You are on page 1of 16

International Journal of Radiation Biology

ISSN: 0955-3002 (Print) 1362-3095 (Online) Journal homepage: https://www.tandfonline.com/loi/irab20

Gene expression following ionising radiation:


Identification of biomarkers for dose estimation
and prediction of individual response

Sylwia Kabacik, Alan Mackay, Narinder Tamber, Grainne Manning, Paul


Finnon, Francois Paillier, Alan Ashworth, Simon Bouffler & Christophe Badie

To cite this article: Sylwia Kabacik, Alan Mackay, Narinder Tamber, Grainne Manning, Paul
Finnon, Francois Paillier, Alan Ashworth, Simon Bouffler & Christophe Badie (2011) Gene
expression following ionising radiation: Identification of biomarkers for dose estimation and
prediction of individual response, International Journal of Radiation Biology, 87:2, 115-129, DOI:
10.3109/09553002.2010.519424

To link to this article: https://doi.org/10.3109/09553002.2010.519424

Published online: 10 Nov 2010.

Submit your article to this journal

Article views: 598

View related articles

Citing articles: 84 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=irab20
Int. J. Radiat. Biol., Vol. 87, No. 2, February 2011, pp. 115–129

Gene expression following ionising radiation: Identification


of biomarkers for dose estimation and prediction of individual
response

SYLWIA KABACIK1, ALAN MACKAY2, NARINDER TAMBER2, GRAINNE MANNING1,


PAUL FINNON1, FRANCOIS PAILLIER1, ALAN ASHWORTH2, SIMON BOUFFLER1, &
CHRISTOPHE BADIE1
1
Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Health Protection Agency,
Chilton, Didcot, Oxfordshire, and 2Breakthrough Breast Cancer Research Centre, the Institute of Cancer Research,
London, UK

(Received 18 June 2010; Revised 3 August 2010; Accepted 25 August 2010)

Abstract
Purpose: To establish a panel of highly radiation responsive genes suitable for biological dosimetry and to explore inter-
individual variation in response to ionising radiation exposure.
Materials and methods: Analysis of gene expression in response to radiation was carried out using three independent
techniques (Microarray, Multiplex Quantitative Real-Time Polymerase Chain Reaction (MQRT- PCR) and nCounter1
Analysis System) in human dividing lymphocytes in culture and peripheral blood leukocytes exposed ex vivo from the same
donors.
Results: Variations in transcriptional response to exposure to ionising radiation analysed by microarray allowed the
identification of genes which can be measured accurately using MQRT PCR and another technique allowing direct count of
mRNA copies. We have identified genes which are consistently up-regulated following exposure to 2 or 4 Gy of X-rays at
different time points, for all individuals in blood and cultured lymphocytes. Down-regulated genes including cyclins,
centromeric and mitotic checkpoint genes, particularly those associated with chromosome instability and cancer could be
detected in dividing lymphocytes only.
Conclusions: The data provide evidence that there are a number of genes which seem suitable for biological dosimetry using
peripheral blood, including sestrin 1 (SESN1), growth arrest and DNA damage inducible 45 alpha (GADD45A), cyclin-
dependent kinase inhibitor 1A (CDKN1A), cyclin G1 (CCNG1), ferredoxin reductase (FDXR), p53 up-regulated mediator
of apoptosis (BBC3) and Mdm2 p53 binding protein homolog (MDM2). These biomarkers could potentially be used for
triage after large-scale radiological incidents and for monitoring radiation exposure during radiotherapy.

Keywords: ionising radiation, biodosimetry, individual response, gene expression, biomarkers, peripheral blood lymphocytes

exposed individuals followed by more accurate


Introduction
assessment of doses in those exposed to determine
Ionising radiation (IR) is a ubiquitous environmental the most appropriate approach to treatment. As in
hazard and a well-known carcinogen. In general, cases of accidental exposure, physical dosimetry is
exposures are low but accidents and incidents have rarely available, and biological dosimetry, based on
led to significant exposures (Blakely et al. 2005). the scoring of chromosomal damage – specifically
The response to any radiation accident or incident dicentric chromosomes – in blood samples, plays a
involving actual or potential IR exposure requires key role (International Atomic Energy Agency
accurate and rapid assessment of the doses received [IAEA] 2001). The dicentric assay is accurate and
by individuals. This is required for initial ‘triage’ of sensitive; nonetheless, it is a labour-intensive assay

Correspondence: Dr Christophe Badie, Cancer Genetics and Cytogenetics, Radiation Effects Department, Health Protection Agency, Centre for Radiation,
Chemical and Environmental Hazards, Chilton, Didcot, Oxon, OX11 0RQ, UK. Tel: þ44 (0)1235 822698. Fax: þ44 (0)1235 833891.
E-mail: christophe.badie@hpa.org.uk
ISSN 0955-3002 print/ISSN 1362-3095 online Ó 2011 Informa UK, Ltd.
DOI: 10.3109/09553002.2010.519424
116 S. Kabacik et al.

(42 days from sampling for first results), and it has a biomarkers of human exposure to radiation after an
limited sample capacity available for mass screening accident (Dressman et al. 2007, Chaudhry 2008),
following an accident or incident. A recent estimation and selected molecular biomarkers are likely to
including the 15 European Union countries where provide a more robust approach for high-throughput
biological dosimetry is established, gives a total minimally invasive radiation biodosimetry (Paul and
capacity for dosimetric triage of about 1500 cases per Amundson 2008).
week analysed with a dicentric assay (Wojcik et al. Gene expression responses in blood lymphocytes
2010). Therefore, it is clear that any minimally can also be used to understand the genetic basis for
invasive methods that could provide individual dose variation in human radiosensitivity (Correa and
estimates more rapidly with greater sample through- Cheung 2004), to characterise tumour subtypes
put would be of great value in incident management. (e.g., Payton et al. 2009), to assess inter-individual
Recently, many attempts have been made to susceptibility to DNA damaging agents for the
improve dosimetry using different end-points. Dif- prediction of therapeutic response to drugs (Fry
ferent cytogenetic techniques have been proposed, for et al. 2008), to predict clinical outcome in human
example, using premature chromosome condensa- cancers (Carter et al. 2006) and to identify in vivo
tion (Lindholm et al. 2010) or analysis of micronuclei radiation-induced biomarkers (Amundson et al.
(Willems et al. 2010). Other techniques have shown 2004). For example, we recently have shown (Badie
some potential, and studies on protein biomarkers et al. 2008) that cyclin-dependent kinase inhibitor
such as histone phosphorylation (g-H2AX) (Garty 1A (CDKN1A) transcriptional response associates
et al. 2010), global proteomics approaches (Ossetrova with abnormal sensitivity to radiation treatment.
and Blakely 2009) and changes in gene expression Several studies have identified potential biomar-
following IR exposure (Amundson et al. 1999, Fan kers of radiation exposure. But there has been
et al. 2001) are promising. These studies are based on relatively little effort placed on the identification of
the knowledge that exposure to IR induces complex genes that respond consistently in cycling and non-
changes at the level of RNA transcripts (quantity) cycling cells from all donors.
as well as proteins (stability, post-transcriptional In this study, we compared the gene expression
changes and quantity). response to IR in dividing, mitogen stimulated
Over recent years, a greater understanding has lymphocytes and blood resting leukocytes exposed
been obtained of the transcriptional response in cells ex vivo from three healthy normal donors. The goals
to radiation exposure. Exposure of cells to ionising of this study were to establish a panel of highly
radiation activates multiple signal transduction path- radiation responsive genes suitable for radiation
ways, which result in complex alterations in gene dosimetry, to compare the gene expression profiles
expression. Expression of specific genes can depend of cycling cells (stimulated T-lymphocytes) and non
on radiation dose (Grace et al. 2002, Amundson and dividing gap 0 (Go) blood leukocytes from the same
Fornace 2003, Stassen et al. 2003), dose-rate donors and to explore inter-individual variation in
(Amundson et al. 2003), radiation quality response to IR and to compare three methods for
(Woloschak and Chang-Liu 1995) and lapse between quantifying gene expression.
stress and analysis (Franco et al. 2005), making
gene-expression profiling a potentially powerful and
informative approach for biological dosimetry Materials and methods
(Blakely et al. 2003, Chaudhry 2008). A linear
Blood collection
dose-response relationship has been obtained for
some genes over a dose range of 1–3 Gy (Grace et al. Peripheral blood samples were obtained anon-
2002), 0.5–4 Gy (Kang et al. 2003) and down to ymously from three healthy female working indivi-
doses as low as 2 cGy (Amundson and Fornace duals (age range 37–53 years old) with informed
2003). Such studies have utilised human lympho- consent and ethical approval from Oxford Research
blastoid cells (Jen and Cheung 2003), non-cycling Ethics Committee. Lymphocyte samples were pur-
human fibroblasts (Ding et al. 2005), confluent ified by separation on Histopaque-1077 (Sigma-
human keratinocytes (Franco et al. 2005) and Aldrich, Poole, UK) and frozen in liquid nitrogen
peripheral blood lymphocytes (Kang et al. 2003). prior to use. These lymphocytes were used to
The transcriptional responses to the genotoxic produce T-cell lines.
stresses can be monitored on a global scale using
microarrays (Tusher et al. 2001, Gruel et al. 2006) or
Cell growth
more accurately and sensitively in a few specific
genes by quantitative real time polymerase chain T-lymphocyte cultures were prepared using the
reaction (QRT-PCR) (Grace et al. 2003). Radiation method described previously (O’Donovan et al.
responsive genes could potentially be used as 1995, Finnon et al. 2008).
Gene expression response to ionising radiation 117

