Ionizing radiation-induced DNA injury and damage detection in patients with breast cancer (2024)

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Ionizing radiation-induced DNA injury and damage detection in patientswith breast cancer (1)

Link to Publisher's site

Genet Mol Biol. 2015 Oct-Dec; 38(4): 420–432.

PMCID: PMC4763322

PMID: 26692152

Author information Article notes Copyright and License information PMC Disclaimer

Abstract

Breast cancer is the most common malignancy in women. Radiotherapy is frequently usedin patients with breast cancer, but some patients may be more susceptible to ionizingradiation, and increased exposure to radiation sources may be associated to radiationadverse events. This susceptibility may be related to deficiencies in DNA repairmechanisms that are activated after cell-radiation, which causes DNA damage,particularly DNA double strand breaks. Some of these genetic susceptibilities inDNA-repair mechanisms are implicated in the etiology of hereditary breast/ovariancancer (pathologic mutations in the BRCA 1 and 2 genes), but otherless penetrant variants in genes involved in sporadic breast cancer have beendescribed. These same genetic susceptibilities may be involved in negativeradiotherapeutic outcomes. For these reasons, it is necessary to implement methodsfor detecting patients who are susceptible to radiotherapy-related adverse events.This review discusses mechanisms of DNA damage and repair, genes related to thesefunctions, and the diagnosis methods designed and under research for detection ofbreast cancer patients with increased radiosensitivity.

Keywords: breast cancer, ionizing radiation, DNA damage, DNA double strand break, DNA repair analysis

Background

Breast cancer is the leading cause of cancer morbidity and death in women in developedcountries and countries with emerging economies (Ripperger et al., 2009; Youlden et al., 2012). According to Globocan, 1.67 millionnew cases of breast cancer were diagnosed in 2012 and ranks as the fifth cause of deathfrom cancer overall (522,000 deaths). A global increase has been estimated to around16,500 yearly new cases of this neoplasia by 2020. (Knaul et al., 2009)

Radiation therapy is an efficient treatment for cancer. About 50% of patients withmalignant breast tumors receive radiation therapy and most patients seem tolerate it,but some suffer severe adverse effects induced by the therapy. This variability ofresponse may be caused by several factors, like age, life style, inflammatory responses,oxidative stress, genetic predisposition and variants in genes involved in the responseto radiation-induced DNA damage (Smirnov etal., 2012; Hornhardt etal., 2014). Therefore, it is important to develop new diagnostictechniques for predicting responses to cancer treatment and for identifying patientssusceptible to radiation-related toxicity.

Any disturbance that results in the loss of genomic integrity may induce cell cyclederegulation and uncontrolled cell proliferation. Cells are continuously exposed to DNAdamaging agents and have developed mechanisms to respond to genome damage. Double-strandDNA breaks (DSB), although rare, are perhaps the most lethal mechanism and are oftenproduced by ionizing radiation (Pastink etal., 2001; Siever etal., 2003). The BRCA-1 and BRCA-2 proteins are involved in DSBdamage repair, and several mutations in these genes increase the risk for developingbreast and other neoplasias (Roy etal., 2012).

Ionizing Radiation-Associated DNA Damage, Radiotherapy and Mechanisms of DNARepair

Ionizing radiation effects in the cell

Ionizing radiation is a type of high-energy radiation that is able to releaseelectrons from atoms and molecules generating ions which can break covalent bonds.Ionizing radiation directly affects DNA structure by inducing DNA breaks,particularly, DSBs. Secondary effects are the generation of reactive oxygen species(ROS) that oxidize proteins and lipids, and also induce several damages to DNA, likegeneration of abasic sites and single strand breaks (SSB). Collectively, all thesechanges induce cell death and mitotic failure.

Ionizing radiation can be divided into X-rays, gamma rays, alpha and beta particlesand neutrons. Quiescent and slowly dividing cells are less radiosensitive, like thoseconstituting the nervous system, while cells with high proliferation rates are moreradiosensitive, like bone marrow, skin, and epithelial cells of the gastro-intestinaltract, among others. The radiation dose is measured in units gray (Gy), a measure ofthe amount of radiation absorbed by 1 kg of tissue (Dunne-Daly, 1999).

Radiotherapy

Radiotherapy is a treatment aimed at shrinking the tumor mass or at eliminatingresidual tumor cells by exposing the tumor to ionizing radiation. Radiotherapyregimes mostly use X- and gamma radiation (Masuda andKamiya, 2012). Radiation affects tumor and healthy irradiated cellsindistinctly. Radiotherapy is used as the standard treatment for breast cancer aftermastectomy; but this therapy may be also used prophylactically or palliatively toreduce the risk of tumor recurrence or to relieve symptoms caused by tumor growth andassociated metastases, respectively (Delaneyet al., 2005). Radiation therapy can be delivered byexternal-beam radiation or internal radiation. External-beam radiation therapy iscreated electronically by a linear accelerator which produces photon beams known asX-rays, with electric potentials in the range of 4 to 20 mega Volts. Patients receiveradiation doses in daily sessions for several weeks, and the radiation dose may beadministered in three different schemes: accelerated fractionation,hyperfractionation and hypofractionation. Accelerated fractionation means a radiationscheme in which the total dose of radiation is divided into small doses, and thetreatments are given more than once per day. The total dose of radiation isadministered in a shorter period of time (fewer days) compared to standard radiationtherapy (weeks). A reduction in the treatment time may reduce the repopulation oftumor cells, resulting in a better locoregional control. In hyperfractionedtreatment, the total radiation dose is divided into smaller doses, and it isadministered more than once a day; but in the same period as standard radiotherapy(days or weeks). Dose reduction may reduce the toxicity risk, although the total doseis increased. Hypofractionated radiation treatment is given once a day or less often.The total dose is divided into larger doses and is administered over a shorter periodthan standard radiotherapy. This scheme reduces patient visits and cost, and fewerside effects are noticed when compared to conventional radiation therapy.

The internal radiation therapy, also called brachytherapy, is released fromgamma-radiation sources such as radioactive isotopes like 60Co and137Cs, which are placed within the patient's body. This type ofradiation can deliver high doses of focalized radiation with an electric potential inthe range of 0.6 to 1 megaVolt and causes less damage to normal tissues (Patel and Arthur, 2006).

DNA repair after ionizing radiation

Ionizing radiation causes DSBs directly, but in addition base damages due to indirecteffects are also induced. This radiation causes formation of ROS (reactive oxygenspecies) which are indirectly involved in DNA damage. These ROS generates apurinic /apyrimidinic (abasic) sites in the DNA, SSBs, sugar moiety modifications, anddeaminated adducted bases (Redon etal., 2010; Aparicio etal., 2014). When DNA is damaged, the repair machinery of thecell is activated and stops the cell cycle at specific control checkpoints to repairDNA damage and prevent continuation of the cycle. It is known that the intrinsicradiosensitivity of tumor cells is strongly influenced by the cells DSB repaircapability (Mladenov et al.,2013). If tumor cells are able to efficiently repair the radiation damage,resistance to radiation develops, enabling cells to survive and replicate. If thedamage remains unrepaired, these mechanisms induce programmed cell death or apoptosisto prevent accumulation of mutations in daughter cells (Deckbar et al., 2011; Guo et al., 2011).

As mentioned, ionizing radiation inevitably reaches normal tissue, inducing bystandereffects in tumor-adjacent normal cells that may contribute to chromosomal aberrationsand to increase the risk for new malignancies. High doses of radiation may producetoxicity and reduce the patient's prognosis (Brownet al., 2015). Individual radiation treatment based onDSB repair capability could predict toxicity to surrounding tissues, therebyimproving treatment safety. DSB repair capability depends not just on gene integrity,but also on gene expression. In addition to germinal mutations affecting genes likeBRCA 1 and 2 or other related genes, genetic and epigeneticmechanisms may reduce or abrogate the expression of genes involved in DSB repair(Bosviel, et al., 2012).The DNA repair capability could be relevant to decide on the appropriate treatmentfor cancer patients, and functional tests may provide valuable information for theseclinical decisions.

DSB repair pathways

DSB repair is achieved in three ways: non-hom*ologous end joining (NHEJ), conservativehom*ologous recombination (HR) and single-strand alignment, also callednon-conservative hom*ologous recombination (SSA) (Langerak and Russell, 2011). HR is considered an error-free mechanismbecause it uses an undamaged DNA guide strand to repair the DSB, and the original DNAis reconstituted without loss of genetic information, but this mechanism proceedsslowly and is only exerted at the S/G2 phases of the cell cycle. NHEJ and SSA areconsidered error-prone and mutagenic mechanisms because the processing of DNA endsmay incur in loss or modification of genetic information at the repaired DSB ends.NHEJ is the most common mechanism of DSB repair in eukaryotic cells. In thismechanism, the DNA strands at the DSB are cut or modified, and the ends are ligatedtogether regardless of hom*ology, generating deletions or insertions. Although thisprocess is error-prone, this mechanism can fix the DNA damage quickly, because it isnot restricted to a single cell cycle phase, thus preventing increased geneticinstability (Do et al.,2014). These mechanisms are detailed below and in the Figure 1. The main proteins involved in the early steps of DSBdetection, chromatin remodeling and DNA repair are listed in Table 1.

