35 research outputs found

    Defining the mechanistic role of hSSB1 : a novel single-stranded DNA binding protein essential for DNA repair

    Get PDF
    DNA is under constant attack from the external and internal environment. It is imperative to repair and maintain the vital genetic information of DNA. Otherwise it leads to an accumulation of mutations that alters the normal function of DNA and in turn causes a disorder in cellular metabolism. During repair, DNA unwinds into single-stranded DNA (ssDNA) and is even more vulnerable to damage. This is where human single-strand DNA binding protein 1 (hSSB1) binds and protects ssDNA. It is known that hSSB1 exists both as part of the MRN (MRE11, RAD50, NBS1) repair complex and the SOSS1 complex (made up of INTS3 and C9orf80). It is also known to initiate an appropriate repair mechanism by recruiting other proteins to the site of damage. There is an accumulating body of research on hSSB1 function in Double Strand Break Repair (DSBR). It promotes the ataxia telangiectasia mutated (ATM) kinase signalling cascade and also recruits the MRN complex to the site of double strand breaks (DSBs) in order to initiate DSBR via Homologous Recombination (HR). Recently, it was discovered that hSSB1 is capable of forming higher order oligomers and can function in the oxidative DNA damage response. The most common oxidative DNA damage is the 7,8-dihydro-8-oxoguanine (8-oxoG) adduct, which is the direct result of reactive oxygen species (ROS) produced during regular cellular respiration. If this damage goes unrepaired it may result in G:C to T:A transversions. These lesions are normally processed by the Base Excision Repair (BER) pathway, which involves human oxoguanine glycosylase (hOGG1) that cleaves the DNA backbone and removes the offending base. So far, it is understood that hSSB1 levels increase in response to oxidative damage; also, cells depleted of hSSB1 are hypersensitive to oxidative damage and are also unable to efficiently remove 8-oxoG adducts. The recruitment of hOGG1 to chromatin is dependent on hSSB1 and hSSB1 can promote hOGG1 cleavage of 8-oxoG. This thesis examines the mechanism of hSSB1 oligomerisation under oxidative conditions. hSSB1 forms dimers and tetramers and this oligomerisation is likely mediated by inter-domain disulfide bond formation. These oligomers can also be synthetically created through the use of a thiol reactive cross-linker. Oxidised hSSB1 binds to 8-oxoG damaged ssDNA with higher affinity than non-damaged ssDNA, likely indicating a direct role for oxidised hSSB1 in the recognition of 8-oxo-G lesions. Furthermore, hSSB1 and hOGG1 directly interact with a moderate binding affinity in the presence of 8-oxoG damaged ssDNA. Finally, a model of the tetramer is proposed using the recent crystal structure of monomeric hSSB1 as a template. The data presented here along with the proposed structural model allows hSSB1 to be placed in the oxidative DNA damage response pathway and gives crucial insight into the possible role of the oligomer in this process. As heightened levels of oxidative stress are associated with cancer (as well as aging and Alzheimer’s disease), understanding the molecular mechanisms of how cells repair oxidative DNA damage will be essential in the development of novel therapeutic treatments

    hSSB1 (NABP2/OBFC2B) is regulated by oxidative stress

    Get PDF
    The maintenance of genome stability is an essential cellular process to prevent the development of diseases including cancer. hSSB1 (NABP2/ OBFC2A) is a critical component of the DNA damage response where it participates in the repair of double-strand DNA breaks and in base excision repair of oxidized guanine residues (8-oxoguanine) by aiding the localization of the human 8-oxoguanine glycosylase (hOGG1) to damaged DNA. Here we demonstrate that following oxidative stress, hSSB1 is stabilized as an oligomer which is required for hSSB1 to function in the removal of 8-oxoguanine. Monomeric hSSB1 shows a decreased affinity for oxidized DNA resulting in a cellular 8-oxoguanine-repair defect and in the absence of ATM signaling initiation. While hSSB1 oligomerization is important for the removal of 8-oxoguanine from the genome, it is not required for the repair of double-strand DNA-breaks by homologous recombination. These findings demonstrate a novel hSSB1 regulatory mechanism for the repair of damaged DNA.Publisher PDFPeer reviewe

    hSSB1 phosphorylation is dynamically regulated by DNA-PK and PPP-family protein phosphatases

