/−/DNA‐PKcs−/− mice displayed an accelerated rate of telomere shortening when compared with Terc−/− controls, suggesting a functional interaction between both activities in maintaining telomere length. In addition, we also provide direct demonstration that DNA‐PKcs is essential for both end‐to‐end fusions and apoptosis triggered by critically short telomeres. Our data predict that, in telomerase‐deficient cells, i.e. human somatic cells, DNA‐PKcs abrogation may lead to a faster rate of telomere degradation and cell cycle arrest in the absence of increased apoptosis and/or fusion of telomere‐exhausted chromosomes. These results suggest a critical role of DNA‐PKcs in both cancer and aging.


Cytogenetic and Genome Research | 2004

Strand-specific fluorescence in situ hybridization: the CO-FISH family

Susan M. Bailey; Edwin H. Goodwin; Michael N. Cornforth

The ability to prepare single-stranded chromosomal target DNA allows innovative uses of FISH technology for studies of chromosome organization. Standard FISH methodologies require functionally single-stranded DNAs in order to facilitate hybridization between the probe and the complementary chromosomal target sequence. This usually involves denaturation of double-stranded probes to induce temporary separation of the DNA strands. Strand-specific FISH (CO-FISH; Chromosome Orientation-FISH) involves selective removal of newly replicated strands from DNA of metaphase chromosomes which results in single-stranded target DNA. When single-stranded probes are then hybridized to such targets, the resulting strand-specific hybridization is capable of revealing a level of information previously unattainable at the cytogenetic level. Mammalian telomeric DNA consists of tandem repeats of the (TTAGGG) sequence, oriented 5′→3′ towards the termini of all vertebrate chromosomes. Based on this conserved structural organization, CO-FISH with a telomere probe reveals the absolute 5′→3′ orientation of DNA sequences with respect to the pter→qter direction of chromosomes. Development and various applications of CO-FISH will be discussed: detection of cryptic inversions, discrimination between telomeres produced by leading- versus lagging-strand synthesis, and replication timing of mammalian telomeres.


Molecular and Cellular Biology | 2003

Role of Mammalian Rad54 in Telomere Length Maintenance

Isabel Jaco; Purificación Muñoz; Fermín A. Goytisolo; Joanna Wesoly; Susan M. Bailey; Guillermo E. Taccioli; Maria A. Blasco

ABSTRACT The homologous recombination (HR) DNA repair pathway participates in telomere length maintenance in yeast but its putative role at mammalian telomeres is unknown. Mammalian Rad54 is part of the HR machinery, and Rad54-deficient mice show a reduced HR capability. Here, we show that Rad54-deficient mice also show significantly shorter telomeres than wild-type controls, indicating that Rad54 activity plays an essential role in telomere length maintenance in mammals. Rad54 deficiency also resulted in an increased frequency of end-to-end chromosome fusions involving telomeres compared to the controls, suggesting a putative role of Rad54 in telomere capping. Finally, the study of mice doubly deficient for Rad54 and DNA-PKcs showed that telomere fusions due to DNA-PKcs deficiency were not rescued in the absence of Rad54, suggesting that they are not mediated by Rad54 activity.


The EMBO Journal | 2010

SNMIB/Apollo protects leading‐strand telomeres against NHEJ‐mediated repair

Yung C. Lam; Shamima Akhter; Peili Gu; Jing Ye; Anaïs Poulet; Marie Josèphe Giraud-Panis; Susan M. Bailey; Eric Gilson; Randy J. Legerski; Sandy Chang

Progressive telomere attrition or deficiency of the protective shelterin complex elicits a DNA damage response as a result of a cells inability to distinguish dysfunctional telomeric ends from DNA double‐strand breaks. SNMIB/Apollo is a shelterin‐associated protein and a member of the SMN1/PSO2 nuclease family that localizes to telomeres through its interaction with TRF2. Here, we generated SNMIB/Apollo knockout mouse embryo fibroblasts (MEFs) to probe the function of SNMIB/Apollo at mammalian telomeres. SNMIB/Apollo null MEFs exhibit an increased incidence of G2 chromatid‐type fusions involving telomeres created by leading‐strand DNA synthesis, reflective of a failure to protect these telomeres after DNA replication. Mutations within SNMIB/Apollos conserved nuclease domain failed to suppress this phenotype, suggesting that its nuclease activity is required to protect leading‐strand telomeres. SNMIB/Apollo−/−ATM−/− MEFs display robust telomere fusions when Trf2 is depleted, indicating that ATM is dispensable for repair of uncapped telomeres in this setting. Our data implicate the 5′–3′ exonuclease function of SNM1B/Apollo in the generation of 3′ single‐stranded overhangs at newly replicated leading‐strand telomeres to protect them from engaging the non‐homologous end‐joining pathway.