Ionizing radiation-induced DNA injury and damage detection in patientswith breast cancer (2)

DSB repair pathways. In NHEJ, the KU70/KU80 heterodimer binds to the DSB,protects it from degradation by exonucleases, and acts as a repressor of HR.The KU70/80 heterodimer recruits and activates the DNA-PKcs and KU70 interactswith XRCC4. Then, the DNA ligase IV interacts with the KU heterodimer to ligatethe DNA ends. If required for ligation, PNKP binds to phosphorylated XRCC4 toprocess the DNA ends. In the HR pathway the MRN complex is recruited at the DSBends and CtIP binds to the MRN complex activating an exonuclease activity whichcreates single strand segments at the borders of the DSB that are extended bythe EXO1 3′- 5′ exonuclease. Then, hSSB1 binds to free ends and RPA (anheterometic complex formed by RPA70, RPA32 and RPA14) protects againstdegradation. RPA is replaced by RAD51-BRCA2. RAD51 nucleoprotein searches forand invades the hom*ologues sequences, from sister chromatid, to form a Hollidayjunction. The sister chromatids are joined by cohesin proteins to facilitatethe interconnection of the DSB to the hom*ologous recombination. Subsequently,RAD51 is removed leaving a free 3′-OH and DNA is synthesized by the DNApolymerase δ using the hom*ologous chromatid as a template. Resolvase enzymessolve the Holliday junction and the DNA ends are joined by DNA ligase I. TheSSA pathway is not conservative and depends on the presence of repeatedsequences flanking the DSB. In this mechanism, the MRN complex joined to CtIPcleaves the 5′-end of one strand of DNA to expose microhom*ology sequences.hom*ologous sequences are aligned, while nonaligned regions are removed by theERCC1/XPF nucleases. Then, DNA ends are joined by DNA ligase III.

Table 1

DNA repair and cell cycle control genes.

GeneNameFunctionCromosomal location
AKT1v-akt murine thymoma viral oncogenehom*olog 1Serine/threonine kinase. Regulatescomponents of the apoptotic machinery.14q32.32
ATMAtaxia telangiectasia mutatedSerine threonine protein kinase.Activates cell cycle checkpoints upon DSB induction acting as a DNAdamage sensor.11q22-q23
BAP1BRCA1 associated protein-1(ubiquitin carboxy-terminal hydrolase)Binds to BRCA1. Involved in cellcycle, response to DNA damage and chromatin dynamics.3p21.1
BIRP1BRCA1 protein interaction withc-terminal helicaseReceptor-interacting proteinforming a complex with BRCA1. Active during DSB repair.17q22.2
BRCA1Breast cancer 1DNA repair,ubiquitination andtranscriptional regulation to maintain genomic stability. Induces cellcycle arrests after ionizing irradiation.17q21
BRCA2Breast cancer 2Involved in DSB repair and/orhom*ologous recombination in meiosis.13q12
CDKsCell Division Protein KinaseCell cycle kinases.10q21.2
CDKN1BCyclin-dependent kinase inhibitor1BCell-cycle progression at G1.12p13.1-p12
CCND1Cyclin D1Regulates cell cycle duringG1/S, also interacts with a network of repair proteins including RAD51 toregulate HR11q13
CCND3Cyclin D3Regulates G1/S transition in cellcycle6p21.1
RBBP8Retinoblastoma Binding ProteinEndonuclease that functions withMRX complex in the first step of the DSB repair.18q11.2
EP3003 00 kDa E1A-Binding proteingeneRegulates transcriptionvia chromatin remodeling. Regulated by acetylation inDNA damage response.22q13.2
EXO1Exonuclease 15’-3’ Exonuclease1q43
FGFR2Fibroblast growth factor receptor2Cell surface tyrosine kinasereceptor regulating cell proliferation, migration and apoptosis.10q25.3-q26
HIST1H2BCHistone cluster 1, H2BCCore histone playing roles in DNArepair, replication and chromosomal stability.6p22.1
H2AXH2A Histone Family, Member XRequired for checkpoint-mediatedarrest of cell cycle progression in response to low doses of ionizingradiation and for efficient DSB repair when modified by C-terminalphosphorylation.11q23.3
KU70Thyroid Autoantigen 70 kDaBinding to DSB ends and inhibitionof exonuclease activity at these ends.22q13.2
LIG4Ligase IVDNA ligase involved in DNAnon-hom*ologous end joining (NHEJ) required for DSB repair.13q33.3
LSP1Lymphocyte-specific protein 1Actin binding protein F.11p15.5
MDC1Mediator of DNA Damage Checkpoint1Mediator-adaptor protein inresponse to DNA damage, active during the S and G2/M phases of cellcycle.6p21.3
MLL3Myeloid/lymphoid or mixed-lineageleukaemia 3Part of the ASCOM complex regulatedby acetylation to induce expression of p53 targets such as p21 in DNAdamage response.7q36.1
MRE11Meiotic Recombination 11Endonuclease, exonuclease, MRN/Xcomplex-5.11q21
NBN1NibrinComponent of the MRN/X complex.Plays a critical role in the cellular response to DNA damage and themaintenance of chromosome integrity. Regulator of cell cycle checkpointsin meiosis.8q21.3
PALB2Partner and localizer of BRCACritical role in HR repair byrecruiting BRCA2 and RAD51.16p12.1
PTENPhosphatase and tensin hom*ologTumor suppressor protein. Active inDNA repair through interactions with the Chk1 and the P53 pathways.Regulator of the RAD51 activity.10q23.3
RAD50RAD50 hom*olog SacharomycescerevisiaeProtein involved in DSBrepair, required for NHEJ and HR.5q23-q31
RAP80Ubiquitin Interaction MotifContaining 1Recognize ubiquitinated H2A andH2AX histones and recruits the BRCA1/BARD1 heterodimer at DSB.5q35.2
RB1RetinoblastomaTumor suppressor protein, mediatescell cycle arrest.17q22.2
Rif1RAP1 interacting factor hom*olog(yeast)Required for cell cycle arrest atS-phase in response to DNA damage.2q23.3
RNF168RING Finger ProteinE3 ubiquitin-protein ligaserequired for recruiting repair proteins to DNA damage sites.3q29
TGFβ1Transforming growth factor β1Multifunctional peptides thatregulate cell proliferation, migration, adhesion, differentiation, andother functions.19q13.1
TopBP1Topoisomerase (DNA) II BindingProteinS-phase checkpoint regulator.3q22.1
TOX3Tox high mobility group box familymember 3Involved in alteration of chromatinstructure.16q12.1
TP53Tumor protein p53Tumor suppressor protein, cellcycle arrest, apoptosis, senescence and DNA repair.17p13
XLF/CernunnosNon hom*ologous End-JoiningFactorScaffold protein. Serve as a bridgebetween XRCC4 and the other NHEJ factors.2q35
XRCC4X-Ray Repair ComplementingDefectiveScaffold protein involved inNHEJ.5q14.2
53BP1Tumor Protein P53 BindingProteinAdaptor protein, chromatin reader.Promotes NHEJ.15q15.3

Non-hom*ologous end joining (NHEJ)

Canonical NHEJ (C-NHEJ) is a conservative end-joining process, and this pathway isalso essential for V(D)J recombination during T- and B-cell lymphocytedevelopment. NHEJ is not restricted to a particular phase of the cell cycle, butoccurs preferentially during the G0, G1 and the early Sphases (Chistiakov et al.,2008; Deckbar et al.,2011; Malu et al.,2012a,b). NHEJ involves ligationof break DNA ends and does not require sequence hom*ology. The first step in theprocess is the recognition of the DNA ends by the KU heterodimer composed by theKU70 and KU80 proteins. The heterodimer binds to DNA ends and protects them fromfurther degradation (Williams etal., 2014). Crystallographic studies of the KU70/80heterodimer showed that it adopts a ring-shaped structure encircling the duplexDNA helix which reaches the DNA ends (Walkeret al., 2001). The KU subunits are similar in domainorganization; they have an amino-terminal von Willebrand domain participating inthe KU heterodimerization (Fell andSchild-Poulter, 2012). The KU70/80 heterodimer forms a scaffold at theDNA ends and recruits and activates the DNA-dependent protein kinase catalyticsubunit (DNA-PKcs). DNA-PKcs form a pincer-shaped structure which creates acentral channel mediating the ability of DNA-PKcs to bind double strand DNA (Sibanda et al., 2010; Davis et al., 2014).Subsequently, the X-ray repair complementing defective repair protein in Chinesehamster cells 4 (XRCC4) interacts with the KU70 subunit and another critical NHEJscaffolding protein, enabling enzymes to interact with the DSB region. DNA ligaseIV directly interacts with the KU heterodimer, an interaction mediated by thetandem BRCA1 C-terminal (BRCT) domains found in the C- terminus of DNA ligase IV(Ochi et al., 2014).Next, the PNKP (polynucleotide kinase-phosphatase) interacts with phosphorylatedXRCC4. Structural analysis showed that this scaffold forms filaments interactingwith the DNA ends and forms a bridge which stabilizes the ends of the DSB (Hammel et al., 2010; Ochi et al., 2014). It hasalso been shown that XRCC4 joins to unphosphorylated PNKP, but with less affinity.Other proteins, such as aprataxin, aprataxin and PNKP like factor (APLF), andXRCC4-like factor (XLF) also bind XRCC4.

Usually, DSB ends are irregular and show other defects, like abasic strandsegments that must be solved before NHEJ occurs. If phosphate or adenylate groupsare present at the DSB ends, DNA end processing may be required for subsequentligation. PNKP is a kinase/phosphatase responsible for adding phosphate to the 5‘OH end and remove the phosphate groups at the 3′ end (Bernstein et al., 2005). Aprataxin is anucleotide hydrolase and transferase which catalyzes the removal of adenylategroups covalently linked to 5′ phosphate termini (Grundy et al., 2013). When DSB asymmetries must befixed, the exonuclease Artemis is phosphorylated and binds to DNA-PKcs to trimredundant ends. KU has 5′deoxyribose-5-phosphate (5′-dRP)/AP lyase activityinvolved in cleaving redundant abasic single strands present at DSB ends (Roberts et al., 2010). TheWerner syndrome Rec Q helicase like protein (WRN) joins the KU heterodimer andXRCC4 and stimulate an exonuclease 3′ to 5′ activity (Gu et al., 2010; Malu et al., 2012). Sometimes filling of gapsin the strands at the DSB site is required, and this function may be accomplishedby the X family polymerases (μ and λ polymerases) (Capp et al., 2006, 2007).