    Get PDF
    This work was supported by a National Health and Medical Research Council project grant [1066550], an Australian Research Council project grant [DP 120103099] and by a Queensland Health Senior Clinical Research Fellowship awarded to K.J.O. This work was also supported by the Wellcome Trust [094476/Z/10/Z], which funded the purchase of the TripleTOF 5600 mass spectrometer at the BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews. NWA was supported by a scholarship awarded by Cancer Council Queensland. E.B. is supported by an Advance Queensland Research Fellowship.The maintenance of genomic stability is essential for cellular viability and the prevention of diseases such as cancer. Human single-stranded DNA-binding protein 1 (hSSB1) is a protein with roles in the stabilisation and restart of stalled DNA replication forks, as well as in the repair of oxidative DNA lesions and double-strand DNA breaks. In the latter process, phosphorylation of threonine 117 by the ATM kinase is required for hSSB1 stability and efficient DNA repair. The regulation of hSSB1 in other DNA repair pathways has however remained unclear. Here we report that hSSB1 is also directly phosphorylated by DNA-PK at serine residue 134. While this modification is largely suppressed in undamaged cells by PPP-family protein phosphatases, S134 phosphorylation is enhanced following the disruption of replication forks and promotes cellular survival. Together, these data thereby represent a novel mechanism for hSSB1 regulation following the inhibition of replication.Publisher PDFPeer reviewe

    A novel role for the immunophilin FKBP52 in motor coordination

    Get PDF
    FKBP52 is a ubiquitously distributed immunophilin that has been associated with wideranging functions in cell signalling as well as hormonal and stress responses. Amongst other pathways, it acts via complex-formation with corticosteroid receptors and has consequently been associated with stress- and age-related neurodegenerative disorders including Alzheimer’s and Parkinson’s diseases. Reduced levels of FKBP52 have been linked to tau dysfunction and amyloid beta toxicity in AD. However, FKBP52’s role in cognition and neurodegenerative disorder-like phenotypes remained to be elucidated. The present study aimed therefore at investigating the cognitive and behavioural effects of reduced FKBP52 levels of genetically modified mice during ageing. Female and male FKBP52+/+, FKBP52+/- and FKBP52-/- mice were compared at two-, ten-, twelve-, fifteenand eighteen-months-of-age in a series of behavioural tests covering specie-specific behaviour, motor activity and coordination, fear-, spatial and recognition memory as well as curiosity and emotionality. Whilst cognitively unimpaired, FKBP52+/- mice performed worse on an accelerating rotating rod than FKBP52+/+ littermates across all age-groups suggesting that FKBP52 is involved in processes controlling motor coordination. This deficit did not exacerbate with age but did worsen with repeated testing; pointing towards a role for FKBP52 in learning of tasks requiring motor coordination abilities. This study contributes to the knowledge base of FKBP52’s implication in neurodegenerative diseases by demonstrating that FKBP52 by itself does not directly affect cognition and may therefore rather play an indirect, modulatory role in the functional pathology of AD, whereas it directly affects motor coordination, an early sign of neurodegenerative damages to the brain

    Effect of angiotensin-converting enzyme inhibitor and angiotensin receptor blocker initiation on organ support-free days in patients hospitalized with COVID-19

    Get PDF
    IMPORTANCE Overactivation of the renin-angiotensin system (RAS) may contribute to poor clinical outcomes in patients with COVID-19. Objective To determine whether angiotensin-converting enzyme (ACE) inhibitor or angiotensin receptor blocker (ARB) initiation improves outcomes in patients hospitalized for COVID-19. DESIGN, SETTING, AND PARTICIPANTS In an ongoing, adaptive platform randomized clinical trial, 721 critically ill and 58 non–critically ill hospitalized adults were randomized to receive an RAS inhibitor or control between March 16, 2021, and February 25, 2022, at 69 sites in 7 countries (final follow-up on June 1, 2022). INTERVENTIONS Patients were randomized to receive open-label initiation of an ACE inhibitor (n = 257), ARB (n = 248), ARB in combination with DMX-200 (a chemokine receptor-2 inhibitor; n = 10), or no RAS inhibitor (control; n = 264) for up to 10 days. MAIN OUTCOMES AND MEASURES The primary outcome was organ support–free days, a composite of hospital survival and days alive without cardiovascular or respiratory organ support through 21 days. The primary analysis was a bayesian cumulative logistic model. Odds ratios (ORs) greater than 1 represent improved outcomes. RESULTS On February 25, 2022, enrollment was discontinued due to safety concerns. Among 679 critically ill patients with available primary outcome data, the median age was 56 years and 239 participants (35.2%) were women. Median (IQR) organ support–free days among critically ill patients was 10 (–1 to 16) in the ACE inhibitor group (n = 231), 8 (–1 to 17) in the ARB group (n = 217), and 12 (0 to 17) in the control group (n = 231) (median adjusted odds ratios of 0.77 [95% bayesian credible interval, 0.58-1.06] for improvement for ACE inhibitor and 0.76 [95% credible interval, 0.56-1.05] for ARB compared with control). The posterior probabilities that ACE inhibitors and ARBs worsened organ support–free days compared with control were 94.9% and 95.4%, respectively. Hospital survival occurred in 166 of 231 critically ill participants (71.9%) in the ACE inhibitor group, 152 of 217 (70.0%) in the ARB group, and 182 of 231 (78.8%) in the control group (posterior probabilities that ACE inhibitor and ARB worsened hospital survival compared with control were 95.3% and 98.1%, respectively). CONCLUSIONS AND RELEVANCE In this trial, among critically ill adults with COVID-19, initiation of an ACE inhibitor or ARB did not improve, and likely worsened, clinical outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT0273570