Expression values from spots with hybridisation


Irradiation of lymphocyte cultures and blood
artefacts or extremely low intensities were flagged in
Cultured lymphocytes were disaggregated and Genepix 5.1 (Axon Instruments/Molecular Devices
seeded at a density of 4 6 105 cells/ml in GR10 Corporation Sunnyvcale, CA, USA) and then con-
medium. Cells or fresh whole blood were irradiated verted to missing values. The raw intensity values
at 2 or 4 Gy using a Siemens Stabilipan (Siemens were then converted to log2 ratios of sample to
AG, Munich, Germany) therapy X-ray set (output reference (M values) and log2 average spot intensity
14 mA, 250 kV peak, 1.2 mm Cu HVL, 0.7 Gy (A values) for all subsequent pre-processing and
min71) at room temperature at the Medical Re- analysis.
search Council (MRC), Harwell. Fresh blood and Data from lymphocyte samples and blood samples
lymphocyte cultures were maintained at 378C before were pre-processed separately. Probes with missing
and after irradiation. RNA Later (Applied Biosys- values in more than 80% of hybridisations within
tems/Ambion, Warrington WA3 7QH, UK) was each cell type were removed from subsequent
added 2 or 24 h after irradiation and cells were kept analysis. The remainder of missing values were
at 7808C until RNAs were extracted. imputed using a k-nearest neighbour imputation.
The loess local regression function was used to
remove dye biases in log2 ratios across the slide.
RNA purification from lymphocytes and whole
After loess normalisation log2 ratios were subjected
blood samples
to quantile normalisation using the affy package in R.
Total RNA from human blood leukocytes (white Final processed data sets represented 10736 probes
blood cells) was prepared by using LeukoLOCKTM for the analysis of lymphocytes and 9698 probes for
Total RNA Isolation Kit (Applied Biosystems/ the analysis of blood samples.
Ambion, Warrington, Cheshire, UK) (for MQRT- Differential gene expression was identified using
PCR and microarrays). This kit eliminates plasma, the significance analysis of microarrays (SAM) in the
platelets and red blood cells including reticulocytes. same package. Paired 2-class SAM comparisons
RiboPureTM-Blood Kit (Applied Biosystems/Am- were made between irradiated samples and respec-
bion) was used to extract RNA for Nanostring. Total tive controls in each tissue, at each time point and
RNA from human T-cell lines was prepared by using each dose. Individual comparisons used an overall
RNAqueous1-4PCR (Applied Biosystems/Ambion) false discovery rate of 5% as a cut-off and were
kit. DNA contamination was removed by using ranked according to their fold change. A complete
DNA-freeTM (Applied Biosystems/Ambion). RNA list of differentially expressed genes was made by
quantity was assessed by Nanodrop ND1000, and concatenating the results of these tests and ranking
RNA quality was assessed on 1.3% agarose gel. For the combined gene lists based upon the sum of their
multiplex PCR, reverse transcriptase reactions were individual ranks in each test.
performed using High Capacity cDNA Reverse
transcription kit, (Applied Biosystems, Foster City,
Multiplex quantitative Real-Time-PCR
CA, USA) according to manufacturer’s protocol
(MQ-RT- PCR)
with 700 ng of total RNA.
Real-time PCR was performed using iQ5 thermo-
cyclers (Bio-Rad, Hercules, CA, USA). All reactions
cDNA microarray hybridisation procedure and
were run in triplicate using PerfeCTa1 MultiPlex
data extraction
qPCR SuperMix (Quanta Biosciences, Inc.
Total RNA integrity was confirmed on an Agilent Gaithersburg, MD, USA), primer and probe sets
2100 BioAnalyser (Agilent Technologies, South for target genes at 300 nM concentration each and
Queensferry, West Lothian, UK) before linear T7 2.5 ml of cDNA in 30 ml reaction volume. 30 6-
amplification using an aminoallyl MessageAmp Kit Carboxyfluorescein (FAM), 6- Hexachlorofluores-
(Applied Biosystems/Ambion). Amplified RNA cein (HEX), Texas Red and CY5 (Eurogentec Ltd,
(1 mg) was coupled with either CY3 or CY5 and Fawley, Hampshire, UK) were used as fluorochrome
hybridised to Breakthrough 20K cDNA microarrays reporters for the hydrolysis probes analysed in
in replicate dye swap hybridisations, as previously multiplexed reactions. A list of the primers and
described (Mackay et al. 2009). The Breakthrough probes which we designed is given in Table I.
20K microarray platform and all primary microarray Cycling parameters were 2 min at 958C, then 45
data have been submitted to Array Express database cycles of 10 s at 958C and 60 s at 608C. Data were
(http://www.ebi.ac.uk/microarray-as/ae/, with acces- collected and analysed by iQ5 Detection System
sion number E-TABM-1083). Annotation of the software. Gene target Ct (cycle threshold) values
Breakthrough 20K cDNA microarray was based on were normalised to a Hypoxanthine-Guanine phos-
build 216 of the Unigene database. phoribosyltransferase 1 (HPRT1) internal control.
118 S. Kabacik et al.