When DSB ends of two DNA segments are clean and compatible they are ligated by DNAligase IV (Jahan et al.,2014). Ligase IV activity is stimulated by XRCC4 (Gu et al., 2007). Incompatible ends may bejoined by an interaction between ligase IV and XLF.

There is also an alternative NHEJ pathway (A-NHEJ) which is independent of theKU70/KU80 heterodimer activity. In this mechanism, DNA ends are excised by themeiotic recombination 11 protein (MRE11) and the retinoblastoma binding protein 8(RBBP8, synonymous of CtIP) exonucleases (Guet al., 2010, Hammelet al., 2010), exposing microhom*ology regions whichcan be aligned, allowing the filling of the empty segments by the X familypolymerases. Thereafter, XRCC1 and ligase III may complete the end-joining process(Frit et al., 2014).C-NHEJ is a more conservative end-joining process, but its efficacy may beaffected by the highly error-prone activity of the A-NHEJ pathway, theadaptability of the C-NHEJ to repair irregular ends, and the incompatibility ofsome DNA ends (Bétermier et al.,2014).

hom*ologous recombination (HR)

HR for DSB repair requires a hom*ologous DNA sequence provided by the sisterhom*ologous chromatid to restore a DSB lesion. Therefore, this process is onlyactive during the S and G2 cell-cycle phases, where this sister chromatid isavailable as a template (Krejci etal., 2012). HR starts with the binding of the MRN complex tothe DSB ends. The MRN complex is constituted by the MRE11 protein, the rad 50hom*olog S. cerevisiae protein (RAD50) and the nibrin protein(NBS1) (Richard et al.,2011a,b). Then, the 3 ‘ends ofthe DSB are digested by the exonuclease activity of the MRE11/CtIP to generatefree ends at the DSB that are extended by the EXO1 3′- 5′ exonuclease activity(Limbo et al., 2007).Subsequently, the single-strand DNA binding protein 1 (hSSB1) binds to the free 3’ends and joins the replication protein A (RPA) to protect these free ends fromfurther degradation, to prevent inappropriate annealing that could lead to genomicrearrangements and to prevent hairpin formation (Chen et al., 2013). RPA is a heterotrimeric complexformed by RPA70, RPA32 and RPA14 also involved in the control of DNA replicationand repair mechanisms (Sleeth etal., 2007). Then, RPA is replaced by an array of RAD51proteins assembled to eight BRC domains of the breast cancer 2 (BRCA2) protein andthe participation of five additional proteins (RAD51B/RAD51C/RAD51D/XRCC2/XRCC3)(West, 2003). Rad51 is a recombinasewhich forms a pre-synaptic RAD51-BRCA2 nucleoprotein filament on the DNA (Williams and Michael 2010). The RAD51-BRCA2nucleoprotein filaments search and invade the hom*ologues sequences to form aHolliday junction structure (Masson etal., 2001). The sister chromatids are joined by the cohesinproteins SMC1, 3, 5 and 6. These proteins facilitate the cohesion of the DSB andthe intact hom*ologous strands to propitiate the hom*ologous recombination (Kim et al., 2002, Kong et al., 2014). After theinvasion of the sister chromatid (synapses) and the alignment of hom*ologous DNAsequences, RAD51 is removed leaving a free 3′-OH end enabling the repairing DNAsynthesis by the DNA polymerase δ in the 3′-5′ direction with the help ofresolvases, like the structure-specific endonuclease subunit (MUS81), theessential meiotic structure-specific endonuclease 1 (EME1), and the Hollidayjunction 5′ flap endonuclease (GEN1) (Constantinouet al., 2002). Once the synthesis of the repairedDNA is completed, these enzymes resolve the Holliday junction and the DNA ends arejoined by the DNA ligase I (Matos and West2014). Although not completely understood, the BRCA1 protein plays animportant role in directing the scaffolding of the Rad51-BRCA2 filaments and alsointeracts with the histone H2AX (described below) during HR repair (O'Donovan and Livingston, 2010).

The HR repair method is considered error-free, because it uses the hom*ologoussequence of the sister chromatid as a template for synthesis. It has been proposedthat chromosome condensation makes it difficult to search for hom*ologous sequencesin the nucleus, and therefore NHEJ is more frequently employed by cells to repairDSB (Deckbar et al., 2011;Langerak and Russell, 2011). The highfidelity of HR is also proposed to explain the low sensitivity and cellularresistance of cells in S/G2 phase to ionizing radiation. Therefore it is suggestedthat resistance to radiotherapy is mediates by HR (Somaiah et al., 2013).

Single-strand alignment (SSA)

SSA can be regarded as a special form of HR repair. This repair mechanism is notconservative and is dependent on the presence of repeated sequences flanking theDSB. It begins with the cleavage of the 5′-end of one strand of DNA to exposemicrohom*ologies. This is mediated by a protein complex composed of the CtIP andthe MRN complex, followed by the alignment of the hom*ologous ends. Nonalignedregions are removed by the ERCC1/XPF nucleases (resulting in a loss of nucleotidesin the DNA chain) and then, the DNA ends are joined by the DNA ligase III (Salles et al., 2011; Liu et al., 2014). Evidencesuggests that SSA repair can elicit the formation of the pathological chromosometranslocations related with cancer (Manthey andBailis, 2010).

Radiosensitivity in Breast Cancer Patients

Radiosensitivity is the susceptibility of the cells or tissues to ionizing radiation.Some patients may be more sensitive to radiation. Sensitivity results from the toxiceffects of radiotherapy resulting in lesions of the patient's normal tissues. Theseeffects may be acute or late, depending on the time of their manifestation. Acuteeffects occur during the treatment or shortly after and they are usually reversibleand occur in rapidly proliferating tissues, like skin, gastrointestinal tract andhematopoietic tissues. Late effects manifest six months or later after the treatment.Late effects can be permanent, mainly affecting slowly proliferating tissues such askidneys, heart, and the nervous system, and may involve systemic deregulations of theendocrine system (Barnett et al.,2009). Radiation promotes DSB as mentioned above, and this damage isdetrimental for genome integrity (Chistiakovet al., 2008; Rübeet al., 2008; Henríquez-Hernández et al., 2011).

Mechanisms of hypersensitivity to ionizing radiation are still unclear, but isestimated that 70% of hypersensitivity cases are due to genetic variants (Turesson et al., 1996). Asmentioned above, mutations in the ATM gene are associated withextreme hypersensitivity to ionizing radiation (Masuda and Kamiya, 2012), and polymorphisms in genes likeXRCC3 and RAD51 increase the risk ofradiosensitivity (Vral et al.,2011). These genes are also implicated in breast cancer. Mayer et al. (2011) analyzedgene expression in peripheral blood lymphocytes of breast and cervical cancerpatients. They identified 153 genes altered by ionizing radiation. These genes areinvolved in cell cycle control and apoptosis in response to radiation. Of these, 67genes were useful to discriminate between normal reacting patients and subjects withsevere radiosensitivity. However, the analyses were performed on lymphocytes, and theauthors comment that an analysis of expression in different tissues would be requiredto define a more precise gene signature (Mayeret al., 2011).

The 7,8-dihydro-8-oxo-2′-deoxyguanosine (8-oxo-dG) base damage is produced byionizing radiation and is repaired by nucleotide excision followed by removal of thisabnormal deoxynucleoside out of the cell (Evanset al., 2010). 8-oxo-dG has been used as a urinarymarker of oxidative stress and has been associated with lung cancer (Il'yasova et al., 2012) andgastrointestinal diseases (Ock etal., 2012). It has also been proposed as a marker forradiosensitivity (Erhola et al.,1997, Roszkowski and Olinski, 2012).Haghdoost et al. (2001)studied 8-oxo-dG urinary levels in breast cancer patients before and after adjuvantradiotherapy (4 to 6 Gy). Radiosensitive patients showed skin redness in the radiatedareas and significantly increased urinary levels of 8-oxo-dG, and these authorsproposed the use of this deoxynucleoside as a urinary biomarker for radiosensitivity.This biomarker facilitates the study of individual radiosensitivity, since theabnormal metabolite maybe measured by ELISA (Haghdoost et al., 2001). In a study by Skiöld et al. (2013),radiation-induced oxidative stress response was analyzed by the 8-oxo-dG biomarker inserum from ex-vivo irradiated leukocytes samples obtained frombreast cancer patients that developed severe acute skin reactions (RTOG [RadiotherapyOncology Group Criteria] grade 3-4) during radiotherapy and from patients with breastcancer showing no early skin reactions after radiotherapy (RTOG grade 0). The authorsdemonstrated that patients with RTGO grade 0 showed increased extracellular serumlevels of 8-oxo-dG, in contrast with the significantly low serum levels observed inpatients with RTOG grades 3 and 4, indicating that 8-oxo-dG is a useful biomarker toanalyze cellular responses to ionizing radiation (Skiöld et al., 2013). Nonetheless, 8-oxo-dG can alsoresult from cell exposure to oxidative stress by ROS, as may occur when tissues areexposed to environmental pollutants (Hecht,1999). For these reasons this biomarker is not specific for ionizingradiation but, as in the case of the studies by Skiöld et al. (2013), it is helpful as a comparativeex vivo test of irradiated cells to define the biological effectsof ionizing radiation. Extracellular levels of 8-oxo-dG are appropriate indicators ofthe cells capability to repair the DNA damage caused by ROS.