    Backbone 1H, 13C and 15N resonance assignments of the OB domain of the single stranded DNA-binding protein hSSB1 (NABP2/OBFC2B) and chemical shift mapping of the DNA-binding interface

    No full text
    Single-stranded DNA-binding proteins (SSBs) are highly important in DNA metabolism and play an essential role in all major DNA repair pathways. SSBs are generally characterised by the presence of an oligonucleotide binding (OB) fold which is able to recognise single-stranded DNA (ssDNA) with high affinity. We discovered two news SSBs in humans (hSSB1 and hSSB2) that both contain a single OB domain followed by a divergent spacer region and a charged C-terminus. We have extensively characterised one of these, hSSB1 (NABP2/OBFC2B), in numerous important DNA processing events such as, in DNA double-stranded break repair and in the response to oxidative DNA damage. Although the structure of hSSB1 bound to ssDNA has recently been determined using X-ray crystallography, the detailed atomic level mechanism of the interaction of hSSB1 with ssDNA in solution has not been established. In this study we report the solution-state backbone chemical shift assignments of the OB domain of hSSB1. In addition, we have utilized NMR to map the DNA-binding interface of hSSB1, revealing major differences between recognition of ssDNA under physiological conditions and in the recently determined crystal structure. Our NMR data in combination with further biophysical and biochemical experiments will allow us to address these discrepancies and shed light onto the structural basis of DNA-binding by hSSB1 in solution

    hSSB1 (NABP2/ OBFC2B) is required for the repair of 8-oxo-guanine by the hOGG1-mediated base excision repair pathway

    Get PDF
    The maintenance of genome stability is essential to prevent loss of genetic information and the development of diseases such as cancer. One of the most common forms of damage to the genetic code is the oxidation of DNA by reactive oxygen species (ROS), of which 8-oxo-7,8-dihydro-guanine (8-oxoG) is the most frequent modification. Previous studies have established that human single-stranded DNA-binding protein 1 (hSSB1) is essential for the repair of double-stranded DNA breaks by the process of homologous recombination. Here we show that hSSB1 is also required following oxidative damage. Cells lacking hSSB1 are sensitive to oxidizing agents, have deficient ATM and p53 activation and cannot effectively repair 8-oxoGs. Furthermore, we demonstrate that hSSB1 forms a complex with the human oxo-guanine glycosylase 1 (hOGG1) and is important for hOGG1 localization to the damaged chromatin. In vitro, hSSB1 binds directly to DNA containing 8-oxoguanines and enhances hOGG1 activity. These results underpin the crucial role hSSB1 plays as a guardian of the genome

    Backbone and side-chain H-1, C-13 and N-15 resonance assignments of the OB domain of the single stranded DNA binding protein from Sulfolobus solfataricus and chemical shift mapping of the DNA-binding interface

    No full text
    Single stranded DNA binding proteins (SSBs) are present in all known cellular organisms and are critical for DNA replication, recombination and repair. The SSB from the hyperthermophilic crenarchaeote Sulfolobus solfataricus (SsoSSB) has an unusual domain structure with a single DNA-binding oligonucleotide binding (OB) fold coupled to a flexible C-terminal tail. This ‘simple’ domain organisation differs significantly from other known SSBs, such as human replication protein A (RPA). However, it is conserved in another important human SSB, hSSB1, which we have recently discovered and shown to be essential in the DNA damage response. In this study we report the solution-state backbone and side-chain chemical shift assignments of the OB domain of SsoSSB. In addition, using the recently determined crystal structure, we have utilized NMR to reveal the DNA-binding interface of SsoSSB. These data will allow us to elucidate the structural basis of DNA-binding and shed light onto the molecular mechanism by which these ‘simple’ SSBs interact with single-stranded DNA
    corecore