Table I. Oligonucleotide primers and probes used in this study for MQRT-PCR analysis. They were designed by using in house programs
specifically developed to identify multiplex PCR compatible oligonucleotides.

Database acc no.

Genes Genebank/dbSNP PCR primers, Fwd, Rev Probes

HPRT1 NM_000194.2 TCAGGCAGTATAATCCAAAGATGGT CGCAAGCTTGCTGGTGAAAAGGACCC


AGTCTGGCTTATATCCAACACTTCG
CCNG1 NM_004060.3, GGAGCTGCAGTCTCTGTCAAG AACTGCTACACCAGCTGAATGCCC
NM_199246.1
TGACATCTAGACTCCTGTTCCAA
PLK3 NM_004073.2 ATCAGCGCGAGAAGATCCTA CGAGACCTGCAGCACCGCC
GATGTTGTCAGCGTCCTCAA
CDC25C NM_022809, ATGCATCATCAGGACCACAA TGAGTTGCTGAGGTGTCGAAGCCA
NM_001790
GCCAGTGGCTGGAATGTTA
PRC1 NM_199414, AATCTGATGCTACTTCTGGAATC CAATTCAACCAACATCCAGTCCTGAGAA
NM_199413,
NM_003981
AGCCACAGCTGGTTGACTGA
SESN1 NM_014454 GCTGTCTTGTGCATTACTTGTG ACATGTCCCACAACTTTGGTGCTGG
CTGCGCAGCAGTCTACAG
FDXR NM_024417, GTACAACGGGCTTCCTGAGA CGGGCCACGTCCAGAGCCA
NM_004110
CTCAGGTGGGGTCAGTAGGA
ATF3 NM_001030287, AGGTTTGCCATCCAGAACAA CCTCTGCCACCGGATGTCCTCT
NM_001040619,
NM_001674
CTGACAGTGACTGATTCC
MDM2 NM_002392 CCATGATCTACAGGAACTTGGTAGTA CAATCAGCAGGAATCATCGGACTCAG
ACACCTGTTCTCACTCACAGATG
DDB2 NM_000107 GTCACTTCCAGCACCTCACA AGCCTGGCATCCTCGCTACAACC
ACGTCGATCGTCCTCAATTC
BBC3 NM_014417.3, CGGAGACAAGAGGAGCAG CCCTCACCCTGGAGGGTCCTGT
NM_001127240.1
NM_001127241.1
NM_001127242.1
GGAGTCCCATGATGAGATTG
CDKN1A NM_000389.3, GCAGACCAGCATGACAG TTTCTACCACTCCAAACGCCGGCT
NM_078467.1
TAGGGCTTCCTCTTGGA
GADD45A NM_001924.2 CTGCGAGAACGACATCAAC ATCCTGCGCGTCAGCAACCCG
AGCGTCGGTCTCCAAGAG
PHPT1 NM_001135861.1, TCGCTCTCATTCCTGATGTG CTTGTAGCCGCGCACGATCTCCTT
NM_014172.4
TCGTAGATGTCCGCATGGTA

Ct values were converted to transcript quantity using The technology uses molecular barcodes and single
standard curves obtained by serial dilution of PCR- molecule imaging to detect and count hundreds of
amplified DNA fragments of each gene. The linear unique transcripts in a single reaction. The nCounter
dynamic range of the standard curves covering six Reporter Probes allow for direct measurement of
orders of magnitude (serial dilution from 3.2 6 1074 individual RNA molecules and provides a digital
to 8.2 6 10710) gave PCR efficiencies between 93% signal.
and 105% for each gene with R2 4 0.998. Relative Briefly, target molecules are detected by hybridisa-
gene expression levels after irradiation were similarly tion to Capture and Reporter Probes, each approxi-
obtained for comparison with pre-irradiation controls. mately 50 nucleotides in length and targetting a
contiguous 100-base target region. Target regions of
each mRNA are screened to eliminate direct and
Nanostring method
inverted repeat elements and evaluated for cross-
The nCounter Analysis System uses a novel digital hybridisation against the human RefSeq database.
technology (NanoString Technologies1, Inc., Seat- Potential 50-base probes are then selected for
tle, WA, USA) that is based on direct multiplexed melting temperatures (Tm) between 78–838C. Cap-
measurement of gene expression (Geiss et al. 2008). ture and Reporter Probes are ligated to the synthetic
Gene expression response to ionising radiation 119

DNA backbones containing the barcode as described The list of the top 20 genes up and down-regulated
(Geiss et al. 2008), and supplied in one tube in cultured lymphocytes and blood in response to
containing all Capture probes, and one tube contain- irradiation is presented in Table III, and the
ing all Reporter probes. corresponding heatmaps of microarray data are
Hybridisations are set up in four pipetting steps as shown in Figure 1.
follows: For each sample, 100 ng of purified RNA or The number of genes seems to be relatively similar
lysate in 5 ml total is added, 10 ml hybridisation buffer, at 2 and 24 h with more up-regulated genes in blood
10 ml supplied Reporter Probe mixture, and lastly 5 ml whereas down-regulated genes are mostly observed
of supplied Capture Probe mixture in separate thin- in cultured lymphocytes. Some genes are specifically
walled PCR strip tubes. The tubes are then covered up-regulated in cultured lymphocytes Table IIIA or
and incubated at 658C for 12–18 h in a thermocycler Blood Table IIIC; however, among the genes
with heated lid. After hybridisation, the samples are significantly up-regulated in blood and cultured
processed in the PrepStation and counted in the lymphocytes, nine genes are consistently found in
DigitalAnalyzer. The number of counts of each gene blood and lymphocytes in the top 20 list for at least
in the CodeSet is analysed by Microsoft Excel one dose at one time point in all three repeat
software. Nanostring assays were performed by experiments, (Table III): sestrin 1 (SESN1), growth
Nanostring Technologies1, Inc., Seattle. arrest and DNA damage inducible 45 alpha (GAD-
D45A), cyclin G1 (CCNG1), RNA binding motif
protein 15 (RBM15), CDKN1A, TNF receptor
Results
superfamily, member 6 (FAS), F-box and WD repeat
Three independent experiments have been carried domain containing two (FBXW2), protein phospha-
out on the blood leukocyte population as well as tise, Mg2þ/Mn2þ dependent, 1D (PPM1D) and
cultured T-lymphocytes from the same three donors. biogenesis of lyosomal organelles complex-1, subunit
Blood and T-lymphocytes were irradiated using 2 (BLOC1S2), with the first four being present at all
three doses (0, 2 and 4 Gy of X-rays) and two time time point for all doses.
points (2 and 24 h post-irradiation). Blood leuko- Other genes are specifically very good markers for
cytes trapped on the filters (LeukoLOCKTM) and blood such as PCNA (proliferating-cell nuclear
T-lymphocytes were kept 7808C thus allowing RNA antigen) which is involved in DNA replication, repair
from all samples to be extracted and analysed of DNA damage, chromatin structure maintenance,
simultaneously. Freezing the cells does not interfere chromosome segregation and cell-cycle progression
with the gene expression results (data not shown) (Stoimenov and Helleday 2009) or for dividing
and allows retrospective analysis of large batches of lymphocytes such as ATF3 (Activating transcription
samples. factor 3), a transcription factor that plays a regulatory
Altogether 108 samples have been analysed by role in inflammation, cell division apoptosis and
DNA array (216 arrays) and MQRT-PCR. Micro- DNA damage (Fan et al. 2002). Both are signifi-
array analyses identified the genes significantly up cantly up-regulated at all doses and time points in all
and down-regulated in response to irradiation in the donors as shown on the heatmaps presented in
three experiments in blood and lymphocytes. The Figure 1.
number of significantly responsive genes for each There are a large number of genes down-regulated
time and dose are presented in Table II. Overall, in irradiated dividing lymphocytes (Tables II and
combining the data, we detected 570 genes up- IIIB) unlike blood leukocytes where there are very
regulated and 31 probes down-regulated in blood few genes consistently and significantly down-regu-
samples as compared with 232 probes up-regulated lated (Table II and Table IIID), especially, at the
and 1357 probes down-regulated in lymphocyte earlier time point (2 h). This is not surprising as
cultures using a false discovery rate of 55%. these down-regulated genes are mainly associated
with cell cycle and are only responsive in cycling
Table II. Radiation responsive genes (up and down-regulated) in lymphocytes in culture (as budding uninhibited by
blood and cultured lymphocytes in response to radiation. Genes benzimidazoles 1 homolog [BUB1], cyclin A2
were identified in paired SAM analyses using a false discovery rate [CCNA2], kinesin family member 23 [KIF23],
of 5%. protein regulator of cytokinesis 1 [PRC1] or cen-
Blood Lymphocytes tromere protein E, 312 kDa [CENPE]).
MQRT-PCR assays have been used to confirm the
2h 24 h 2h 24 h
results obtained by microarray analysis in individual
2 Gy 4 Gy 2 Gy 4 Gy 2 Gy 4 Gy 2 Gy 4 Gy donors for three up-regulated genes (SESN1, GAD-
D45A and CDKN1A) as well as one down-regulated
up 296 234 13 247 31 100 107 116
down 0 1 7 29 29 267 113 1109
gene (PRC1) (Figure 2). Overall, all the genes
identified by microarray were confirmed by the other
120 S. Kabacik et al.