Certain phenotypes of breast cancer have been associated with locoregional recurrence(LRR). Brollo et al. (2013)suggested that HER2+ tumors are more susceptible to ionizing radiation, while Voduc et al. (2010) observedthat LRR seemed higher in patients with triple negative marker breast cancer,although the number of LRR events was small. At present, there are no molecularmethods to discriminate between patients with high and low LRR (Britten et al., 2013). In addition, there is notenough information regarding the possible adverse effects of radiotherapy that mayinduce genomic and epigenetic modifications and changes in gene-expression profilesin breast cancer.

Henríquez-Hernández et al.(2011) analyzed isolated peripheral blood lymphocytes (PBLs) from patientswith advanced breast cancer treated ex vivo with high radiotherapydoses to study ionizing radiation resistance. They showed that lymphocytes frompatients with low DNA damage and high apoptosis rates had low risks of radiationadverse events.

Studies analyzing the type of repair that occurs when cells are exposed to radiationand the correlation with abnormal expression of certain genes involved in DSB repairhave also been conducted. In vitro studies of Bca11 (familial breastcancer cell line) and Bca10 (sporadic breast cancer cell line) cell lines showed highNHEJ repair activity and direct HR non-conservative repair in the Bca11 cell line.The Bca10 cell line also showed an increase in non-conservative repair of direct HR,but to a lesser degree than Bca11. Consequently, repair mechanisms in these celllines may cause deletions in the DNA sequence and cell cycle deregulation (Keimling et al., 2008). Theseauthors performed a study in PBLs from patients with sporadic breast cancer, healthywomen with familial risk of breast cancer, and healthy controls, and theydemonstrated increased NHEJ and SSA in both, cancer patients and subjects athereditary risk, vs. the healthy controls. This study suggested thatthese two groups are prone to extended non-conservative DSB repairing mechanisms.Based on these results, Keimling etal. (2012) implemented a test to analyze DSB repair invitro.

Techniques for DSB Repair Analysis

Some tests have been devised to assess DNA damage in response to diverse substances,microorganisms, or environmental conditions. Some of these tests are describedbelow.

Comet assay

The alkaline comet assay involves measurement of DNA damage in SSB and DSB. Thismethod is fast and cheap. It provides important information about the risk ofdiseases related to oxidative stress (Alapetiteet al., 1999; Dusinskaand Collins, 2008). In this assay, cells are embedded in a thin layer ofa*garose on a thin glass slide, cells are lysed in a solution containing detergent andNaCl, releasing the DNA from the proteins bound to it, but leaving DNA fragmentsstill attached to the nuclear membrane. Then, the plate is incubated in an alkalinesolution, an electrophoresis is run and DNA is stained with ethidium bromide. DNAfragments travel to the anode forming a comet-like image when viewed by fluorescencemicroscopy (Fikrová et al.,2011, Baumgartner et al.,2012). The image of the comet head denotes the DNA content and the tail thefrequency of DNA breaks (Figure 2B). Softwareprograms designed to analyze the comet image allow measurement of DNA content andtail length. The length of the comet tail correlates with the level of DNAdamage.

Ionizing radiation-induced DNA injury and damage detection in patientswith breast cancer (3)

General assays for detecting DNA damage (A)Immunohistochemistry with antibodies directed against γ-H2AX: peripheral bloodmononuclear cells are isolated, nuclei are stained with DAPI and withantibodies directed at γ-stained H2AX and visualized under fluorescentmicroscopy. (B) Comet assay: the comet assay is also performed onmononuclear cells. The cells are embedded in agarose on a thin glass slide,cells are lysed and incubated in an alkaline solution. Subsequently, DNAfragments are separated by electrophoresis and stained with ethidium bromide.The comet-like image is viewed under a fluorescence microscope. The length ofthe comet tail indicates the frequency of DNA breaks

Hair et al. (2010) used amodified comet assay method in which slides with cells embedded in agarose wereincubated with three different treatments: 1) alkaline electrophoresis to detect SSBinduced radiation and alkaline-labile sites; 2) electrophoresis of cells treated withformamidopyrimidine [Fapy] -DNA glycosylase (Fpg); this releases the damaged purines,leaving apurinic sites (AP sites) that are subsequently cleaved with the cellular APlyase, producing single strand fragments which can be visualized in the comet assay,and 3) electrophoresis after treatment of the cells with bacterial endonucleaseEndoIII, which cleaves the damage strands at sites presentingoxidized pyrimidines, thus increasing the sensitivity of the comet assay by leavinggaps in mutated bases (Hair et al.,2010).

Some disadvantages of the comet assay are the variability between different protocolsand between laboratories, which makes it difficult to define ionizing radiationtoxicities, so this issue will require adoption of standardized and comparableprotocols (Forchhammer et al.,2010; Henríquez-Hernández etal., 2012; Azqueta etal., 2014). Sirota etal. (2014) studied inter-laboratory variation of comet assayfactors, like slide brands, duration of alkali treatment and electrophoresisconditions, and they found that laboratory differences were associated withelectrophoresis conditions, especially the temperature during alkalineelectrophoresis, which affects the rate of conversion of alkali labile sites tosingle stranded breaks (Sirota etal., 2014). Additionally, it has been suggested thatimplementation of a standard software will be required for comet assay interpretation(Fikrová et al.,2011).

γ-H2AX

The histone H2AX variant of the histone H2A is present in subsets of nucleosomes (2to 25% of the total H2A) and has been implicated in DSB repair. When H2AX isphosphorylated at the serine residue 139 by phosphoinositide-3-kinase-related proteinkinases (PIKKs), the phosphate group adopts a γ position in the protein, constitutingthe gamma H2AX (γ-H2AX) configuration (Rogakouet al., 1998; Rothkammand Horn, 2009). This phosphoprotein acts in early events of DNA repair bydecondensing the chromatin near the DSB (Kruhlaket al., 2006). Additionally, γ H2AX joins to the DSBends forming a “γH2AX focus” which is extended for several Mb at the sides of theDSB. A method used for the analysis of DNA damage is the measurement of γ-H2AX usingantibodies against

In the γ-H2AX assays, peripheral blood is collected and mononuclear cells areseparated and fixed on a glass surface. Then, an immunohistochemistry withanti-γ-H2AX antibody is performed and the results are analyzed by fluorescencemicroscopy in which fluorescent foci are measured (Figure 2A). This test may be also analyzed by flow cytometry or by westernblot (Kinner et al., 2008;Dickey et al., 2009; Podhorecka et al., 2010).

γ-H2AX foci measurements in patients before and after radiotherapies using low andhigh doses of ionizing radiation have shown a linear relationship between DNA damageand exposure to radiation. The initial number of γ-H2AX foci is consistent with DSBsin the cells. After a while, the γ-H2AX foci disappear due to the DNA repair (Rübe et al., 2008; Horn et al., 2011). This methodis sensitive for measuring DNA repair in patients undergoing radiotherapy, but it isalso applied in other fields, such as DNA damage analysis due to occupationalexposure or contact with environmental pollutants, cigarette smoke, drugs, etc‥ It isimportant to note that these co-exposures may affect the results in radiotherapypatients and, hence, should be considered on an individual basis. Furthermore,phosphorylation of H2AX is observed in the absence of DSB in the replication process,in mitosis and during DNA fragmentation in apoptosis. Therefore, the test must beable to distinguish between apoptotic and non-apoptotic cells (Dickey et al., 2009).

Comet assay and γ-H2AX methods described above help to assess DNA damage and repair,but do not allow discrimination of the type of damage, like SSB or DSB. It is alsoimportant to analyze whether the damage is repaired and what kind of repair mechanismis operating to assess whether cells are sensitive or resistant to ionizingradiation.

Engineered proteins to detect spontaneous DSB

Shee et al. (2013) developeda new synthetic technology to quantify DSBs in bacterial and mammalian cells. Thismethod use the green fluorescent-protein (GFP) fused to the GAM protein (GAM-GFP), aviral protein from bacteriophage Mu, which shares sequence hom*ology with theeukaryotic proteins KU80 and KU70 involved in NHEJ (Aparicio et al., 2014). Unlike the KU protein, the GAMprotein is not involved in DNA repair reactions. GAM binds to DNA and inhibits avariety of exonucleases involved in DNA repair (Abraham and Symonds, 1990; fa*gagnaet al., 2003; Sheeet al., 2013). This advance allows the study andquantification of DNA breaks. In this method, the I-SceIendonuclease is used to make site specific DSBs and cells are transfected with a MuGAM-GFP fusion expression vector. The GAM-GFP protein joins the DSBs formed by theI-SceI treatment, generating fluorescence at the damaged siteswhich can be analyzed by fluorescence microscopy. Since the GAM-GFP protein competeswith KU proteins, this results in low levels of DNA damage, thus limiting thistechnology to the study of DSB repair by HR (Sheeet al., 2013).

Identification of repair mechanisms by specific DNA substrates

As mentioned above, Keimling et al.(2012) developed an in vitro method in which PBLs aretransfected with marker plasmids for enabling discrimination of the mechanismsinvolved in DSB repair: HR, NHEJ, and SSA (Figure3A). In this procedure, PBLs are transduced in three different experimentswith separate plasmids, each containing the EGFP reporter gene followedby differentsequences amenable to undergo one of the different mechanisms of DNA repair definedabove. Cells in the three groups are co-transduced with a plasmid codifying forI-SceI as the inductor of DSB repair events. Fluorescencedetection after 24 h by flow cytometry in any of the three transduced cells of thepanel measures the events of each individual operating mechanism, allowing moredetailed information about DSB repair in individual patients (Figure 3B). This test is amenable for high-throughput sampleprocessing and analysis (Boehden etal., 2002; Keimling etal., 2012).

Ionizing radiation-induced DNA injury and damage detection in patientswith breast cancer (4)

Specific assays for detecting DNA damage (A) The EJ-EGFPplasmids contains a mutated version of the EGFP gene (green light bar) createdby inserting a restriction site for the meganuclease I-SceIflanked by a 5 bp microhom*ology sites (black arrows); this plasmid was designedto be repaired by NHEJ. The Δ-EGFP/3’EGFP and Δ-EGFP/5’EGFP plasmids contain anarray of an EGFP mutated gene containing an I-SceI site (greenlight bar) followed by a spacer (purple bar) and EGFP gene versions truncatedat their flanking 3’ and 5’ ends, respectively (dark green bars) which allowthe reconstitution of the wild-type version of the marker geneby SSA and HR, respectively. (B) Analysis of DSB repair: The assayis performed in three cultures of peripheral blood lymphocytes (PBLs),transduced separately with each of the plasmid versions designed fordiscrimination of SSA, NHEJ and HR. The cultures are co-transduced with anadditional plasmid expressing the I-SceI enzyme. Aftergenerating DBS in the target plasmids by the expressed restriction enzyme, DNArepair in PBLs repair by each of the different DNA repair pathway may bemonitored by restoration of the wild-type version of EGFP 24 h aftertransduction by measuring EGFP florescence by flow cytometry.

Conclusions

Detection of genetic alterations in genes associated with breast cancer, particularlygenes related to DSB repair, may allow the diagnosis for genetic patients with breastcancer, but current methods based on genomic methodologies to detect mutations areexpensive and not suitable for screening subjects under risk for increased DSB events.Almost 20% of the breast cancer patients will show acute complications due toradiotherapy. Hence, evaluation of DSB repair is a useful tool for assessing breastcancer risk and predicting the response and complications associated with conventionalradiotherapy. Methods for studying DSB repair in PBLs are less expensive and suitablefor designing high-throughput analyses for screening subjects at high risk for cancer ingeneral, to anticipate adverse events and to offer individualized therapies. Thesemethods will be relevant for preventing unnecessary radiation exposure, for screening ofpatients which will not benefit from radiotherapy, and for adjusting radiotherapyregimes in patients requiring this therapeutic option, in order to avoid adverse effectsassociated with DSB in tissues that can ameliorate a patient's prognosis.

A general comparison of methods shows that the comet assay assesses the amount of DNAdamage, is inexpensive and is easy to perform in conventional laboratories. However itdoes not provide detailed information about the DNA lesion (SSB or DSB) and neither theDSB repair mechanism (NHEJ, SSA or HR). Another disadvantage of this method is theinter-protocol and the inter-laboratory variability in results. Nonetheless, this testis useful as a preliminary tool for assessing DNA damage. Detection of γ-H2AX is also asimple procedure and measurement of γ-H2AX may be performed by fluorescent microscopy,but the technique is also amenable for flow cytometry or western blot assays, which mayrender a more precise quantification than the comet assay. However, the detection ofγ-H2AX does not discriminate between SSB and DSB. Furthermore, γ-H2AX may bephosphorylated during mitosis or apoptosis, resulting in false positives. The methoddeveloped by Shee et al. (2013)is more sensitive for DSB detection. It uses the GAM protein linked to EGFP, which joinsthe ends of the DSB and prevents DNA repair. Cells with DSB may be measured byfluorescent microscopy or flow cytometry. This technique requires molecular and cellbiology techniques which may constitute an obstacle for diagnostic laboratories. Themethod developed by Keimling et al.(2012) enables the discrimination and measurement of the type of DSB repairmechanism. This method also uses techniques of molecular and cell biology, which maycomplicate its implementation in diagnostic laboratories, but this refined technologymay have a great impact in defining a patient's risk to DSB induced by ionizingradiation.

Further advances in the discovery of genes involved in DNA repair and additional factorsaffecting genome stability will prompt the implementation of better technologies tostudy DNA damage in the clinical setting so as to avoid radiation-relatedtoxicities.

Acknowledgments

This work received sponsorship from the PAICYT-UANL CS943-11 call for research.

Footnotes

Associate Editor: Carlos F. M. Menck

References

  • Abraham ZH, Symonds N. Purification of overexpressed gam gene protein from bacteriophage Muby denaturation-renaturation techniques and a study of its DNA-bindingproperties. Biochem J. 1990;269:679–684. [PMC free article] [PubMed] [Google Scholar]
  • Alapetite C, Thirion P, De la Rochefordie A, Cosset JM, Moustacchi E. Analysis by alkaline comet assay of cancer patients with severereactions to radiotherapy: Defective rejoining of radioinduced dna strand breaksin lymphocytes of breast cancer patients. Int J Cancer. 1999;90:83–90. [PubMed] [Google Scholar]
  • Aparicio T, Baer R, Gautier J. DNA double-strand break repair pathway choice andcancer. DNA Repair. 2014;19:169–175. [PMC free article] [PubMed] [Google Scholar]
  • Azqueta A, Slyskova J, Langie SAS, Gaivão ION, Collins A. Comet assay to measure DNA repair: Approach andapplications. Front Genet. 2014;5:1–8. [PMC free article] [PubMed] [Google Scholar]
  • Barnett GC, West CML, Dunning AM, Elliott RM, Coles CE, Pharoah PDP, Burnet NG. Normal tissue reactions to radiotherapy: Towards tailoring treatmentdose by genotype. Nat Rev Cancer. 2009;9:134–142. [PMC free article] [PubMed] [Google Scholar]
  • Baumgartner A, Kurzawa-Zegota M, Laubenthal J, Cemeli E, Anderson D. Comet-assay parameters as rapid biomarkers of exposure todietary/environmental compounds an in vitro feasibility study on spermatozoa andlymphocytes. Mutat Res. 2012;743:25–35. [PubMed] [Google Scholar]
  • Bernstein NK, Williams RS, Rakovszky ML, Cui D, Green R, Karimi-Busheri F, Mani RS, Galicia S, Koch CA, Cass CE, et al. The molecular architecture of the mammalian DNA repair enzyme,polynucleotide kinase. Mol Cell. 2005;17:657–670. [PubMed] [Google Scholar]
  • Bétermier M, Bertrand P, Lopez BS. Is non-hom*ologous end-joining really an inherently error-proneprocess. PLoS Genet. 2014;10: [PMC free article] [PubMed] [Google Scholar]
  • Boehden GS, Su S, Rimek A, Preuss U, Scheidtmann K, Wiesmu L. DNA substrate dependence of p53-mediated regulation of double-strandbreak repair. Mol Cell Biol. 2002;22:6306–6317. [PMC free article] [PubMed] [Google Scholar]
  • Bosviel R, Garcia S, Lavediaux G, Michard E, Dravers M, Kwiatkowski F, Bignon Y, Bernard-Gallon DJ. BRCA1 promoter methylation in peripheral blood DNA was identified insporadic breast cancer and controls. Cancer Epidemiol. 2012;36:177–182. [PubMed] [Google Scholar]
  • Britten A, Rossier C, Taright N, Ezra P, Bourgier C. Genomic classifications and radiotherapy for breastcancer. Eur J Pharmacol. 2013;717:67–70. [PubMed] [Google Scholar]
  • Brollo J, Kneubil MC, Botteri E, Rotmensz N, Duso BA, Fumagalli L, Locatelli MA, Criscitiello C, Lohsiriwat V, Goldhirsch A, et al. Locoregional recurrence in patients with HER2 positive breastcancer. Breast. 2013;22:856–862. [PubMed] [Google Scholar]
  • Brown LC, Mutter RW, Halyard MY. Benefits, risks, and safety of external beam radiation therapy forbreast cáncer. Int J Womens Health. 2015;7:449–458. [PMC free article] [PubMed] [Google Scholar]
  • Capp JP, Boudsocq F, Bertrand P, Laroche-Clary A, Pourquier P, Lopez BS, Cazaux C, Hoffmann JS, Canitrot Y. The DNA polymerase λ is required for the repair of non-compatible DNAdouble strand breaks by NHEJ in mammalian cells. Nucleic Acids Res. 2006;34:2998–3007. [PMC free article] [PubMed] [Google Scholar]
  • Capp JP, Boudsocq F, Besnard AG, Lopez BS, Cazaux C, Hoffmann JS, Canitrot Y. Involvement of DNA polymerase μ in the repair of a specific subset ofDNA double-strand breaks in mammalian cells. Nucleic Acids Res. 2007;35:3551–3560. [PMC free article] [PubMed] [Google Scholar]
  • Chen H, Lisby M, Symington L. RPA coordinates DNA end resection and prevents formation of DNAhairpins. Mol Cell. 2013;50:589–600. [PMC free article] [PubMed] [Google Scholar]
  • Chistiakov DA, Voronova NV, Chistiakov PA. Genetic variations in DNA repair genes, radiosensitivity to cancer andsusceptibility to acute tissue reactions in radiotherapy-treated cancerpatients. Acta Oncol. 2008;47:809–824. [PubMed] [Google Scholar]
  • Constantinou A, Chen XB, McGowan CH, West SC. Holliday junction resolution in human cells: Two junctionendonucleases with distinct substrate specificities. EMBO J. 2002;21:5577–5585. [PMC free article] [PubMed] [Google Scholar]
  • Davis AJ, Chen BPC, Chen DJ. DNA-PK: A dynamic enzyme in a versatile DSB repairpathway. DNA Repair. 2014;17:21–29. [PMC free article] [PubMed] [Google Scholar]
  • Deckbar D, Jeggo PA, Löbrich M. Understanding the limitations of radiation-induced cell cyclecheckpoints. Crit Rev Biochem Mol Biol. 2011;46:271–283. [PMC free article] [PubMed] [Google Scholar]
  • Delaney G, Jacob S, Featherstone C, Barton M. The role of radiotherapy in cancer treatment estimating optimalutilization from a review of evidence-based clinical guidelines. Cancer. 2005;104:1129–1137. [PubMed] [Google Scholar]
  • Dickey JS, Redon CE, Nakamura AJ, Baird BJ, Sedelnikova OA, Bonner WM. H2AX: Functional roles and potential applications. Chromosoma. 2009;118:683–692. [PMC free article] [PubMed] [Google Scholar]
  • Dusinska M, Collins AR. The comet assay in human biomonitoring: Gene-environmentinteractions. Mutagenesis. 2008;23:191–205. [PubMed] [Google Scholar]
  • Do TA, Brooks JT, Le Neveu MK, La Rocque JR. Double-strand break repair assays determine pathway choice andstructure of gene conversion events in Drosophila melanogaster. G3. 2014;4:425–432. [PMC free article] [PubMed] [Google Scholar]
  • Dunne-Daly CF. Principles of radiotherapy. Br J Hosp Med (Lond) 1999;74:C166–C169. [PubMed] [Google Scholar]
  • Erhola M, Toyokuni S, Okada K, Tanaka T, Hiai H, Ochi H, Uchida K, Osawa T, Nieminen MM, Alho H, et al. Biomarker evidence of DNA oxidation in lung cancer patients:Association of urinary 8-hydroxy-2’-deoxyguanosine excretion with radiotherapy,chemotherapy, and response to treatment. FEBS Lett. 1997;409:287–291. [PubMed] [Google Scholar]
  • Evans MD, Saparbaev M, Cooke MS. DNA repair and the origins of urinary oxidized 2’-deoxyribonucleosides. Mutagenesis. 2010;25:433–442. [PubMed] [Google Scholar]
  • fa*gagna FA, Weller GR, Doherty AJ, Jackson SP. The Gam protein of bacteriophage Mu is an orthologue of eukaryoticKu. EMBO Rep. 2003;4:47–52. [PMC free article] [PubMed] [Google Scholar]
  • Fell VL, Schild-Poulter C. Ku regulates signaling to DNA damage response pathways through theKu70 von Willebrand a domain. Mol Cell Biol. 2012;32:76–87. [PMC free article] [PubMed] [Google Scholar]
  • Fikrová P, Stetina R, Hronek M, Hyspler R, Tichá A, Zadák Z. Application of the comet assay method in clinicalstudies. Wien Klin Wochenschr. 2011;123:693–699. [PubMed] [Google Scholar]
  • Forchhammer L, Johansson C, Loft S, Godschalk RWL, Sabine A, Langie S, Jones GDD, Kwok RWL, Collins AR, Azqueta A, et al. Variation in the measurement of DNA damage by comet assay measured bythe ECVAG y inter-laboratory validation trial. Mutagenesis. 2010;25:113–123. [PubMed] [Google Scholar]
  • Frit P, Barboule N, Yuan Y. Alternative end-joining pathway(s): Bricolage at DNAbreaks. DNA Repair. 2014;17:81–97. [PubMed] [Google Scholar]
  • Grundy GJ, Rulten SL, Zeng Z, Arribas-Bosacoma R, Iles N, Manley K, Oliver A, Caldecott KW. APLF promotes the assembly and activity of non-hom*ologous end joiningprotein complexes. EMBO J. 2013;32:112–125. [PMC free article] [PubMed] [Google Scholar]
  • Gu J, Lu H, Tippin B, Shimazaki N, Goodman MF, Lieber MR. XRCC4:DNA ligase IV can ligate incompatible DNA ends and can ligateacross gaps. EMBO J. 2007;26:1010–1023. [PMC free article] [PubMed] [Google Scholar]
  • Gu J, Li S, Zhang X, Wang LC, Niewolik D, Schwarz K, Legerski RJ, Zandi E, Lieber MR. DNA-PKcs regulates a single-stranded DNA endonuclease activity ofArtemis. DNA Repair. 2010;9:429–437. [PMC free article] [PubMed] [Google Scholar]
  • Guo G-S, Zhang F-M, Gao R-J, Delsite R, Feng Z-H, Powell SN. DNA repair and synthetic lethality. Int J Oral Sci. 2011;3:176–179. [PMC free article] [PubMed] [Google Scholar]
  • Hair JM, Terzoudi GI, Hatzi VI, Lehockey KA, Srivastava D, Wang W, Pantelias GE, Georgakilas AG. BRCA1 role in the mitigation of radiotoxicity and chromosomalinstability through repair of clustered DNA lesions. Chem Biol Interact. 2010;188:350–358. [PubMed] [Google Scholar]
  • Hammel M, Yu Y, Fang S, Lees-Miller SP, Tainer JA. XLF regulates filament architecture of the XRCC4. Ligase IVcomplex. Structure. 2010;18:1431–1442. [PMC free article] [PubMed] [Google Scholar]
  • Haghdoost S, Svoboda P, Ingemar N. Can 8-oxo-dg be used as a predictor for individualradiosensitivity? Int J Radiat Oncol Biol Phys. 2001;50:405–410. [PubMed] [Google Scholar]
  • Hecht SS. DNA adduct formation from tobacco-specificN-nitrosamines. Mutat Res. 1999;424:127–142. [PubMed] [Google Scholar]
  • Henríquez-Hernández LA, Carmona-Vigo R, Pinar B, Bordón E, Lloret M, Núñez MI, Rodríguez-Gallego C, Lara PC. Combined low initial DNA damage and high radiation-induced apoptosisconfers clinical resistance to long-term toxicity in breast cancer patientstreated with high-dose radiotherapy. Radiat Oncol. 2011;6:1–8. [PMC free article] [PubMed] [Google Scholar]
  • Henríquez-Hernández LA, Bordón E, Pinar B, Lloret M, Rodríguez-Gallego C, Lara PC. Prediction of normal tissue toxicity as part of the individualizedtreatment with radiotherapy in oncology patients. Surg Oncol. 2012;21:201–206. [PubMed] [Google Scholar]
  • Horn S, Barnard S, Rothkamm K. Gamma-H2AX-based dose estimation for whole and partial body radiationexposure. PloS One. 2011;6: [PMC free article] [PubMed] [Google Scholar]
  • Hornhardt S, Rößler U, Sauter W, Rosenberger A, Illig T, Bickeböller H, Wichmann H, Gomolka M. Genetic factors in individual radiation sensitivity. DNA Repair. 2014;16:54–65. [PubMed] [Google Scholar]
  • Il'yasova D, Scarbrough P, Spasojevic I. Urinary biomarkers of oxidative status. Clin Chim Acta. 2012;413:1446–1453. [PMC free article] [PubMed] [Google Scholar]
  • Jahan F, Kweon J, Wang Y, Han E, Kan Y, Lichter N, Weisensel N, Hendrickson EA. A role for XLF in DNA repair and recombination in human somaticcells. DNA Repair. 2014;15:39–53. [PMC free article] [PubMed] [Google Scholar]
  • Keimling M, Kaur J, Bagadi SAR, Kreienberg R, Wiesmüller L, Ralhan R. A sensitive test for the detection of specific DSB repair defects inprimary cells from breast cancer specimens. Int. J Cancer. 2008;123:730–736. [PubMed] [Google Scholar]
  • Keimling M, Deniz M, Varga D, Stahl A, Schrezenmeier H, Kreienberg R, Hoffmann I, König J, Wiesmüller L. The power of DNA double-strand break (DSB) repair testing to predictbreast cancer susceptibility. FASEB J. 2012;26:2094–2104. [PubMed] [Google Scholar]
  • Kim JS, Krasieva TB, LaMorte V, Malcolm A, Taylor R, Yokomori K. Specific recruitment of human cohesin to laser-induced DNAdamage. J Biol Chem. 2002;277:45149–45153. [PubMed] [Google Scholar]
  • Kinner A, Wu W, Staudt C, Iliakis G. Gamma-H2AX in recognition and signaling of DNA double-strand breaks inthe context of chromatin. Nucleic Acids Res. 2008;36:5678–5694. [PMC free article] [PubMed] [Google Scholar]
  • Knaul FM, Nigenda G, Lozano RCM, Arreola-Ornelas H, Langer A, Frenk J. Cáncer de mama en México: Una prioridad apremiante. Salud Públ Mex. 2009;51:335–344. [PubMed] [Google Scholar]
  • Kong X, Ball AR, Jr, Pham HX, Zeng W, Chen H, Schmiesing JA, Kim J, Berns M, Yokomori K, Ball AR, et al. Distinct functions of human Cohesin-SA1 and Cohesin-SA2 in doublestrand break repair. Cell Mol Biol. 2014;34:685–698. [PMC free article] [PubMed] [Google Scholar]
  • Krejci L, Altmannova V, Spirek M, Zhao X. hom*ologous recombination and its regulation. Nucleic Acids Res. 2012;40:5795–5818. [PMC free article] [PubMed] [Google Scholar]
  • Kruhlak MJ, Celeste A, Dellaire G, Fernandez-Capetillo O, Müller WG, McNally JG, Bazett-Jones DP, Nussenzweig A. Changes in chromatin structure and mobility in living cells at sitesof DNA double-strand breaks. Int J Cell Biol. 2006;172:823–834. [PMC free article] [PubMed] [Google Scholar]
  • Langerak P, Russell P. Regulatory networks integrating cell cycle control with DNA damagecheckpoints and double-strand break repair. Philos Trans R Soc Lond B Biol Sci. 2011;366:3562–3571. [PMC free article] [PubMed] [Google Scholar]
  • Limbo O, Chahwan C, Yamada Y, Bruin RAM, Wittenberg C, Russell P. Ctp1 is a cell cycle-regulated protein that functions with Mre11complex to control double-strand break repair by hom*ologousrecombination. Mol Cell. 2007;28:134–146. [PMC free article] [PubMed] [Google Scholar]
  • Liu C, Srihari S, Cao K-AL, Chenevix-Trench G, Simpson PT, Ragan MA, Khanna KK. A fine-scale dissection of the DNA double-strand break repairmachinery and its implications for breast cancer therapy. Nucleic Acids Res. 2014;42:6106–6127. [PMC free article] [PubMed] [Google Scholar]
  • Malu S, De Ioannes P, Kozlov M, Greene M, Francis D, Hanna M, Pena J, Escalante CR, Kurosawa A, Erdjument-Bromage H, et al. Artemis C-terminal region facilitates V(D)J recombination through itsinteractions with DNA Ligase IV and DNA-PKcs. J Exp Med. 2012a;209:955–963. [PMC free article] [PubMed] [Google Scholar]
  • Malu S, Malshetty V, Francis D, Cortes P. Role of non-hom*ologous end joining in V(D)Jrecombination. Immunol Res. 2012b;54:233–246. [PubMed] [Google Scholar]
  • Manthey GM, Bailis AM. Rad51 inhibits translocation formation by non-conservative hom*ologousrecombination in Saccharomyces cerevisiae. PloS One. 2010;5: [PMC free article] [PubMed] [Google Scholar]
  • Masson JY, Tarsounas MC, Stasiak AZ, Stasiak A, Shah R, McIlwraith MJ, Benson FE, West SC. Identification and purification of two distinct complexes containingthe five RAD51 paralogs. Genes Dev. 2001;8:3296–3307. [PMC free article] [PubMed] [Google Scholar]
  • Masuda Y, Kamiya K. Molecular nature of radiation injury and DNA repair disordersassociated with radiosensitivity. Int J Lab Hematol. 2012;95:239–245. [PubMed] [Google Scholar]
  • Matos J, West SC. Holliday junction resolution: Regulation in space andtime. DNA Repair. 2014;19:176–181. [PMC free article] [PubMed] [Google Scholar]
  • Mayer C, Popanda O, Greve B, Fritz E, Illig T, Eckardt-Schupp F, Gomolka M, Benner A, Schmezer P. A radiation-induced gene expression signature as a tool to predictacute radiotherapy-induced adverse side effects. Cancer Lett. 2011;302:20–28. [PubMed] [Google Scholar]
  • Mladenov E, Magin S, Soni A, Iliakis G. DNA double-strand break repair as determinant of cellularradiosensitivity to killing and target in radiation therapy. Front Oncol. 2013;3:1–18. [PMC free article] [PubMed] [Google Scholar]
  • Ochi T, Wu Q, Blundell TL. The spatial organization of non-hom*ologous end joining: From bridgingto end joining. DNA Repair. 2014;17:98–109. [PMC free article] [PubMed] [Google Scholar]
  • Ock C, Kim E, Choi DJ, Lee HJ, Hahm K, Chung MH. 8-Hydroxydeoxyguanosine: Not mere biomarker for oxidative stress, butremedy for oxidative stress-implicated gastrointestinal diseases. World J Gastroenterol. 2012;18:302–308. [PMC free article] [PubMed] [Google Scholar]
  • O'Donovan PA, Livingston DM. BRCA1 and BRCA2: Breast/ovarian cancer susceptibility gene productsand participant ins in double strand break repair. J Carcinog. 2010;31:961–967. [PubMed] [Google Scholar]
  • Pastink A, Eeken JCJ, Lohman PHM. Genomic integrity and the repair of double-strand DNAbreaks. Mutat Res. 2001;481:37–50. [PubMed] [Google Scholar]
  • Patel RR, Arthur DW. The emergence of advanced brachytherapy techniques for commonmalignancies. Hematol Oncol Clin North Am. 2006;20:97–118. [PubMed] [Google Scholar]
  • Podhorecka M, Skladanowski A, Bozko P. H2AX Phosphorylation: Its role in DNA damage response and cancertherapy. J Nucleic Acids. 2011;2010: [PMC free article] [PubMed] [Google Scholar]
  • Redon CE, Nakamura AJ, Zhang Y-W, Ji JJ, Bonner WM, Kinders RJ, Parchment RE, Doroshow JH, Pommier Y. Histone gammaH2AX and poly(ADP-ribose) as clinical pharmacodynamicbiomarkers. Clin Cancer Res. 2010;16:4532–4542. [PMC free article] [PubMed] [Google Scholar]
  • Richard DJ, Cubeddu L, Urquhart AJ, Bain A, Bolderson E, Menon D, White MF, Khanna KK. HSSB1 interacts directly with the MRN complex stimulating itsrecruitment to DNA double-strand breaks and its endo-nucleaseactivity. Nucleic Acids Res. 2011a;39:3643–3651. [PMC free article] [PubMed] [Google Scholar]
  • Richard DJ, Savage K, Bolderson E, Cubeddu L, So S, Ghita M, Chen DJ, White MF, Richard K, Prise KM, et al. HSSB1 rapidly binds at the sites of DNA double-strand breaks and isrequired for the efficient recruitment of the MRN complex. Nucleic Acids Res. 2011b;39:1692–1702. [PMC free article] [PubMed] [Google Scholar]
  • Ripperger T, Gadzicki D, Meindl A, Schlegelberger B. Breast cancer susceptibility: Current knowledge and implications forgenetic counselling. Eur J Med Genet. 2009;17:722–731. [PMC free article] [PubMed] [Google Scholar]
  • Roberts SA, Strande N, Burkhalter MD, Strom C, Havener JM, Hasty P, Ramsden DA. Ku is a 5'dRP/AP lyase that excises nucleotide damage near brokenends. Nature. 2010;464:1214–1217. [PMC free article] [PubMed] [Google Scholar]
  • Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. Double-stranded breaks induce Histone H2AX phosphorylation on Serine139. J Biol Chem. 1998;273:5858–5868. [PubMed] [Google Scholar]
  • Roszkowski K, Olinski R. Urinary 8-oxoguanine as a predictor of survival in patients undergoingradiotherapy. Cancer Epidemiol Biomarkers Prev. 2012;2012:629–635. [PubMed] [Google Scholar]
  • Rothkamm K, Horn S. gamma-H2AX as protein biomarker for radiation exposure. Ann Ist Super Sanita. 2009;45:265–271. [PubMed] [Google Scholar]
  • Roy R, Chun J, Powell SN. BRCA1 and BRCA2: Different roles in a common pathway of genomeprotection. Nat Rev Cancer. 2012;12:68–78. [PMC free article] [PubMed] [Google Scholar]
  • Rübe CE, Grudzenski S, Kühne M, Dong X, Rief N, Löbrich M, Rübe C. DNA double-strand break repair of blood lymphocytes and normal tissuesanalysed in a preclinical mouse model: Implications for radiosensitivitytesting. Clin Cancer Res. 2008;14:6546–6555. [PubMed] [Google Scholar]
  • Salles D, Mencalha AL, Ireno IC, Wiesmüller L, Abdelhay E. BCR-ABL stimulates mutagenic hom*ologous DNA double-strand break repairvia the DNA-end-processing factor CtIP. Carcinogenesis. 2011;32:27–34. [PubMed] [Google Scholar]
  • Shee C, Cox BD, Gu F, Luengas EM, Joshi MC, Chiu L-Y, Magnan D, Halliday JA, Frisch RL, Gibson JL, et al. Engineered proteins detect spontaneous DNA breakage in human andbacterial cells. Genes Chromosomes. 2013;2: [PMC free article] [PubMed] [Google Scholar]
  • Sibanda BL, Chirgadze DY, Blundell TL. Crystal structure of DNA-PKcs reveals a large open-ring cradlecomprised of HEAT repeats. Nature. 2010;463:118–121. [PMC free article] [PubMed] [Google Scholar]
  • Siever OM, Heinimann K, Tomlinson IPM. Genomic instability - The engine of tumorigenesis? Perspectives. 2003;3:1–8. [PubMed] [Google Scholar]
  • Sirota NP, Zhanataev AK, Kuznetsova EA, Khizhnyak EP, Anisina EA, Durnev AD. Some causes of inter-laboratory variation in the results of cometassay. Mutat Res Genet Toxicol Environ Mutagen. 2014;770:16–22. [PubMed] [Google Scholar]
  • Skiöld S, Naslund I, Brehwens K, Andersson A, Wersall P, Lidbrink E, Harms-Ringdahl M, Wojcik A, Haghdoost S. Radiation-induced stress response in peripheral blood of breast cancerpatients differs between patients with severe acute skin reactions and patientswith no side effects to radiotherapy. Mutat Res Genet Toxicol Environ Mutagen. 2013;756:152–157. [PubMed] [Google Scholar]
  • Sleeth KM, Sørensen CS, Issaeva N, Dziegielewski J, Bartek J, Helleday T. RPA mediates recombination repair during replication stress and isdisplaced from DNA by checkpoint signalling in human cells. J Mol Cell Biol. 2007;373:38–47. [PubMed] [Google Scholar]
  • Smirnov DA, Brady L, Halasa K, Morley M, Solomon S, Cheung VG. Genetic variation in radiation-induced cell death. Genome Res. 2012;22:332–339. [PMC free article] [PubMed] [Google Scholar]
  • Somaiah N, Yarnold J, Lagerqvist A, Rothkamm K, Helleday T. hom*ologous recombination mediates cellular resistance and fractionsize sensitivity to radiation therapy. Radiother aOncol. 2013;108:155–161. [PubMed] [Google Scholar]
  • Turesson I, Nyman J, Holmberg E, Oden A. Prognostic factors for acute and late skin reactions in radiotheraphypatients. Int J Radiat Oncol Biol Phys. 1996;36:1065–1075. [PubMed] [Google Scholar]
  • Voduc KD, Cheang MCU, Tyldesley S, Gelmon K, Nielsen TO, Kennecke H. Breast cancer subtypes and the risk of local and regionalrelapse. J Clin Oncol. 2010;28:1684–1691. [PubMed] [Google Scholar]
  • Vral A, Willems P, Claes K, Poppe B, Perletti ABG, Thierens H. Combined effect of polymorphisms in Rad51 and XRCC3 on breast cancerrisk and chromosomal radiosensitivity. Mol Med Rep. 2011;185:901–912. [PubMed] [Google Scholar]
  • Walker JR, Corpina RA, Goldberg J. Structure of the Ku heterodimer bound to DNA and its implications fordouble-strand break repair. Nature. 2001;412:607–614. [PubMed] [Google Scholar]
  • West SC. Molecular views of recombination proteins and theircontrol. Nat Rev. 2003;4:1–11. [PubMed] [Google Scholar]
  • Williams AB, Michael WM. Eviction notice: New insights into Rad51 removal from DNA duringhom*ologous recombination. Mol Cell. 2010;37:157–158. [PubMed] [Google Scholar]
  • Williams GJ, Hammel M, Radhakrishnan SK, Ramsden D, Lees-Miller SP, Tainer JA. Structural insights into NHEJ: Building up an integrated picture ofthe dynamic DSB repair super complex, one component and interaction at atime. DNA Repair. 2014;17:110–120. [PMC free article] [PubMed] [Google Scholar]
  • Youlden DR, Cramb SM, Dunn NAM, Muller JM, Pyke CM, Baade PD. The descriptive epidemiology of female breast cancer: An internationalcomparison of screening, incidence, survival and mortality. Cancer Epidemiol. 2012;36:237–248. [PubMed] [Google Scholar]

Articles from Genetics and Molecular Biology are provided here courtesy of Sociedade Brasileira de Genética

Ionizing radiation-induced DNA injury and damage detection in patients
with breast cancer (2024)

FAQs

How does ionizing radiation cause breast cancer? ›

The researchers found that when levels of a tumor suppressor protein called phosphatase and tensin hom*olog (PTEN) were low in a type of breast tissue called stroma, it increased the likelihood that exposure to radiation would trigger tumor growth.

What type of DNA damage does ionizing radiation cause? ›

Ionizing radiation directly affects DNA structure by inducing DNA breaks, particularly, DSBs. Secondary effects are the generation of reactive oxygen species (ROS) that oxidize proteins and lipids, and also induce several damages to DNA, like generation of abasic sites and single strand breaks (SSB).

What does ionizing radiation do to cancer? ›

Ionising radiation, such as gamma rays, X-rays and radioactive particles can cause cancer by damaging DNA. However, how this happens, or how many tumours are caused by radiation damage has not been known.

What is the effect of ionizing radiation on DNA quizlet? ›

Interactions of ionizing radiation with DNA molecules may cause the loss of or change in a nitrogenous base in the DNA chain. Direct consequence of this damage is an alteration of the base sequence, a mutation.

How sick does radiation for breast cancer make you? ›

Common side effects during treatment may include: Mild to moderate fatigue. Skin irritation, such as itchiness, redness, peeling or blistering, similar to what you might experience with a sunburn. Breast swelling.

What is the absolute risk for radiation-induced breast cancer? ›

On average, the radiation-induced excess relative risk and excess absolute risk of breast cancer at age 50 years were 0.51 Gy1 (95% confidence interval (CI): 0.33, 0.71) and 10.8 cases/10,000 person-years/Gy (95% CI: 7.0, 14.6), respectively.

What are 3 effects of ionizing radiation? ›

High doses of ionizing radiation can lead to various effects, such as skin burns, hair loss, birth defects, illness, cancer, and death. The basic principle of toxicology, “the dose determines poison,” applies to the toxicology of ionizing radiation as well as to all other branches of toxicology.

Which ionizing radiation has the highest risk of cancer? ›

High-energy radiation, such as x-rays, gamma rays, alpha particles, beta particles, and neutrons, can damage DNA and cause cancer. These forms of radiation can be released in accidents at nuclear power plants and when atomic weapons are made, tested, or used.

Does ionizing radiation stay in the body? ›

Does any radiation stay in the body after an imaging exam? After a radiographic, fluoroscopic, CT, ultrasound, or MRI exam, no radiation remains in your body. For nuclear medicine imaging, a small amount of radiation can stay in the body for a short time.

What can radiation do to your DNA? ›

Ionizing radiation damages the genetic material in reproductive cells and results in mutations that are transmitted from generation to generation.

What type of DNA damage may not be reversible? ›

In contrast to DNA damage, a mutation is a change in the base sequence of the DNA. A mutation cannot be recognized by enzymes once the base change is present in both DNA strands, and thus a mutation cannot be repaired. At the cellular level, mutations can cause alterations in protein function and regulation.

Which of the following are effects of ionizing radiation on cells? ›

When ionizing radiation interacts with cells, it can cause damage to the cells and genetic material (i.e., deoxyribonucleic acid, or DNA). If not properly repaired, this damage can result in the death of the cell or potentially harmful changes in the DNA (i.e., mutations).

What does radiation do to the breast? ›

The main short-term side effects of external beam radiation therapy to the breast are: Swelling in the breast. Skin changes in the treated area similar to a sunburn (redness, skin peeling, darkening of the skin) Fatigue.

How does radiation affect breast implants? ›

Radiation therapy may cause the reconstructed breast to become much firmer and to sit higher on the chest. When the radiated tissue around an implant, called a capsule, becomes firm, this is known as capsular contracture.

Can electromagnetic radiation cause breast cancer? ›

Large prospective cohort studies and a meta-analysis that combined the results from 15 studies have found no link between EMF and breast cancer. Learn more about EMF and breast cancer risk. Learn about the strengths and weaknesses of different types of studies.

Can laptop radiation cause breast cancer? ›

The risk for breast cancer was similar in the highest quartile of EMF-exposure from computer use (relative risk 1.05, 95% confidence interval (CI) 0.95 – 1.16) compared to the lowest quartile, after adjustment for numerous established risk factors, for all states combined.

Top Articles
ZOO Crypto World price today, ZOO to USD live price, marketcap and chart | CoinMarketCap
Can You Pawn 10k Gold? | Meriden Pawn
Public Opinion Obituaries Chambersburg Pa
Stadium Seats Near Me
Don Wallence Auto Sales Vehicles
Roblox Developers’ Journal
How do you mix essential oils with carrier oils?
Doby's Funeral Home Obituaries
You can put a price tag on the value of a personal finance education: $100,000
Phillies Espn Schedule
2021 Lexus IS for sale - Richardson, TX - craigslist
Oc Craiglsit
Hca Florida Middleburg Emergency Reviews
Aldi Sign In Careers
Dr Adj Redist Cadv Prin Amex Charge
Vandymania Com Forums
Jang Urdu Today
Google Doodle Baseball 76
Ratchet & Clank Future: Tools of Destruction
Leccion 4 Lesson Test
라이키 유출
Scout Shop Massapequa
Catherine Christiane Cruz
Reptile Expo Fayetteville Nc
Raz-Plus Literacy Essentials for PreK-6
Bjerrum difference plots - Big Chemical Encyclopedia
[PDF] NAVY RESERVE PERSONNEL MANUAL - Free Download PDF
Craigslist Battle Ground Washington
Sand Dollar Restaurant Anna Maria Island
Webworx Call Management
Revelry Room Seattle
Abga Gestation Calculator
Ofw Pinoy Channel Su
Dreamcargiveaways
Metra Union Pacific West Schedule
Mgm Virtual Roster Login
SF bay area cars & trucks "chevrolet 50" - craigslist
The Thing About ‘Dateline’
Mars Petcare 2037 American Italian Way Columbia Sc
Indiana Jones 5 Showtimes Near Cinemark Stroud Mall And Xd
Metro Pcs Forest City Iowa
Obituaries in Hagerstown, MD | The Herald-Mail
Cnp Tx Venmo
Who Is Responsible for Writing Obituaries After Death? | Pottstown Funeral Home & Crematory
No Boundaries Pants For Men
Nina Flowers
Breaking down the Stafford trade
Mcoc Black Panther
Ronnie Mcnu*t Uncensored
Myapps Tesla Ultipro Sign In
Gummy Bear Hoco Proposal
Latest Posts
Article information

Author: Geoffrey Lueilwitz

Last Updated:

Views: 5757

Rating: 5 / 5 (80 voted)

Reviews: 95% of readers found this page helpful

Author information

Name: Geoffrey Lueilwitz

Birthday: 1997-03-23

Address: 74183 Thomas Course, Port Micheal, OK 55446-1529

Phone: +13408645881558

Job: Global Representative

Hobby: Sailing, Vehicle restoration, Rowing, Ghost hunting, Scrapbooking, Rugby, Board sports

Introduction: My name is Geoffrey Lueilwitz, I am a zealous, encouraging, sparkling, enchanting, graceful, faithful, nice person who loves writing and wants to share my knowledge and understanding with you.