Cancer Research | 2009

Telomere Dysfunction and DNA-PKcs Deficiency: Characterization and Consequence

Eli S. Williams; Rebekah Klingler; Brian Ponnaiya; Tanja Hardt; Evelin Schröck; Susan P. Lees-Miller; Katheryn Meek; Robert L. Ullrich; Susan M. Bailey

The mechanisms by which cells accurately distinguish between DNA double-strand break (DSB) ends and telomeric DNA ends remain poorly defined. Recent investigations have revealed intriguing interactions between DNA repair and telomeres. We were the first to report a requirement for the nonhomologous end-joining (NHEJ) protein DNA-dependent protein kinase (DNA-PK) in the effective end-capping of mammalian telomeres. Here, we report our continued characterization of uncapped (as opposed to shortened) dysfunctional telomeres in cells deficient for the catalytic subunit of DNA-PK (DNA-PKcs) and shed light on their consequence. We present evidence in support of our model that uncapped telomeres in this repair-deficient background are inappropriately detected and processed as DSBs and thus participate not only in spontaneous telomere-telomere fusion but, importantly, also in ionizing radiation-induced telomere-DSB fusion events. We show that phosphorylation of DNA-PKcs itself (Thr-2609 cluster) is a critical event for proper telomere end-processing and that ligase IV (NHEJ) is required for uncapped telomere fusion. We also find uncapped telomeres in cells from the BALB/c mouse, which harbors two single-nucleotide polymorphisms that result in reduced DNA-PKcs abundance and activity, most markedly in mammary tissue, and are both radiosensitive and susceptible to radiogenic mammary cancer. Our results suggest mechanistic links between uncapped/dysfunctional telomeres in DNA-PKcs-deficient backgrounds, radiation-induced instability, and breast cancer. These studies provide the first direct evidence of genetic susceptibility and environmental insult interactions leading to a unique and ongoing form of genomic instability capable of driving carcinogenesis.


Radiation Research | 2008

Resveratrol Reduces Radiation-Induced Chromosome Aberration Frequencies in Mouse Bone Marrow Cells

Ronald E. Carsten; Annette M. Bachand; Susan M. Bailey; Robert L. Ullrich

Abstract Carsten, R. E., Bachand, A. M., Bailey, S. M. and Ullrich, R. L. Resveratrol Reduces Radiation-Induced Chromosome Aberration Frequency in Mouse Bone Marrow Cells. Radiat. Res. 169, 633–638 (2008). Resveratrol, a polyphenol compound with reported antioxidant and anticarcinogenic effects, a wide range of molecular targets, and toxicity only at extreme doses, has received considerable attention. We evaluated the radioprotective effect of orally administered resveratrol on the frequencies of chromosome aberrations in irradiated mouse bone marrow cells. CBA/CaJ mice were divided into four groups: (1) no treatment, (2) resveratrol only, (3) radiation only, and (4) resveratrol and radiation. Resveratrol treatment (100 mg/kg daily) was initiated 2 days prior to irradiation. Bone marrow was then harvested at 1 and 30 days after a single dose of 3 Gy whole-body γ radiation. A statistically significant (P < 0.05) reduction in the mean total chromosome aberration frequency per metaphase at both times postirradiation in the resveratrol and radiation group compared to the radiation-only group was observed. This study is the first to demonstrate that resveratrol has radioprotective effects in vivo. These results support the use of resveratrol as a radioprotector with the potential for widespread application.


Cancer Research | 2005

NBS1 knockdown by small interfering RNA increases ionizing radiation mutagenesis and telomere association in human cells

Ying Zhang; Chang U.K. Lim; Eli S. Williams; Junqing Zhou; Qinming Zhang; Michael H. Fox; Susan M. Bailey; Howard L. Liber

Hypomorphic mutations which lead to decreased function of the NBS1 gene are responsible for Nijmegen breakage syndrome, a rare autosomal recessive hereditary disorder that imparts an increased predisposition to development of malignancy. The NBS1 protein is a component of the MRE11/RAD50/NBS1 complex that plays a critical role in cellular responses to DNA damage and the maintenance of chromosomal integrity. Using small interfering RNA transfection, we have knocked down NBS1 protein levels and analyzed relevant phenotypes in two closely related human lymphoblastoid cell lines with different p53 status, namely wild-type TK6 and mutated WTK1. Both TK6 and WTK1 cells showed an increased level of ionizing radiation-induced mutation at the TK and HPRT loci, impaired phosphorylation of H2AX (gamma-H2AX), and impaired activation of the cell cycle checkpoint regulating kinase, Chk2. In TK6 cells, ionizing radiation-induced accumulation of p53/p21 and apoptosis were reduced. There was a differential response to ionizing radiation-induced cell killing between TK6 and WTK1 cells after NBS1 knockdown; TK6 cells were more resistant to killing, whereas WTK1 cells were more sensitive. NBS1 deficiency also resulted in a significant increase in telomere association that was independent of radiation exposure and p53 status. Our results provide the first experimental evidence that NBS1 deficiency in human cells leads to hypermutability and telomere associations, phenotypes that may contribute to the cancer predisposition seen among patients with this disease.