Table III. Top 20 most up-regulated genes in cultured lymphocytes (A) and peripheral whole blood (C) and most down-regulated in
cultured lymphocytes (B) and peripheral whole blood (D) in response to irradiation. The numbers indicate the ranking of genes based on
fold change (1 being the most responsive gene at a particular dose and time-point). False discovery rate of less than 5%. Some of the genes
did not pass this level of significance for a specific dose or time point (NS: not significant).

Symbol Description Unigene entry 2 h 2 Gy 2 h 4 Gy 24 h 2 Gy 24 h 4 Gy

(A) Most significantly up-regulated genes in irradiated cultured lymphocytes.


GADD45A Growth arrest and DNA-damage-inducible, alpha Hs.80409 1 1 1 1
SESN1 Sestrin 1 Hs.591336 3 5 3 3
CDKN1A Cyclin-dependent kinase inhibitor 1A (p21, Cip1) Hs.370771 2 3 7 5
ATF3 Activating transcription factor 3 Hs.460 5 6 13 4
FAS Fas (TNF receptor superfamily, member 6) Hs.244139 6 8 4 11
FBXW2 F-box and WD repeat domain containing 2 Hs.494985 7 14 6 6
RBM15 RNA binding motif protein 15 Hs.435947 4 4 42 8
ANKRA2 Ankyrin repeat, family A (RFXANK-like), 2 Hs.239154 23 33 31 9
PPM1D Protein phosphatase 1D magnesium-dependent, Hs.591184 11 20 60 35
delta isoform
C12orf5 Chromosome 12 open reading frame 5 Hs.504545 12 32 48 43
RRM2B Ribonucleotide reductase M2 B (TP53 inducible) Hs.512592 22 54 45 16
C11orf24 Chromosome 11 open reading frame 24 Hs.303025 19 47 85 29
IFNG Interferon, gamma Hs.856 NS 9 2 2
FBXO22 F-box protein 22 Hs.591115 28 69 94 80
CCNG1 Cyclin G1 Hs.79101 10 23 10 NS
BLOC1S2 Biogenesis of lysosome-related organelles Hs.576605 NS 15 12 18
complex-1, subunit 2
PRDM1 PR domain containing 1, with ZNF domain Hs.436023 NS 19 21 15
TncRNA Trophoblast-derived non-coding RNA Hs.523789 14 39 NS 7
RPS27L Ribosomal protein S27-like Hs.108957 NS 44 5 20
LRRC58 Leucine rich repeat containing 58 Hs.518084 NS 36 23 14
(B) Most significantly down-regulated genes in irradiated cultured lymphocytes.
BUB1 BUB1 budding uninhibited by benzimidazoles Hs.469649 8 19 NS 242
1 homolog (yeast)
CCNA2 Cyclin A2 Hs.58974 22 22 NS 474
KIF23 Kinesin family member 23 Hs.270845 6 3 NS 686
NCAPG Non-SMC condensin I complex, subunit G Hs.567567 20 79 NS 720
PPP1R3D Protein phosphatase 1, regulatory (inhibitor) Hs.42215 26 34 NS 771
subunit 3D
PRC1 Protein regulator of cytokinesis 1 Hs.567385 17 26 NS 826
CENPE Centromere protein E, 312kDa Hs.75573 5 11 NS 913
LDHA Lactate dehydrogenase A Hs.2795 NS NS 1 1
HMMR Hyaluronan-mediated motility receptor Hs.72550 1 2 NS NS
(RHAMM)
DKFZp762E1312 Hypothetical protein DKFZp762E1312 Hs.532968 2 1 NS NS
DLG7 Discs, large homolog 7 (Drosophila) Hs.77695 3 5 NS NS
CBLN2 Cerebellin 2 precursor Hs.569851 NS NS 8 2
PTGES3 Prostaglandin E synthase 3 (cytosolic) Hs.50425 NS NS 6 5
INSIG1 Insulin induced gene 1 Hs.520819 NS NS 3 8
BNIP3 BCL2/adenovirus E1B 19kDa Hs.144873 NS NS 2 9
interacting protein 3
PIF1 PIF1 50 -to-30 DNA helicase homolog Hs.112160 4 7 NS NS
(S. cerevisiae)
PTMA Prothymosin, alpha (gene sequence 28) Hs.459927 NS NS 13 4
PSRC1 Proline/serine-rich coiled-coil 1 Hs.405925 9 8 NS NS
RRM2 Ribonucleotide reductase M2 polypeptide Hs.226390 NS NS 7 11
KIF14 Kinesin family member 14 Hs.3104 7 18 NS NS
(C) Most significantly up-regulated genes in irradiated whole blood.
PCNA Proliferating cell nuclear antigen Hs.147433 1 1 1 1
DUSP21 Dual specificity phosphatase 21 Hs.534478 5 6 11 7
SESN1 Sestrin 1 Hs.591336 6 5 22 11
GADD45A Growth arrest and DNA-damage-inducible, alpha Hs.80409 2 3 38 15
TMEM30A Transmembrane protein 30A Hs.108530 4 4 40 18
CCNG1 Cyclin G1 Hs.79101 3 2 62 26
RBM15 RNA binding motif protein 15 Hs.435947 108 13 13 10
RPS27L Ribosomal protein S27-like Hs.108957 7 NS 3 4

(continued)
Gene expression response to ionising radiation 121

Table III. (Continued).

Symbol Description Unigene entry 2 h 2 Gy 2 h 4 Gy 24 h 2 Gy 24 h 4 Gy

CDKN1A Cyclin-dependent kinase inhibitor 1A (p21, Cip1) Hs.370771 39 NS 6 3


IER5 Immediate early response 5 Hs.15725 50 NS 12 14
FAS Fas (TNF receptor superfamily, member 6) Hs.244139 32 NS 23 24
HSP90AA1 Heat shock protein 90kDa alpha (cytosolic), Hs.525600 73 NS 17 12
class A member 1
FBXW2 F-box and WD repeat domain containing 2 Hs.494985 90 NS 49 30
ARL6IP1 ADP-ribosylation factor-like 6 interacting protein 1 Hs.634882 104 NS 64 105
PERP PERP, TP53 apoptosis effector Hs.520421 56 NS 105 119
UBC Ubiquitin C Hs.520348 86 NS 134 99
BLOC1S2 Biogenesis of lysosome-related organelles Hs.576605 64 NS 169 90
complex-1, subunit 2
ZFR Zinc finger RNA binding protein Hs.435231 40 NS 179 156
PPM1D Protein phosphatase 1D magnesium-dependent, Hs.591184 48 NS 185 153
delta isoform
STX11 Syntaxin 11 Hs.118958 215 NS 72 150
(D) Most significantly down-regulated genes in irradiated whole blood.
BANK1 B-cell scaffold protein with ankyrin repeats 1 Hs.480400 NS NS 1 2
IGJ Immunoglobulin J polypeptide Hs.651109 NS NS 2 4
IL2RB Interleukin 2 receptor, beta Hs.474787 NS NS 3 3
EDG1 Endothelial differentiation, sphingolipid Hs.154210 NS NS 5 13
G-protein-coupled receptor, 1
GLCCI1 Glucocorticoid induced transcript 1 Hs.131673 NS NS 7 24
CD8A CD8a molecule Hs.85258 NS NS 6 26
KLRF1 Killer cell lectin-like receptor subfamily F, member 1 Hs.183125 NS NS NS 1
TCL1A T-cell leukemia/lymphoma 1A Hs.2484 NS 1 NS NS
GZMK Granzyme K (granzyme 3; tryptase II) Hs.277937 NS NS 4 NS
SLC2A3 Solute carrier family 2, member 3 Hs.419240 NS NS NS 5
TRA@ T cell receptor alpha locus Hs.74647 NS NS NS 6
IGHG1 Immunoglobulin heavy constant gamma 1 (G1m marker) Hs.510635 NS NS NS 7
GFRA3 GDNF family receptor alpha 3 Hs.58042 NS NS NS 8
FGFBP2 Fibroblast growth factor binding protein 2 Hs.98785 NS NS NS 9
FOXRED1 FAD-dependent oxidoreductase domain containing 1 Hs.317190 NS NS NS 10
INSIG1 Insulin induced gene 1 Hs.520819 NS NS NS 11
SH3BP5 SH3-domain binding protein 5 (BTK-associated) Hs.654642 NS NS NS 12
PBEF1 Pre-B-cell colony enhancing factor 1 Hs.489615 NS NS NS 14
ZNF182 Zinc finger protein 182 Hs.189690 NS NS NS 15
FOLH1 Folate hydrolase (prostate-specific membrane antigen) 1 Hs.654487 NS NS NS 16

NS: Not Significant (false discovery rate higher than 5%).

assays (i.e., MQRT-PCR and the Nanostring tech- stimulated lymphocytes from the same donors.
nology); nevertheless, some promising candidate CDKN1A and GADD45A are known to be good
biomarkers were not identified in the microarray data. biomarkers of radiation exposure (Jen and Cheung
Ferredoxin reductase (FDXR) for example is not 2005) but they are also involved in signal transduc-
present on the array and could not therefore be tion pathways important for genome stability.
detected. For p53 up-regulated mediator of apoptosis SESN1 is an anti-oxidant gene activated through
(BBC3), also known as PUMA, and Mdm2 p53 p53 transcriptional programme as are CDKN1A and
binding protein homolog (MDM2), the primers and GADD45A (Budanov and Karin 2008).
probes used for quantitative PCR were designed to PRC1 encodes a regulator of cytokinesis which
cover most if not all the alternative transcripts. Some contributes to the correct formation of the spindle
alternative transcripts might be more radiation- during the metaphase (Li et al. 2004). It is responsive
responsive than others and it is possible that the to IR in cultured lymphocytes where cells are
oligonucleotides on the array were not detecting blocked in their cell-cycle progression and modestly
the radiation-responsive ones, thus missing the up- up-regulated at 2 Gy/24 h (as detected by MQRT-
regulation in response to IR revealed by MQRT-PCR. PCR) as surviving cells override the block and
MQRT-PCR confirmed that these three up- progress through mitosis. PRC1 is a good marker
regulated genes which are significantly responsive of radiation exposure in dividing lymphocytes but is
in all individuals, at all time points (2–24 h) at not radiation-responsive in blood. There are no
different doses (2 and 4 Gy) in blood as well as in genes which are down-regulated in all samples in
122 S. Kabacik et al.

Figure 1. Heat maps of microarray data including some of the most radiation responsive genes are shown in cultured lymphocytes (left
panels) and blood (right panels). A colour bar showing the level of expression is shown in the bottom right of the Figure.

blood and lymphocytes. It should be noted that other or time points and both of these methods appear to
genes like FDXR, MDM2 (Figure 3) and damage- have a much superior ‘dynamic range’ over which
specific DNA binding protein 2, 48 kDa (DDB2) they can detect changes. It is important to note that
(data not shown) although not detected by micro- the Nanostring method avoids possible bias due to
array analysis (or not represented on the arrays) are enzymatic amplification of RNA prior to analysis.
also good candidates for biodosimetry; they showed a These methods enable highly sensitive detection and
consistent up-regulation between 2 and 24 h after quantification of the expression of specific gene. For
exposure to 2 and 4 Gy. FDXR is particularly example, BBC3 and PLK3 (polo-like Kinase 3)
promising as the fold of change is about 15-fold at genes, are up-regulated and were both identified as
24 h and can be easily detectable even at lower doses radiation responsive in all samples by MQRT-PCR
and later time points (data not shown). (Figure 5) but not by microarray. The data presented
We have compared the three techniques used in for four genes, suggest that, the contribution of
this study, and the data for four radiation-responsive genetic factors to gene expression response to IR
genes (CDKN1A, GADD45A, PHPT1, CCNG1) exposure is strong enough to affect the level of
are presented in Figure 4. There is a good agreement response; the highest transcriptional response in
between the three methods in that the up-regulation blood appear to be also the highest in cultured
of expression in blood and lymphocytes is detected lymphocytes. Donor two for example (middle
by all three techniques. The nCounter Analysis sample), is consistently showing a stronger up-
System and particularly the MQRT-PCR are more regulation in blood and T-lymphocytes for the four
accurate for detecting differences between doses and/ genes presented; in other words, when comparing
Gene expression response to ionising radiation 123

Figure 2. Gene expression changes in four key index genes. Log2 ratios of each gene are plotted for each donor after 2 and 24 h in response
to either 2 or 4 Gy of X-rays. CDKN1A (panel A), GADD45A (panel B), PRC1 (panel C) and SESN1 (panel D) gene expression changes
obtained by Microarray and MQRT-PCR in cultured lymphocytes (upper panels) and blood (lower panels) for the three donors (1, 2 and 3).
Microarray: Gene expression changes in four key index genes. Log2 ratios of each gene are plotted for each donor after 2 and 24 h in
response to either 2 Gy or 4 Gy X-ray irradiation. Data are presented for one of the three replicate experiments. MQRT-PCR: Gene
expression changes in the same four key index genes. Fold changes in expression compared to non-irradiated cells (and relative to HPRT
gene) of each gene are plotted for each donor after 2 and 24 h in response to either 2 or 4 Gy irradiation. The Figures represent the ratio of
level of expression after irradiation divided by the non-irradiated cell expression levels. The mean of three independent experiments with
triplicate reactions are presented. Values were normalised using HPRT as a standard. Data for donor 1 are presented in blue, in green for
donor 2 and in red for donor 3. Error bars indicate the standard error of the mean (SEM) for n ¼ 3 independent experiments.
124 S. Kabacik et al.

Figure 2. (Continued).

individuals, the up-regulation level in cultured cells previous findings demonstrating that genetic factors
is representative of the level of up-regulation in blood influence the differences in individual transcriptional
(although not true for all genes, e.g., CDKN1A and response as shown in heterozygous carriers of
SESN1 in Figure 1). This is in good agreement with Nijmegen breakage syndrome (Cheung and Ewens
Gene expression response to ionising radiation 125

monitored the expression level of numerous genes


amongst which some may be suitable for biological
dosimetry.
The following genes have been identified either by
microarray analysis or by MQRT-PCR: SESN1,
GADD45A, CDKN1A, CCNG1, FDXR, BBC3
DDB2, PHPT1, MDM2, PCNA, PLK3 as well as
FAS, FAS and v-mic v-myc myelocytomatosis viral
oncogene homolog (c-MYC) (data not shown) as
being up-regulated in blood in the three donors
analysed. For most of these genes radiation re-
sponses are long lasting with CDKN1A, SESN1 and
GADD45A still being up-regulated at 48 h post-
exposure (2 Gy) (data not shown). It will be very
important to demonstrate that the up-regulation of
the identified biomarkers last in time (72 h or even
longer). These experiments will have to be carried
out on blood exposed in vivo as in our experience
RNA extracted from blood after more than 48 h
tends to show some degradation. Our data confirm
previous work (Meadows et al. 2008) which showed
that gene expression response to radiation is specific,
can be long lasting and is quantitatively accurate. It is
probable that these methods can be refined in the
Figure 3. Up-regulation in cultured lymphocytes and blood of two near future by for example, sorting of lymphocytes
genes MDM2 (panel A) and FDXR (panel B) not previously sub-populations which have a more pronounced
identified by microarray. Fold changes in expression assessed by transcriptional response to radiation. For instance,
MQRT-PCR compared to non-irradiated cells (and relative to it has been shown that several biological responses in
HPRT gene) of each gene are plotted for each donor after 2 and
cluster of differentiation (CD) 4þ cells could be more
24 h in response to either 2 or 4 Gy irradiation. The Figures
represent the ratio of level of expression after irradiation divided by sensitive to low doses of radiation than CD56þ and
the un-irradiated cell expression levels. The mean of three CD8þ (Gruel et al. 2008) although these refinements
independent experiments with triplicate reactions are presented. may prove to be time-consuming and not suitable for
Values were normalised using HPRT as a standard. Error bars mass screening. Other tissues sustain long term
indicate the standard error of the mean (SEM) for n ¼ 3
changes in gene expression after in vivo exposure to
independent experiments.
IR. Mouse liver maintains a high level of CDKN1A
following a sublethal dose of 6 Gy for at least 10
weeks (Pawlik et al. 2009).
2006) and Ataxia telangiectasia (Watts et al. 2002, In order to study the impact of IR on changes in
Smirnov and Cheung 2008). gene expression in cycling cells, we used a protocol
where leukocytes are stimulated to obtain extended-
term cultures of human T-lymphocytes exclusively
Discussion
(O’Donovan et al. 1995); the gene expression
We have used whole genome microarray expression profiles are specific to dividing T-lymphocytes. On
profiling to identify genes which could potentially help the other hand, resting blood leukocytes are con-
to estimate quantitatively radiation exposures across a stituted of different cell-types such as monocytes and
dose range relevant for medical decision making in a neutrophils. Each sub-population has a specific
radiologic emergency. MQRT- PCR was performed expression pattern and some of the differences in
to confirm the array results and increase the sensitivity gene expression detected here are cell-type depen-
to evaluate inter-individual differences. We also tested dant. Nevertheless, many of the genes which expres-
a recently developed high throughput digital technol- sion is significantly down-regulated by IR-exposure
ogy (NanoString nCounter) (Geiss et al. 2008) on in cycling T-lymphocytes are associated with the cell-
selected genes to assess its potential role in quantita- cycle, not to a specific cell type. Genes like BUB1,
tive radiation dosimetry. CCNA2, PRC1, CENPE (Table IIB) and others,
Exposure to IR leads to complex cellular responses like cell division cycle 25 homolog C (CDC25C),
that include changes in gene expression, and these cyclin B1 (CCNB1), mitotic arrest deficient-2
gene expression responses can differ between in- (MAD2L1) are down-regulated and there repression
dividuals. Nevertheless, in the present study, we was confirmed by MQRT-PCR. Although they are
126 S. Kabacik et al.

Figure 4. Comparison of data obtained from the three different techniques (MQRT-PCR, microarray and NanoString nCounter). The final
ratios, expressed as fold change for the three methods, were generated by comparing expression levels in irradiated samples with basal levels
in non-irradiated samples. The mean of three independent experiments (microarray) and of triplicate reactions (MQRT-PCR and
NanoString nCounter) are presented for donor 1.

poor candidates for biological dosimetry purposes as radiation-induced cancer. The biomarkers identified
they are not responsive in blood, the modification in in this study can potentially be used to identify
transcription level in response to IR of genes individuals who have suffered accidental exposure.
associated with chromosome instability and aneu- The existence of heritable radiosensitivity syn-
ploidy is extremely interesting. Genes like PRC1 dromes (e.g., Ataxia telangiectasia [AT]) suggests
which associates with mitotic spindles, BUB1 master that normal tissue reaction severity is determined, at
regulator of spindle assembly checkpoint (Baker least in part, by genetic factors, and these may be
et al. 2009, Marchetti and Venkatachalam 2010) revealed by differences in gene expression. This is
and MAD2L1, haplo-insufficiency of which causes also suggested by the results shown in Figure 5. Lack
premature anaphase and chromosome instability in of or low expression in response to IR exposure could
mammalian cells (Michel et al. 2001) are all down- also help to identify individuals who are predisposed
regulated in cycling lymphocytes in response to IR. to negative outcome of exposure. For example, we
The assessment of expression level of genes asso- have previously shown that post-irradiation expres-
ciated with the spindle assembly checkpoint and sion response of CDKN1A in peripheral blood was
proper chromosome segregation can have important significantly reduced in patients with severe acute
consequences for tumorigenesis and developmental skin reactions (Badie et al. 2008). A clear differential
abnormalities studies. A signature of chromosomal transcriptional response was also demonstrated be-
instability obtained from gene expression profiles has tween healthy donors and individuals with a defined
been identified recently which predicts clinical out- genetic defect like AT (Badie et al. 2008).
come in multiple human cancers (Carter et al. 2006). To summarise, radiation-responsive genes show-
Remarkably, 11 out of the 25 genes which form their ing relatively little intra-individual variation in
classifier are significantly down-regulated in our irradiated blood are the best candidates for dose
irradiated cultured lymphocyte samples. estimation. The main purpose of this manuscript was
There is concern surrounding the increasing use of to establish a panel of highly radiation responsive
IR in medical diagnostics and the associated risk of genes suitable for biological dosimetry and this has
Gene expression response to ionising radiation 127

Figure 5. Individual transcriptional responses to radiation exposure 24 h after 4 Gy in four radiation-responsive genes, PLK3, BBC3,
GADD45A and ATF3 assessed by MQRT-PCR. Error bars indicate the standard error of the mean (SEM) for n ¼ 3 independent
experiments.

been achieved. As the ultimate aim is to be able to patterns of gene expression in association with
estimate the radiation dose, we started additional radiation exposure. The ability of a single gene set
work to study the low dose sensitivity of these genes to predict radiation exposure throughout a window
and to produce full dose-responses and time-courses of time is an important advance in the development
in blood exposed ex vivo and in vivo in mice. of gene expression for practical biological dosimetry
Some others may be particularly good as suscept- (Paul and Amundson 2008, Brengues et al. 2010).
ibility biomarkers, for example the down-regulated Some of the genes identified in this study are
genes in cultured lymphocytes. involved in signal transduction networks which
could well be also relevant to those of radiation
induced cancer. They provide research tools to
Conclusion
examine in vivo dosimetry, but also therapeutic
We have used three different techniques available to response and mechanisms of radiation tumorigen-
study gene expression in response to IR exposure in esis. Confirmation and extension of these results
human blood and cultured lymphocytes from three in vivo with animal models or radiotherapy patient
healthy donors. Changes in gene expression assessed samples may enable the development of simple
by DNA microarrays provided a list of radiation clinical tests to predict the likely level of radiation
responsive genes for which the variations in tran- toxicity and to individualise patient treatment as
scriptional response can be validated and measured well as provide robust tools for biological dosime-
more accurately and with greater sensitivity by try.
MQRT-PCR. The results obtained were validated
by a third independent technique scoring mRNA
Acknowledgements
copies directly.
Array analysis is very useful for identification of We thank Richard Doull (MRC Harwell) for
responsive transcripts but the MQRT-PCR and irradiations. This work was supported in part by
Nanostring methods have distinct advantages for Cancer Research UK (C181/A6976) and Break-
quantitative analysis sensitive for the detection of through Breast Cancer.
128 S. Kabacik et al.

Declaration of interest: The authors report no Finnon P, Robertson N, Dziwura S, Raffy C, Zhang W, Ainsbury
conflicts of interest. The authors alone are respon- L, Kaprio J, Badie C, Bouffler S. 2008. Evidence for significant
heritability of apoptotic and cell cycle responses to ionising
sible for the content and writing of the paper. radiation. Human Genetics J3:485–493.
Franco N, Lamartine J, Frouin V, Le Minter P, Petat C, Leplat JJ,
Libert F, Gidrol X, Martin MT. 2005. Low-dose exposure to
References gamma rays induces specific gene regulations in normal human
Amundson SA, Do KT, Fornace AJ Jr. 1999. Induction of stress keratinocytes. Radiation Research. 3:623–635.
genes by low doses of gamma rays. Radiation Research Fry RC, Svensson JP, Valiathan C, Wang E, Hogan BJ,
152:225–231. Bhattacharya S, Bugni JM, Whittaker CA, Samson LD.
Amundson SA, Fornace AJ Jr. 2003. Monitoring human radiation 2008. Genomic predictors of interindividual differences in
exposure by gene expression profiling: Possibilities and pitfalls. response to DNA damaging agents. Genes Development
Health Physics 85:36–42. 22:2621–2626.
Amundson SA, Lee RA, Koch-Paiz CA, Bittner ML, Meltzer P, Garty G, Chen Y, Salerno A, Turner H, Zhang J, Lyulko O,
Trent JM, Fornace AJ Jr. 2003. Differential responses of stress Bertucci A, Xu Y, Wang H, Simaan N, Randers-Pehrson G,
genes to low dose-rate gamma irradiation. Molecular Cancer Yao YL, Amundson SA, Brenner DJ. 2010. The RABIT: A
Research 6:445–452. rapid automated biodosimetry tool for radiological triage.
Amundson SA, Grace MB, McLeland CB, Epperly MW, Yeager Health Physics 8:209–217.
A, Zhan Q, Greenberger JS, Fornace AJ Jr. 2004. Human Geiss GK, Bumgarner RE, Birditt B, Dahl T, Dowidar N,
in vivo radiation-induced biomarkers: Gene expression changes Dunaway DL, Fell HP, Ferree S, George RD, Grogan T,
in radiotherapy patients. Cancer Research 64:6368–6371. James JJ, Maysuria M, Mitton JD, Oliveri P, Osborn JL, Peng
Badie C, Dziwura S, Raffy C, Tsigani T, Alsbeih G, Moody J, Finnon T, Ratcliffe AL, Webster PJ, Davidson EH, Hood L, Dimitrov
P, Levine E, Scott D, Bouffler S. 2008. Aberrant CDKN1A K. 2008. Direct multiplexed measurement of gene expression
transcriptional response associates with abnormal sensitivity to with color-coded probe pairs. Nature Biotechnology 3:
radiation treatment. British Journal of Cancer 98:1845–1851. 317–325.
Baker DJ, Jin F, Jeganathan KB, van Deursen JM. 2009. Whole Grace MB, McLeland CB, Blakely WF. 2002. Real-time
chromosome instability caused by Bub1 insufficiency drives quantitative RT-PCR assay of GADD45 gene expression
tumorigenesis through tumor suppressor gene loss of hetero- changes as a biomarker for radiation biodosimetry. Interna-
zygosity. Cancer Cell 16:475–486. tional Journal of Radiation Biology 8:1011–1021.
Blakely WF, Miller AC, Grace MB, McLeland CB, Luo L, Grace MB, McLeland CB, Gagliardi SJ, Smith JM, Jackson WE 3rd,
Muderhwa JM, Miner VL, Prasanna PG. 2003. Radiation Blakely WF. 2003. Development and assessment of a quan-
biodosimetry: Applications for spaceflight. Advanced Space titative reverse transcription-PCR assay for simultaneous
Research 31:1487–1493. measurement of four amplicons. Clinical Chemistry. 49:
Blakely WF, Salter CA, Prasanna PG, 2005. Early-response 1467–1475.
biological dosimetry – recommended countermeasure en- Gruel G, Lucchesi C, Pawlik A, Frouin V, Alibert O,
hancements for mass-casualty radiological incidents and Kortulewski T, Zarour A, Jacquelin B, Gidrol X, Tronik-
terrorism. Health Physics 9:494–504. Le Roux D. 2006. Novel microarray-based method for
Brengues M, Paap B, Bittner M, Amundson S, Seligmann B, Korn estimating exposure to ionizing radiation. Radiation Re-
R, Lenigk R, Zenhausern F. 2010. Biodosimetry on small blood search. 166:746–756.
volume using gene expression assay. Health Physics F8:179–185. Gruel G, Voisin P, Vaurijoux A, Roch-Lefevre S, Grégoire E,
Budanov AV, Karin M. 2008. p53 target genes sestrin1 and Maltere P, Petat C, Gidrol X, Voisin P, Roy L. 2008. Broad
sestrin2 connect genotoxic stress and mTOR signaling. Cell modulation of gene expression in CD4þ lymphocyte subpo-
134:451–460. pulations in response to low doses of ionizing radiation.
Carter SL, Eklund AC, Kohane IS, Harris LN, Szallasi Z. 2006. A Radiation Research 170:335–344.
signature of chromosomal instability inferred from gene International Atomic Energy Agency (IAEA). 2001. Cytogenetic
expression profiles predicts clinical outcome in multiple analysis for radiation dose assessment: A manual. Technical reports
human cancers. Nature Genetics 8:1043–1048. Series No. 405, Vienna: IAEA. Available from the website: http://
Chaudhry MA. 2008. Biomarkers for human radiation exposure. www-pub.iaea.org/MTCD/publications/. . ./TRS405_scr.pdf
Journal of Biomedical Science Sep;15(5):557–563. Jen KY, Cheung VG. Transcriptional response of lymphoblastoid
Cheung VG, Ewens WJ. 2006. Heterozygous carriers of Nijmegen cells to ionizing radiation. 2003. Genome Research 132092–
Breakage Syndrome have a distinct gene expression phenotype. 132100.
Genome Research 8:973–979. Jen KY, Cheung VG. 2005. Identification of novel p53 target
Correa CR, Cheung VG. 2004. Genetic variation in radiation- genes in ionizing radiation response. Cancer Research 65:
induced expression phenotypes. American Journal of Human 7666–7673.
Genetics 5:885–890. Kang CM, Park KP, Song JE, Jeoung DI, Cho CK, Kim TH, Bae
Ding LH, Shingyoji M, Chen F, Chatterjee A, Kasai KE, Chen S, Lee SJ, Lee YS. 2003. Possible biomarkers for ionizing
DJ. 2005. Gene expression changes in normal human skin radiation exposure in human peripheral blood lymphocytes.
fibroblasts induced by HZE-particle radiation. Radiation Radiation Research 159:312–319.
Research 4:523–526. Li C, Lin M, Liu J. 2004. Identification of PRC1 as the p53 target
Dressman HK, Muramoto GG, Chao NJ, Meadows S, Marshall gene uncovers a novel function of p53 in the regulation of
D, Ginsburg GS, Nevins JR, Chute JP. 2007. Gene expression cytokinesis. Oncogene 23:9336–9347.
signatures that predict radiation exposure in mice and humans. Lindholm C, Stricklin D, Jaworska A, Koivistoinen A, Paile W,
PLoS Medicine 4:e106. Arvidsson E, Deperas-Standylo J, Wojcik A. 2010. Premature
Fan F, Jin S, Amundson SA, Tong T, Fan W, Zhao H, Zhu X, chromosome condensation (PCC) assay for dose assessment in
Mazzacurati L, Li X, Petrik KL, Fornace AJ Jr, Rajasekaran B, mass casualty accidents. Radiation Research 173:71–78.
Zhan Q. 2001. ATF3 induction following DNA damage is Mackay A, Tamber N, Fenwick K, Iravani M, Grigoriadis A,
regulated by distinct signaling pathways and over-expression of Dexter T, Lord CJ, Reis-Filho JS, Ashworth A. 2009. A high-
ATF3 protein suppresses cells growth. Oncogene 21:7488–7496. resolution integrated analysis of genetic and expression profiles
Gene expression response to ionising radiation 129

of breast cancer cell lines. Breast Cancer Research Treatment primary human acute myeloid leukemia samples. Journal of
118:481–498. Clinical Investigation 119:1714–1726.
Marchetti F, Venkatachalam S. 2010. The multiple roles of Bub1 Petrik KL, Fornace AJ Jr, Rajasekaran B, Zhan Q. 2002. ATF3
in chromosome segregation during mitosis and meiosis. Cell induction following DNA damage is regulated by distinct
Cycle 9:58–63. signaling pathways and over-expression of ATF3 protein
Meadows SK, Dressman HK, Muramoto GG, Himburg H, suppresses cells growth. Oncogene 21:7488–7496.
Salter A, Wei Z, Ginsburg GS, Chao NJ, Nevins JR, Chute Smirnov DA, Cheung VG. 2008. ATM gene mutations result in
JP. 2008. Gene expression signatures of radiation response both recessive and dominant expression phenotypes of genes and
are specific, durable and accurate in mice and humans. PLoS microRNAs. American Journal of Human Genetics 83:243–253.
One 3:e1912. Stassen T, Port M, Nuyken I, Abend M. 2003. Radiation-induced
Michel LS, Liberal V, Chatterjee A, Kirchwegger R, Pasche B, Gerald gene expression in MCF-7 cells. International Journal of
W, Dobles M, Sorger PK, Murty VV, Benezra R. 2001. MAD2 Radiation Biology 79:319–331.
haplo-insufficiency causes premature anaphase and chromosome Stoimenov I, Helleday T. 2009. PCNA on the crossroad of cancer.
instability in mammalian cells. Nature 409:355–359. Biochemical Society Transactions 37:605–613.
O’Donovan MP, Freemantle MR, Hull G, Bell DA, Arlett CF, Tusher VG, Tibshirani R, Chu G. 2001. Significance analysis of
Cole J. 1995. Extended-term cultures of human T-lympho- microarrays applied to the ionizing radiation response.
cytes: A practical alternative to primary human lymphocytes for Proceedings of the National Academy of Sciences of the
use in genotoxicity testing. Mutagenesis 10:189–201. USA 985116–985121.
Ossetrova NI, Blakely WF. 2009. Multiple blood-proteins Watts JA, Morley M, Burdick JT, Fiori JL, Ewens WJ, Spielman
approach for early-response exposure assessment using an RS, Cheung VG. 2002. Gene expression phenotype in
in vivo murine radiation model. International Journal of heterozygous carriers of Ataxia telangiectasia. American
Radiation Biology 85:837–850. Journal of Human Genetics 71:791–800.
Paul S, Amundson SA. 2008. Development of gene expression Willems P, August L, Slabbert J, Romm H, Oestreicher U, Thierens
signatures for practical radiation biodosimetry. International H, Vral A. 2010. Automated micronucleus (MN) scoring for
Journal of Radiation Oncology Biology Physics 71: population triage in case of large scale radiation events.
1236–1244. International Journal of Radiation Biology 6:2–11.
Pawlik A, Delmar P, Bosse S, Sainz L, Petat C, Pietu G, Thierry D, Wojcik A, Lloyd D, Romm H, Roy L. 2010. Biological dosimetry
Tronik-Le Roux D. 2009. Changes in transcriptome after in vivo for triage of casualties in a large-scale radiological emergen-
exposure to ionising radiation reveal a highly specialised liver cy:capacity of the EU member states. Radiation Protection
response. International Journal of Radiation Biology 85:656–671. Dosimetry 138:397–401.
Payton JE, Grieselhuber NR, Chang LW, Murakami M, Geiss Woloschak GE, Chang-Liu CM. 1995. Modulation of expression of
GK, Link DC, Nagarajan R, Watson MA, Ley TJ. 2009. High genes encoding nuclear proteins following exposure to JANUS
throughput digital quantification of mRNA abundance in neutrons or gamma-rays. Cancer Letters 97:169–175.

You might also like