Cancer Research | 2012

Deficiency in Mammalian Histone H2B Ubiquitin Ligase Bre1 (Rnf20/Rnf40) Leads to Replication Stress and Chromosomal Instability

Sophia B. Chernikova; Olga V. Razorenova; John P. Higgins; Brock J. Sishc; Monica Nicolau; Jennifer A. Dorth; Diana A. Chernikova; Shirley Kwok; James D. Brooks; Susan M. Bailey; John C. Game; J. Martin Brown

Mammalian Bre1 complexes (BRE1A/B (RNF20/40) in humans and Bre1a/b (Rnf20/40) in mice) function similarly to their yeast homolog Bre1 as ubiquitin ligases in monoubiquitination of histone H2B. This ubiquitination facilitates methylation of histone H3 at K4 and K79, and accounts for the roles of Bre1 and its homologs in transcriptional regulation. Recent studies by others suggested that Bre1 acts as a tumor suppressor, augmenting expression of select tumor suppressor genes and suppressing select oncogenes. In this study, we present an additional mechanism of tumor suppression by Bre1 through maintenance of genomic stability. We track the evolution of genomic instability in Bre1-deficient cells from replication-associated double-strand breaks (DSB) to specific genomic rearrangements that explain a rapid increase in DNA content and trigger breakage-fusion-bridge cycles. We show that aberrant RNA-DNA structures (R-loops) constitute a significant source of DSBs in Bre1-deficient cells. Combined with a previously reported defect in homologous recombination, generation of R-loops is a likely initiator of replication stress and genomic instability in Bre1-deficient cells. We propose that genomic instability triggered by Bre1 deficiency may be an important early step that precedes acquisition of an invasive phenotype, as we find decreased levels of BRE1A/B and dimethylated H3K79 in testicular seminoma and in the premalignant lesion in situ carcinoma.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Hyper telomere recombination accelerates replicative senescence and may promote premature aging

R. Tanner Hagelstrom; Krastan B. Blagoev; Laura J. Niedernhofer; Edwin H. Goodwin; Susan M. Bailey

Werner syndrome and Bloom syndrome result from defects in the RecQ helicases Werner (WRN) and Bloom (BLM), respectively, and display premature aging phenotypes. Similarly, XFE progeroid syndrome results from defects in the ERCC1-XPF DNA repair endonuclease. To gain insight into the origin of cellular senescence and human aging, we analyzed the dependence of sister chromatid exchange (SCE) frequencies on location [i.e., genomic (G-SCE) vs. telomeric (T-SCE) DNA] in primary human fibroblasts deficient in WRN, BLM, or ERCC1-XPF. Consistent with our other studies, we found evidence of elevated T-SCE in telomerase-negative but not telomerase-positive backgrounds. In telomerase-negative WRN-deficient cells, T-SCE—but not G-SCE—frequencies were significantly increased compared with controls. In contrast, SCE frequencies were significantly elevated in BLM-deficient cells irrespective of genome location. In ERCC1-XPF-deficient cells, neither T- nor G-SCE frequencies differed from controls. A theoretical model was developed that allowed an in silico investigation into the cellular consequences of increased T-SCE frequency. The model predicts that in cells with increased T-SCE, the onset of replicative senescence is dramatically accelerated even though the average rate of telomere loss has not changed. Premature cellular senescence may act as a powerful tumor-suppressor mechanism in telomerase-deficient cells with mutations that cause T-SCE levels to rise. Furthermore, T-SCE-driven premature cellular senescence may be a factor contributing to accelerated aging in Werner and Bloom syndromes, but not XFE progeroid syndrome.


Nature Genetics | 2007

DNA double-strand breaks are not sufficient to initiate recruitment of TRF2.

Eli S. Williams; Jan Stap; Jeroen Essers; Brian Ponnaiya; Martijn S. Luijsterburg; Przemek M. Krawczyk; Robert L. Ullrich; Jacob A. Aten; Susan M. Bailey

Kelvin Y K Chan1, Vera S F Chan2, Yongxiong Chen2, Shea-Ping Yip3, Chen-Lung S Lin2& Ui-Soon Khoo1 1Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Special Administrative Region, China. 2Division of Surgery, Oncology, Reproduction Biology and Anaesthetics, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK. 3Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Special Administrative Region, China. e-mail: [email protected] or [email protected]

Collaboration


Dive into the Susan M. Bailey's collaboration.

Top Co-Authors

Avatar

Edwin H. Goodwin

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Michael N. Cornforth

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Howard L. Liber

Colorado State University

View shared research outputs
Top Co-Authors

Avatar

Joel S. Bedford

Colorado State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

F. Andrew Ray

Colorado State University

View shared research outputs
Top Co-Authors

Avatar

Bradford D. Loucas

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge