Skip to main content
Erschienen in: Radiation Oncology 1/2016

Open Access 01.12.2016 | Research

Transcriptomic analyses of the radiation response in head and neck squamous cell carcinoma subclones with different radiation sensitivity: time-course gene expression profiles and gene association networks

verfasst von: Agata Michna, Ulrike Schötz, Martin Selmansberger, Horst Zitzelsberger, Kirsten Lauber, Kristian Unger, Julia Hess

Erschienen in: Radiation Oncology | Ausgabe 1/2016

Abstract

Background

Acquired and inherent radioresistance of tumor cells is related to tumor relapse and poor prognosis – not only in head and neck squamous cell carcinoma (HNSCC). The underlying molecular mechanisms are largely unknown. Therefore, systemic in-depth analyses are needed to identify key regulators of radioresistance. In the present study, subclones of the CAL-33 HNSCC cell line with different radiosensitivity were analyzed to identify signaling pathways related to the different phenotypes.

Methods

Subclones with altered radiosensitivity were generated by fractionated irradiation of the parental CAL-33 cells. Differences in radiosensitivity were confirmed in colony formation assays. Selected subclones were characterized at the genomic and transcriptomic level by SKY, array CGH, and mRNA-microarray analyses. Time-course gene expression analyses upon irradiation using a natural cubic spline regression model identified temporally differentially expressed genes. Moreover, early and late responding genes were identified. Gene association networks were reconstructed using partial correlation. The Reactome pathway database was employed to conduct pathway enrichment analyses.

Results

The characterization of two subclones with enhanced radiation resistance (RP) and enhanced radiosensitivity (SP) revealed distinct genomic and transcriptomic changes compared to the parental cells. Differentially expressed genes after irradiation shared by both subclones pointed to important pathways of the early and late radiation response, including senescence, apoptosis, DNA repair, Wnt, PI3K/AKT, and Rho GTPase signaling. The analysis of the most important nodes of the gene association networks revealed pathways specific to the radiation response in different phenotypes of radiosensitivity. Exemplarily, for the RP subclone the senescence-associated secretory phenotype (SASP) together with GPCR ligand binding were considered as crucial. Also, the expression of endogenous retrovirus ERV3-1in response to irradiation has been observed, and the related gene association networks have been identified.

Conclusions

Our study presents comprehensive gene expression data of CAL-33 subclones with different radiation sensitivity. The resulting networks and pathways associated with the resistant phenotype are of special interest and include the SASP. The radiation-associated expression of ERV3-1 also appears highly attractive for further studies of the molecular mechanisms underlying acquired radioresistance. The identified pathways may represent key players of radioresistance, which could serve as potential targets for molecularly designed, therapeutical intervention.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13014-016-0672-0) contains supplementary material, which is available to authorized users.

Background

Head and neck squamous cell carcinoma (HNSCC) develops in approx. 139,000 individuals per year in Europe with a survival rate of approx. 70 % at 1 year and approx. 40 % at 5 years after therapy [1]. More than 90 % of head and neck cancers are classified as HNSCC and originate from the oral cavity, nasopharynx, oropharynx, hypopharynx, or larynx, respectively [2]. The major risk factors for HNSCC are tobacco smoking, alcohol abuse, and poor oral health [3, 4]. For oropharyngeal cancers, infection with high-risk human papilloma viruses is another important risk factor [5]. Thus, HNSCC is a very heterogeneous cancer entity also in terms of therapy response. Surgical resection followed by radio(chemo)therapy is the standard treatment of HNSCC patient [6, 7]. In locally advanced HNSCC, surgery is often limited by the complex anatomy of the affected region and, therefore, definitive radiochemotherapy is an important treatment option. However, acquired and/or inherent radioresistance of tumor cells is a common cause for tumor relapse and poor prognosis. Tumor cells derived from HNSCCs after radiotherapy have been reported to be more radioresistant than cell lines established prior to therapy, thus strengthening the clinical relevance of acquired radioresistance [8]. Along these lines, it was proposed that fractionated irradiation might preferentially eradicate radiosensitive cells, whereas radioresistant cells remain largely untouched. Accordingly, recurrent tumors mostly consist of radioresistant cells [8]. Although different potential mechanisms of radioresistance have been proposed and extensively studied, the underlying molecular details remain largely unknown [9]. Systemic in-depth analyses are needed in order to identify the master regulators of acquired radioresistance, which could serve as potential biomarkers and future therapeutic targets in novel combined modality approaches.
In this study, we characterized two subclones (#303 and #327) derived from the CAL-33 HNSCC cell line, which were generated by fractionated radiation treatment of the parental cells. CAL-33 is an HPV-negative HNSCC cell line that has been established by Gioanni et al. (1988) from a biopsy specimen prior to treatment from a squamous cell carcinoma of the tongue from a male patient [10]. The subclones derived thereof differed in radiosensitivity when compared to the parental CAL-33 cell line. Interestingly, one subclone was significantly more radioresistant, whereas the other one was significantly more radiosensitive. In order to identify potential key regulators of altered radiation sensitivity, the subclones were characterized on the genomic and transcriptomic level. Furthermore, time-course gene expression analyses were performed upon irradiation, and gene association network reconstruction and pathway enrichment analyses were utilized to identify signaling pathways related to the observed radiation phenotypes.

Methods

Cell culture

The human head and neck squamous cell carcinoma (HNSCC) cell line CAL-33 was obtained from the German collection of microorganisms and cell cultures (DSMZ). Cells were maintained in DMEM GlutaMAX I medium supplemented with 10 % FCS and 1 % penicillin/streptomycin and cultured at 37 °C/5 % CO2. Cells were mycoplasma-free as tested by the MycoAlert (Lonza) mycoplasma detection kit.

Generation of clones with altered radiation resistance

In order to generate radioresistant subclones of the parental CAL-33 cell line, exponentially growing CAL-33 cells were exposed to fractionated irradiation (Mueller RT-250 γ-ray, tube Thoraeus Filter, 200 kV, 10 mA) according to a schedule commonly used in radiotherapy: A total dose of 20 Gy was given in daily fractions of 2 Gy 5 times per week. For each week, 2 days of recovery time were included. Afterwards, cells were cloned by limiting dilution procedure and grown for 4 to 12 weeks.

Colony forming assay

Clonogenic survival was determined in colony formation assays as described previously [11]. Briefly, cells were seeded into 6-well plates, allowed to adhere for 4 h, and irradiated at 0, 1, 2, 4, 6 or 8 Gy, respectively (Mueller RT-250 γ-ray, Thoraeus Filter, 200 kV, 10 mA). 14 days after irradiation, colonies were fixed and stained with methylene blue. Only colonies consisting of at least 50 cells were scored. Each assay was carried out in duplicates of three different cell densities per each irradiation dose. Results from three independent experiments were subjected to linear-quadratic regression analyses employing the maximum likelihood approach. Differences between curves were evaluated using F-test [12].

Proliferation and cell cycle analyses

Proliferation rates were determined over a period of three days upon seeding 20,000 cells per well into 24-well plates. Cells were harvested by trypsinization, and total cell numbers were determined by manual counting. Cell numbers were plotted against the growth time, and doubling times were calculated by semi-log regression analyses in the exponential growth phase. Dynamics of cell cycle distribution upon irradiation at 4 Gy was analyzed by flow cytometric phospho-histone H3(S10)/propidium iodide (PI) staining as described in [13]. Briefly, cells were collected by trypsinization and fixed in 70 % ethanol. After extensive washing, cells were stained with anti-phospho-histone-H3-Alexa488 (pH3(S10)) antibody (New England Biolabs, Frankfurt, Germany) and PI/RNase staining solution (BD Biosciences, Heidelberg, Germany). Data of 10,000 cells were recorded on an LSRII flow cytometer (BD Biosciences), and cell cycle analyses were performed by using FlowJo 7.6.5 software (Tree Star Inc., Ashland, OR, USA).

Spectral karyotyping (SKY)

Metaphase chromosome spreads were prepared from untreated CAL-33 parental cell line and generated CAL-33 sublines. Colcemid (Roche) was added at a final concentration of 0.1 μg/ml to the culture medium of exponentially growing cells at a density of 6 × 106 cells per 75 cm2. After 3 h of incubation time, cells were washed with PBS, trypsinized, suspended in fresh culture medium followed by hypotonic KCl treatment (75 mM) at 37 °C for 25 minutes. Following centrifugation, cells were resuspended in 2–3 ml of fixation solution and approximately 40–50 μl of cell suspension was dropped on several microscope slides. After one week of ageing at room temperature, spectral karyotyping was performed as described by Hieber et al. [14]. The karyotype of each cell line was determined based on a minimum of 15 metaphases. Chromosomal aberrations were detectable by color junctions within affected chromosomes. Spectral imaging and image analysis were performed with a SpectraCube system and SkyView imaging software (both from Applied Spectral Imaging).

Genomic copy number typing (array CGH)

In order to characterize copy number changes of parental CAL-33 and generated CAL-33 sublines, array comparative genomic hybridization analysis (array CGH) was performed on high-resolution oligonucleotide-based SurePrint G3 Human 180 k CGH microarrays (AMADID 252206, Agilent Technologies). DNA from non-irradiated samples was isolated using the QIAamp DNA Mini Kit (Qiagen). The DNA concentration was quantified with the NanoDrop 1000 Spectrophotometer (Thermo Fisher Scientific, Germany). Slight modifications of the original Agilent array CGH protocol were introduced. 250 ng isolated cell line DNA and 250 ng sex-mismatched normal reference DNA (Promega) were labeled with Cy3 and Cy5, respectively, using the CGH labeling kit for oligo arrays (Enzo) following the Enzo’s protocol. Microcon YM-30 columns (Millipore) were used to remove the unincorporated nucleotides. Subsequent labeled DNA hybridization, washing and scanning of the CGH arrays were continued according to the Agilent’s protocol. The fluorescence intensities were extracted as text files with the Feature Extraction software 10.7 (Agilent Technologies). Obtained data were imported into the R statistical platform (version 3.2.2, www.​r-project.​org) and filtered for quality outliers using the QA measurements generated by the Feature Extraction software. Experimental artifacts were removed from the array CGH data using spatial normalization as suggested and described in MANOR R-package manual [15] and [16]. Array CGH profiles containing a wave bias that appear as waves in plots were removed using ridge regression based algorithms implemented in NoWaves R-package available from http://​www.​few.​vu.​nl/​~mavdwiel/​nowaves.​html [17]. Following normalization and/or wave bias removal, data were segmented using circular binary segmentation algorithms as implemented in DNAcopy R-package [18] in order to detect breakpoints and levels in single array CGH profiles [19]. Chromosomal gains and losses were determined using CGHcall algorithm implemented in CGHcall R-package [20]. To reduce data complexity, copy number calls were transformed into regions using the R-package CGHregions [21].

Irradiation and sample preparation

Cells were seeded into 6-well plates and allowed to adhere for 16 h. The numbers of plated cells were adjusted to the incubation times: For samples collected 0.25, 2, 7, 12, and 24 h after irradiation, 3.5 × 105 cells/well were seeded, whereas for those collected after 48, 72, and 96 h, 1.75 × 105 cells/well were used. Cells were irradiated at 0 or 8 Gy of gamma-irradiation (Mueller RT-250, Thoraeus Filter, 200 kV, 10 mA) at a dose rate of 1.3 Gy/min, and samples were collected after 0.25, 2, 7, 12, 24, 48, 72, and 96 h by scraping on ice. The washed, dry cell pellet was snap frozen and stored at −80 °C. Samples from 3 independent experiments were used for subsequent transcriptomic analyses. Total RNA was isolated using the Rneasy Mini Kit (Qiagen) including a DNase digestion step, according to the manufacturer’s protocol. The concentration of RNA was quantified with a Qubit 2.0 Fluorometer (Life Technologies), and RNA integrity was confirmed with a Bioanalyzer 2100 (Agilent Technologies). Samples with RNA integrity number (RIN) >7 were used in subsequent gene expression microarrays analyses.

Global gene expression profiling

Global gene expression profiling of all CAL-33 cell lines was performed on SurePrint G3 Human Gene Expression 8x60k microarrays (Agilent Technologies, AMADID 28004) using 60 ng of total RNA according to the manufacturer’s protocol (one-color Low Input Quick Amp Labeling Kit, Agilent Technologies). Raw gene expression data were extracted as text files with the Feature Extraction software 11.0.1.1 (Agilent Technologies). All data analysis was conducted using the R statistical platform (version 3.2.2, www.​r-project.​org) [22]. Data quality assessment, filtering, preprocessing, normalization, batch correction based on nucleic acid labeling batches and data analyses were carried out with the Bioconductor R-packages limma, Agi4x44PreProcess and the ComBat function of the sva R-package [2325]. All quality control, filtering, preprocessing and normalization thresholds were set to the same values as suggested in Agi4x44PreProcess R-package user guide [25]. Only HGNC annotated genes were used in the analysis. For multiple microarray probes representing the same gene the optimal probe was selected according to the Megablast score of probe sequences against the human reference sequence (http://​www.​ncbi.​nlm.​nih.​gov/​refseq/​) [26]. If the resulted score was equal for two or more probes, the probe with the lowest differential gene expression FDR value was kept for further analyses since only one expression value per gene was allowed in subsequent gene association network (GAN) reconstruction analysis.

Differential gene expression analysis

The time-course differential gene expression analyses were conducted between irradiated and control cells (sham-irradiated) using a natural cubic spline regression model with three degrees of freedom as described in splineTimeR R-package [27]. Obtained p-values were adjusted by the Benjamini-Hochberg method for false discovery [28]. Genes with an adjusted p-value (FDR, false discovery rate) lower than 0.05 were considered as differentially expressed and associated with radiation response. For the status quo experiment that compares the derived CAL-33 clones with the parental CAL-33 cell line, genes were considered as differentially expressed when a log2 fold-change was higher than 0.5 and a FDR value lower than 0.05.

Identification of early and late responding genes

Temporally differentially expressed genes with fold-change above 2.0 or below 0.5 in any measured time points within the first day after irradiation were considered as early responding genes. Respectively, genes with fold-change above 2.0 or below 0.5 within the second, third or fourth day of irradiation were considered as late responding.

Gene association network reconstruction and identification of important nodes in the reconstructed networks

Temporally differentially expressed genes were subjected to gene association network (GAN) reconstruction using a regularized dynamic partial correlation method [29]. Pairwise relationships between genes over time were inferred based on a dynamic Bayesian network model with shrinkage estimation of covariance matrices as implemented in the GeneNet R-package [30]. Analyses were conducted with a posterior probability of 0.95 for each potential undirected edge. Further, in order to determine the importance of each node in the reconstructed association networks, graph topological analyses based on centrality measures were applied [31]. Three most commonly used centrality measures: degree, shortest path betweenness and closeness describing the importance of gene in a network were combined into one centrality measure [32]. For each gene the three centrality values where ranked and the consensus centrality measure for each node was defined as the mean of the three independent centrality ranks.

Pathway enrichment analysis

The Reactome pathway database was used to conduct the pathway enrichment analysis in order to further investigate the functions of the selected sets of differentially expressed genes [33]. Only pathways containing not more than 600 genes and not less than 20 genes were considered. Thereby, too general and too specific pathways were excluded from the analysis. Statistical significance of enriched pathways was determined by one-sided Fisher’s exact test. The resulting p-values were adjusted for FDR using the Benjamini-Hochberg method.

qRT-PCR technical validation of gene expression data

For technical validation of the gene expression microarray data, RNA samples (500 ng) were reversely transcribed using the QuantiTect Reverse Transcription Kit (Qiagen) according to the manufacturer’s protocol and subjected to qRT-PCR reactions (10 μl) on a ViiA 7 qPCR system (Life Technologies). The following Taqman® Assays were used (Life Technologies): AKT3 (Hs00987350_m1), GADD45A (Hs01077132_m1), MAL (Hs00360838_m1), HOPX (Hs04188695_m1), HYAL3 (Hs00185910_m1), TUBGCP3 (Hs00902139_m1), RGS16 (Hs00892674_m1), TNFAIP3 (Hs00234713_m1). ACTB (Hs01060665_g1) and GAPDH (Hs99999905_m1) served as endogenous reference genes. Relative expression levels were calculated using the ΔΔCt method and Spearman correlation analyses with microarray data were performed. Validation was considered successful for Spearman’s rho > 0.5. Additionally, the fold-change values obtained from microarrays and qRT-PCR were compared.

Results and discussion

Tumor relapse after radiochemotherapy in HNSCC is often linked to intrinsic and/or acquired radioresistance of tumor cells. However, the underlying molecular mechanisms remain largely unknown [9]. To gain knowledge on this fundamental and clinically relevant process, we established an in vitro model of acquired phenotypes of discrepant radiosensitivity in CAL-33 cells. The underlying molecular mechanisms were investigated by static and dynamic global mRNA expression analyses with subsequent network reconstruction and pathway enrichment analyses.

CAL-33 subclones with different phenotypes of radiosensitivity and cytogenetic characteristics

The parental cell line CAL-33 was repeatedly irradiated in order to generate subclones with different phenotypes of radiosensitivity. To analyze acquired alterations in radiosensitivity of the derived CAL-33 subclones, long-term survival upon gamma-irradiation was assessed by colony formation assays (Fig. 1). For further analyses we selected two subclones. Both subclones #303 and #327 showed statistically significant differences (p-values < 0.0001) when compared to the parental CAL-33 cells. Interestingly, subclone #303 showed increased radiosensitivity (sensitive phenotype, SP), whereas subclone #327 was more radioresistant (resistant phenotype, RP) – particularly in the dose range > 4 Gy.
At first glance, the emergence of more radiation sensitive subclones appears unexpected and might be related to clonal evolution that was initiated by the irradiation-induced genomic alterations and that might occur at sublethal doses. This phenomenon has also been observed in previous studies [34, 35]. In comparison to other HNSCC cell lines, CAL-33 is very radioresistant a priori and this might explain why it appears to be very difficult to generate subclones with an even more resistant phenotype.
To analyze structural and numerical chromosomal aberrations in comparison to the parental cell line, the subclones were cytogenetically characterized by SKY analyses. Structural and numerical aberrations identified by SKY in the parental CAL-33 cell line involved chromosomes 3, 7, 8, 9, 16, 18, 20, X, and Y (Fig. 2a). Structural and numerical aberrations of the SP subclone included chromosomes 2, 3, 7, 8, 9, 11, 16, 18, 20, 21, X, and Y (Fig. 2b). Chromosomes 1, 3, 4, 5, 7, 8, 9, 14, 16, 18, 20, X, and Y were affected by aberrations in subclone RP (Fig. 2c).
The obtained SKY results were complemented with copy number analysis by array CGH (Fig. 3 and Additional file 1: Table S1). Array CGH analysis identified 173 regions with aberrant copy number status from which 78, 111 and 132 regions were affected by DNA gains or DNA losses in the CAL-33 parental cells, SP, or RP subclones, respectively (Fig. 3 and Additional file 1: Table S1 and Additional file 2: Table S2). 68 copy number alterations (for SP) and 85 copy number alterations (for RP) were different from the parental CAL-33 cell line. Cytogenetic studies of both clones showed distinct genomic changes in comparison to the parental cell line indicating genomic key alterations for irradiation-related phenotypes on chromosomes 1, 2, 3, 4, 5, 8, 11, 14, 16 and 21. In addition, recurrent CAL-33-specific alterations on chromosomes 3, 7, 18, X and Y were observed showing the authenticity of the newly generated cell lines.

Analysis of proliferation rates and cell cycle distribution

Proliferation rate and cell cycle distribution are important factors, which can affect radiosensitivity and/or resistance. Accordingly, we performed proliferation and cell cycle analyses. In comparison to CAL-33 parental cells, both subclones displayed prolonged doubling times (29 h for subclone SP, 30 h for subclone RP, and 24 h for the parental cell line, Additional file 3: Figure S1, A). This might be due to the observation that under exponential growth conditions, the percentage of mitotic cells in both subclones was decreased (2.1 % for subclone SP, 1.5 % for subclone RP, and 2.9 % for the parental cells), but fails to explain the differences in radiosensitivity (Additional file 3: Figure S1, B). With regard to irradiation-induced G2-arrest, the sensitive subclone SP revealed virtually identical dynamics as the parental CAL-33 cells, whereas in the resistant subclone RP G2-arrest was initially delayed, but also reached its maximum around 12 h after irradiation, and afterwards appeared to be prolonged (Additional file 3: Figure S1, C). In fact, prolonged cell cycle arrest can contribute to radioresistance as cells have more time to repair irradiation-induced DNA damage. However, extended cell cycle arrest can also be indicative for delayed DNA repair. In order to address the mechanisms underlying radioresistance on a molecular level, next we therefore performed unbiased transcriptome analyses of the CAL-33 subclones.

mRNA gene expression analysis of the CAL-33 subclones

Microarray analyses allowed the identification of differences in basal gene expression levels between the derived subclones and the parental CAL-33 cell line. We identified 523 and 1292 differentially expressed genes (FDR < 0.05) for clones SP and RP, respectively, whereas 361 of the genes were overlapping (Additional file 4: Table S3). It is interesting to note that the RP clone exhibited more pronounced transcriptional differences than the SP clone.
Eight of the differentially expressed genes (SP and/or RP clone versus the parental CAL-33 cell line) were arbitrarily chosen for technical validation of the microarray data. Correlation analysis between qRT-PCR and microarray data showed a strong correlation for seven out of eight validated genes (Additional file 5: Table S4). The microarray and qRT-PCR derived fold-changes were in a good agreement.
Subsequently, the differentially expressed genes were subjected to pathway enrichment analyses in order to determine pathways common or specific to the radioresistant or radiosensitive phenotype. In total, 65 and 455 pathways were significantly enriched (FDR < 0.1) for the SP and RP clone, respectively (Additional file 6: Table S5). The top 100 identified pathways ordered according to the highest matching (percentage of differentially expressed genes of all genes in a pathway) were grouped into major pathways (Table 1).
Table 1
Significantly enriched pathways of genes differentially expressed in subclones SP and RP compared to the parental CAL-33 cells
CAL-33 SP vs parental
Common signalling pathways
CAL-33 RP vs parental
Transmembrane transport of small molecules
Signaling by VEGF
Nonhomologous end-joining (NHEJ)
GPCR ligand binding
Extracellular matrix organization
Homologous DNA pairing and strand exchange
Signaling by Rho GTPases
RNA polymerase III transcription initiation
CD28 dependent PI3K/Akt signaling
 
Interferon signaling
NOTCH1 intracellular domain regulates transcription
 
Signaling by interleukins
 
 
Senescence-associated secretory phenotype (SASP)
 
 
NOD1/2 signaling pathway
 
 
TNFR1-induced NFkB signaling pathway
 
 
Toll-like receptors cascades
 
 
Death receptor signaling
 
 
MAPK1/MAPK3 signaling
 
Pathways common (middle) and specific to the radiosensitive (left) or radioresistant (right) phenotype are shown. The corresponding genes and their direction of regulation (up/down) are listed in Additional file 4: Table S3 and Additional file 6: Table S5
This resulted in a set of commonly deregulated pathways compared to the parental cell line but not specific for a particular phenotype of radiation sensitivity. Moreover, pathways specific to the radiosensitive or radioresistant phenotype were identified. These comprised mainly pathways that are known to be affected by ionizing irradiation in HNSCC [3638].

Integration of copy number changes with differentially expressed genes

To verify whether the aberrant expression of genes in the SP and RP subclones can be explained by the observed copy number changes, integration of genomic (array CGH data) and transcriptomic data was performed. For subclone SP, we identified 18 genes with DNA gains being up-regulated and 31 genes with DNA losses being down-regulated, whereas for subclone RP, 44 up-regulated genes showed copy number gains and 5 genes with copy number losses were down-regulated (Table 2).
Table 2
Integration of differentially expressed genes with array CGH data
CAL-33 SP (#303)
CAL-33 RP (#327)
Gene name
FC
 
Gene name
FC
 
PTGS1
16.417
gain
PTGS1
20.52
gain
TLR4
10.333
IL7R
10.643
SLC2A6
7.167
SLC2A6
7.13
TNC
5.478
SLC12A7
6.931
PHF11
4.886
COL5A1
6.608
PAPPA
4.594
CERCAM
6.381
MX2
4.457
PDZD2
5.726
LHFP
4.442
PTGER4
5.05
RGCC
4.285
STXBP1
4.377
GTF2F2
3.767
INPP5E
3.797
BACE2
3.755
NKD2
3.785
NEK3
3.564
SLC1A3
3.784
MSANTD3
3.198
RICTOR
3.675
FNDC3A
2.971
PTGES
3.409
RC3H2
2.77
MVB12B
3.381
INPP5E
2.584
CDK5RAP2
3.362
RPL7A
2.543
PRRC2B
3.155
UFM1
2.485
SEC16A
2.948
SLC25A29
0.263
loss
RC3H2
2.855
RCOR1
0.297
USP20
2.714
HLCS
0.305
RPL7A
2.59
CCDC85C
0.318
CARD9
2.572
IPO5
0.322
TOR1B
2.529
WRB
0.326
TRAF1
2.524
PIGP
0.329
QSOX2
2.492
CDCA4
0.36
TLR4
2.473
ZBTB42
0.361
UGCG
2.463
EVA1C
0.37
ZBTB43
2.396
BTBD6
0.37
C9orf9
2.327
TTC3
0.374
RAD1
2.255
CLN5
0.388
TRIM32
2.241
CCNK
0.389
PTRH1
2.221
DSC3
0.389
CEP72
2.208
PPP1R13B
0.389
DAP
2.035
DYRK1A
0.393
C9orf114
2.019
SIVA1
0.4
SDHA
2.013
BRF1
0.404
DOLPP1
1.997
IMPACT
0.41
RALGPS1
1.996
IFNGR2
0.416
SURF1
1.958
TIAM1
0.423
MTRR
1.855
PCCA
0.426
C5orf42
1.849
EML1
0.432
ANKRD33B
0.609
HMGN1
0.439
OR4C6
0.576
SETD3
0.441
NPR3
0.43
OSBPL1A
0.462
GOLGA6L6
0.505
loss
IFNAR2
0.54
ZNF480
0.532
LAMA3
2.1
NBPF10
1.909
CRIP1
2.463
TPTE
2.342
CDH2
5.373
NBPF9
2.945
Detailed information on identified genes, their CNA status and corresponding fold changes are presented
This integrative data analysis of DNA copy number and gene expression followed by pathway enrichment analysis allowed us to identify related pathways encompassing signaling by Rho GTPases as one of the deregulated pathways in the SP subclone. At the same time we observed DNA loss and downregulation of the TIAM1 gene that belongs to the Rho GTPases signaling pathway. Yang et al. recently showed that high expression of TIAM1 is associated with poor clinical outcome in patients with HNSCC [39]. Similarly, for the RP subclone a gain and upregulation of RAD1 and RICTOR genes was observed which is in accordance with the deregulation of the homologous recombination and PI3K signaling pathways in HNSCC as previously described in [40]. For both of those clones a gain and upregulation of the TLR4 gene and the deregulation of the toll-like kinases signaling pathway including upregulation of the genes PLCG2, TAB3, RPS6KA2 has been detected. Even though contradictory reports exist, the overexpression of TLR4 and activation of related pathway has been described to promote HNSCC tumor development and to ensure tumor protection from the immune system [41].

Time-dependent gene expression in response to ionizing radiation in CAL-33 clones

It was hypothesized that CAL-33 subclones with different phenotypes of radiosensitivity also show differences in gene expression profiles in response to irradiation. Therefore, we performed differential time-course microarray analyses between irradiated (8 Gy) and sham irradiated control cells. 7299, 6980, and 8111 genes were differentially expressed in the CAL-33 parental, SP, and RP subclones, respectively (Table 3). Although the number of differentially expressed genes after irradiation with 8 Gy was comparable for all cell lines studied, approximately 50 % of the affected genes were not identical. More than 1000 genes were exclusively involved in the radiation response of each of the CAL-33 cell clones (Fig. 4). The entirety of differentially expressed genes in response to ionizing radiation is listed and compared for all CAL-33 cell lines in Additional file 7: Table S6.
Table 3
Comparison of detected and differentially expressed genes after irradiation for analyzed cell sublines
CAL-33 (8 Gy vs sham-irradiated)
Parental cell line
Subclone SP
Subclone RP
Total number of detected genes
12529
12259
12714
Number of differentially expressed genes
7299
6980
8111
Number of genes in the network
6256
5709
6859
5 % top genes
313
285
343
To identify common and separate pathways of the radiation response, pathway enrichment analyses were performed on early and late responding genes, respectively (Additional file 8: Table S7). The most interesting pathways with regard to their known role in radiation response were grouped in to major pathways (Table 4).
Table 4
Significantly enriched pathways of early and late responding genes after ionizing radiation
 
Subclone SP
CAL-33 parental
Subclone RP
Early and late responding pathways
Apoptosis
Cellular senescence
Cell cycle
Cellular responses to stress
Signaling by Wnt
Signaling by Rho GTPases
Early responding pathways
DNA double-strand break repair
Signaling by TGF-beta receptor complex
Interferon signaling
Cellular response to hypoxia
TRAF6 mediated NF-kB activation
  
Late responding pathways
Signaling by TGF-beta receptor complex
TRAF6 mediated NF-kB activation
Interferon signaling
Toll-Like receptors cascades
Signaling by interleukins
Extracellular matrix organization
MAPK family signaling cascades
NOD1/2 signaling pathway
PI3K/AKT signaling in cancer
Signaling by VEGF
Signaling by EGFR
Signaling by FGFR
Signaling by NOTCH
Signaling by ERBB2
Signaling by ERBB4
GPCR ligand binding
Base excision repair
GPCR ligand binding
It is interesting to note that all cell lines share a set of deregulated pathways (Table 4). In addition, for each of the radiosensitivity phenotypes (parental, SP, RP) specific pathways were observed. The early responses in gene expression include interferon signaling and in particular for the resistant phenotype the cellular response to hypoxia which is known for a long time to have a crucial role in radioresistance [42, 43]. Also, some other pathways from an early gene expression response that are involved in DNA repair, cell cycle, cellular response to stress and apoptosis impact on survival after irradiation and therefore contribute to radioresistance [9, 4446].
Deregulated pathways related to late responding genes include EGFR- and PI3K/AKT signaling that were already linked to poor clinical outcome and therapy response in HNSCC [4749] in association with ERBB2, ERBB3 and ERBB4 co-expression [5052]. Interestingly, also involvement of ERBB2 and EERB4 receptor signaling was observed in all cell lines as a late response to ionizing radiation. Further late responding pathways include toll-like receptor cascades, interleukin signaling, NF-kB activation and interferon signaling all of which are frequently detected after treatment with ionizing radiation [53, 54]. However, the role of interleukin signaling and immune response is largely unknown so far. This also applies to cellular senescence and the impact of senescence pathways on radioresistance that were also discovered as late responding pathways in our CAL-33 subclones. Although evidence exists that senescence might be associated with the disruption of the tissue microenvironment leading to the secretion of senescence-associated pro-inflammatory factors and to the development of a pro-oncogenic environment [34] its role in radioresistance in rather unclear so far.

Gene association network reconstruction and network analysis

To go beyond pathway enrichment analyses of differentially expressed genes, gene association networks were reconstructed. Parameters of the obtained networks (provided as igraph R-objects in Additional file 9: File S1) for all three CAL-33 cell lines are presented in Table 3. Subsequently, the combined topological centrality measure was used to characterize the biological importance of genes in the reconstructed association networks. We identified nodes (genes) that are likely to control the network by combining three network centrality measures: degree, closeness and shortest path betweenness [32, 55]. The 5 % of the highest ranked genes (listed in Additional file 10: Table S8) were mapped to the Reactome pathways to further evaluate their biological roles. The top ten pathways according to the FDR values are listed in Table 5. All identified pathways are listed in Additional file 11: Table S9. Two of the detected pathways, signaling by Rho GTPases and signaling by Wnt, were in common for all three subclones exposed to irradiation (Table 5). However, most of the pathways were different for the individual subclones. For the parental cell line, we additionally detected pathways associated with cellular response to stress, signaling by EGFR, and cytochrome P450. The most important pathways in the SP subclone were associated with axon guidance, chromatin organization and semaphorin, whereas for the RP subclone the senescence-associated secretory phenotype (SASP) together with GPCR ligand binding were considered as crucial. The co-occurrence of the SASP and GPCR signaling pathways, that are known to be connected [56], indicate that the identification of those pathways might not be accidental and suggest a high importance for the radiation resistant phenotype. In future experiments more HNSCC cell lines should be analyzed.
Table 5
The top pathways after mapping of 5 % highest ranked genes from the reconstructed gene association networks to the Reactome pathways
 
Pathway
Genes
CAL-33 parental
Generic transcription pathway
CCNC, NR4A3, ZNF248, ZNF302, ZNF350, ZNF417, ZNF431, ZNF543, ZNF621, ZNF710, ZNF735
Cellular responses to stress
BAG4, CDKN2D, DEDD2, GABARAPL2, HSPA1A, MAPK10, RBX1
Diseases of signal transduction
CBL, CCNC, CTBP2, FOXO4, RBX1, TGFB1
EPH-ephrin mediated repulsion of cells
CLTCL1, EFNA5, SRC
Neurotransmitter release cycle
CHAT, SNAP25, STXBP1
O-linked glycosylation
ADAMTSL5, CFP, ST3GAL3, THBS2
Signaling by EGFR
CBL, FOXO4, NF1, PAG1, RBX1, SPRY1, SRC
Signaling by Wnt
CTBP2, DAAM1, FRAT1, RAC2, RBBP5, RBX1, SOX3
Signaling by Rho GTPases
ABR, CENPA, DAAM1, PKN3, RAC2, RANGAP1, SRC
Cytochrome P450 - arranged by substrate type
CYP17A1, CYP3A4, CYP4A11
SP subclone
Axon guidance
APH1A, DPYSL3, HSP90AB1, ITGA9, LAMTOR2, NRG1, PLXNA4, PPP2CA, PSPN, RAC1, RDX, RGMB
Generic transcription pathway
AKT2, LAMTOR2, MED26, TBL1XR1, ZNF302, ZNF394, ZNF431, ZNF561, ZNF680, ZNF691, ZNF750, ZNF774
Cell cycle
CDKN2D, EP300, MASTL, MAU2, MCM4, NUPL2, PPP2CA, RAD21, RANGAP1, SKA2, SYCP1
Chromatin organization
ATXN7, EP300, HIST3H2A, KDM4D, KDM5D, SUPT20H, TBL1XR1
Diseases of signal transduction
AKT2, APH1A, BCR, CTBP2, EP300, NRG1, PPP2CA, TBL1XR1
Signaling by Wnt
AKT2, CCDC88C, CTBP2, EP300, PLCB1, PPP2CA, RAC1, RNF146
Semaphorin interactions
DPYSL3, HSP90AB1, PLXNA4, RAC1
Glycolysis
ALDOC, GCK, PPP2CA
Regulation of beta-cell development
AKT2, GCK, IAPP
RHO GTPases activate WASPs and WAVEs
NCKIPSD, RAC1, WAS
RP subclone
Signaling by Rho GTPases
ABR, ACTB, ARHGAP35, ARHGEF7, BCR, HIST1H2BJ, HIST1H2BL, HIST2H2BE, INCENP, ITSN1, NDE1, OBSCN, RHOT1, SRGAP1, WAS
Cell Cycle
ANAPC11, BLM, CDKN1A, CDKN2D, FZR1, HIST1H2BJ, HIST1H2BL, HIST2H2BE, INCENP, KIF23, MAU2, NDE1, NUP62, POM121, PSMC3IP, TK2, WHSC1
Senescence-associated secretory phenotype (SASP)
ANAPC11, CDKN1A, CDKN2D, FZR1, HIST1H2BJ, HIST1H2BL, HIST2H2BE
GPCR ligand binding
CCL19, GPR132, MC1R, MLN, OPN1SW, OPRL1, PTCH2, PTGDR, PTGER1, TAS2R14, TAS2R19, TAS2R45, TBXA2R, WNT10B
Transcriptional regulation by small RNAs
HIST1H2BJ, HIST1H2BL, HIST2H2BE, NUP62, POM121
G alpha (12/13) signaling events
ABR, ARHGEF7, ITSN1, OBSCN, TBXA2R
Post-translational protein modification
ADAMTS19, ARSB, ARSG, BLM, CFP, CNIH1, CNIH2, GALNT10, NUP62, POM121, ST3GAL3
DNA repair
ACTB, BLM, CHD1L, DTL, ERCC6, HIST1H2BJ, HIST1H2BL, HIST2H2BE, WHSC1
Signaling by Wnt
HIST1H2BJ, HIST1H2BL, HIST2H2BE, NFATC1, PLCB1, SOX3, TCF7L2, TMED5, WNT10B
Assembly of the primary cilium
BBS10, CC2D2A, NDE1, NPHP1, PDE6D, TCTEX1D2, TTC30B

Expression of endogenous retrovirus and related pathways

The analysis of differentially expressed genes between the RP and SP subclones and the parental cell line revealed the ERVMER34-1 gene as differentially expressed in both clones. Approximately 8 % of the human genome consists of endogenous retroviruses (ERVs) that have been derived from exogenous retroviruses following infection and DNA integration into germ line cells [57, 58]. Although most of the ERV sequences have defective structures, some of ERV genes still have an open reading frame (ORF) and protein expression [59]. ERV genes can promote homologous and non-homologous recombination and therefore, may introduce new mutations [60, 61]. Furthermore, ERVs may lead to genome instability, and contribute to tumor initiation and progression [62]. The expression of ERV genes has been demonstrated in various cancers, including breast, ovarian, prostate and melanoma [6366]. The differential gene expression analyses of the subclones in comparison to the parental CAL-33 cells revealed ERVMER34-1 gene as differentially expressed in both derived clones (Additional file 4: Table S3). Subsequently, we tested whether any of the retroviral genes were differentially expressed in response to ionizing radiation. Apart from ERVMER34-1, we were able to identify another endogenous retroviral gene, ERV3-1, that was temporally differentially expressed following radiation. The time dependent expression of those genes suggests ERV3-1 expression to be associated with the radiation response, whereas ERVMER34-1 expression exhibits rather random fluctuations (Additional file 12: Figure S2 and Additional file 13: Figure S3). Thus, the ERV3-1 expression clearly implies a possible association with the radiation exposure. The radiation-associated upregulation of ERV3-1 is demonstrated for all subclones starting from the second day of irradiation. To our knowledge, the expression of ERV3-1 following radiation and its influence on radiation resistance has not been addressed in detail so far. A recent study by Lee et al. [67] has demonstrated an increase in the expression of ERV3-1 (HERV-R) env related to a fractionated exposure to γ-radiation in radioresistant A549 lung cancer cells but not in less radioresistant H460 cells. The presented results raise the question whether overexpression of ERV3-1 might be involved in the radiation response of HNSCC cells. To gain knowledge about the potential gene interactions with the ERV3-1 gene we used the gene association networks reconstructed for all CAL-33 subclones and extracted the putative direct or indirect ERV3-1 interaction partners resulting in the first neighborhood genes of ERV3-1 differ between the three analyzed cell lines (Fig. 5). The largest first neighborhood gene association network can be observed for the RP subclone where 29 genes are linked to ERV3-1. For the CAL-33 parental cell line and the SP subclone the first neighborhood gene association network consist only of three (OR2A2, U2AF1L4, C11orf94) and one (FEEH2) potential association partners, respectively. The considerably larger first neighborhood of the ERV3-1 gene for the RP cells suggests a more important role of this gene for acquired radiation resistance. In addition, a Reactome pathway enrichment analysis revealed that the first neighborhood genes of the ERV3-1 gene in RP cells were associated with GPCR signaling (DRD4, OPN1MW, TBXA2R), transmembrane transport of small molecules (ATP1B2, AZGP1, SLC22A17), generic transcription pathway (ZNF419, ZNF550, ZNF782), signaling by Rho GTPases (NCKIPSD), and cell cycle (MAX). However, to our knowledge, the interaction partners of the ERV3-1 gene have not been studied in detail so far, which makes an interpretation difficult and highly speculative at this time. Also further studies have to be performed in order to validate the gene associations with ERV3-1independently.

Conclusion

In conclusion, the present study presents comprehensive gene expression data of CAL-33 subclones of different radiosensitivity. Based on these data networks have been identified that are linked to the radiation response phenotypes. The pathways associated with the resistant phenotype are of special interest focusing on the senescence-associated secretory phenotype (SASP) together and GPCR ligand binding. Also, the radiation-associated expression of the endogenous retrovirus ERV3-1 appears highly attractive for further studies on the molecular mechanisms of acquired radioresistance.

Abbreviations

CGH, comparative genomic hybridization; CNA, copy number aberration; ERV endogenous retrovirus; FDR, false discovery rate; GAN, gene association network; HNSCC, head and neck squamous cell carcinoma; RP, resistant phenotype; SKY, spectral karyotyping; SP, sensitive phenotype

Acknowledgement

We thank Aaron Selmaier, Isabella Zagorski and Laura Dajka from the Research Unit Radiation Cytogenetics for their excellent technical support.

Funding

This study was supported by the German Federal Ministry of Education and Research (ZiSS - 02NUK024B and 02NUK024C) and the Clinical Cooperation Group “Personalized Radiotherapy in Head and Neck Cancer”.

Availability of data and material

Gene expression microarray data will be available at ArrayExpress.

Authors’ contributions

AM: gene expression analyses, bioinformatics and biostatistics analysis, manuscript draft; US: generation and characterization of subclones, sample preparation for gene expression analyses; MS: bioinformatics and biostatistics analysis; HZ: spectral karyotyping; support for study design, critical revision of the manuscript; KL: conceived generation of subclones; support for experimental design/concept, critical revision of the manuscript; KU: support for study design, experimental design/concept, bioinformatics and biostatistics analysis; JH: array CGH analyses; support for study design, experimental design/concept; draft and critical revision of manuscript; All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
Not applicable.

Disclosures

Not applicable.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Anhänge

Additional files

Literatur
2.
Zurück zum Zitat Network NCC. NCCN Clinical Practice Guidelines in Oncology: head and neck cancers. Version 2. 2013. Network NCC. NCCN Clinical Practice Guidelines in Oncology: head and neck cancers. Version 2. 2013.
3.
Zurück zum Zitat Hashibe M, Brennan P, Chuang SC, Boccia S, Castellsague X, Chen C, Curado MP, Dal Maso L, Daudt AW, Fabianova E, et al. Interaction between tobacco and alcohol use and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Cancer Epidemiol Biomarkers Prev. 2009;8(2):541–50.CrossRef Hashibe M, Brennan P, Chuang SC, Boccia S, Castellsague X, Chen C, Curado MP, Dal Maso L, Daudt AW, Fabianova E, et al. Interaction between tobacco and alcohol use and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Cancer Epidemiol Biomarkers Prev. 2009;8(2):541–50.CrossRef
4.
Zurück zum Zitat Guha N, Boffetta P, Wünsch Filho V, Eluf Neto J, Shangina O, Zaridze D, Curado MP, Koifman S, Matos E, Menezes A, et al. Oral health and risk of squamous cell carcinoma of the head and neck and esophagus: results of two multicentric case–control studies. Am J Epidemiol. 2007;66(10):1159–73.CrossRef Guha N, Boffetta P, Wünsch Filho V, Eluf Neto J, Shangina O, Zaridze D, Curado MP, Koifman S, Matos E, Menezes A, et al. Oral health and risk of squamous cell carcinoma of the head and neck and esophagus: results of two multicentric case–control studies. Am J Epidemiol. 2007;66(10):1159–73.CrossRef
5.
Zurück zum Zitat Pannone G, Santoro A, Papagerakis S, Lo Muzio L, De Rosa G, Bufo P. The role of human papillomavirus in the pathogenesis of head & neck squamous cell carcinoma: an overview. Infect Agents Cancer. 2011;6:4.CrossRefPubMedPubMedCentral Pannone G, Santoro A, Papagerakis S, Lo Muzio L, De Rosa G, Bufo P. The role of human papillomavirus in the pathogenesis of head & neck squamous cell carcinoma: an overview. Infect Agents Cancer. 2011;6:4.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Seiwert TY, Salama JK, Vokes EE. The chemoradiation paradigm in head and neck cancer. Nat Clin Pract Oncol. 2007;4(3):156–71.CrossRefPubMed Seiwert TY, Salama JK, Vokes EE. The chemoradiation paradigm in head and neck cancer. Nat Clin Pract Oncol. 2007;4(3):156–71.CrossRefPubMed
7.
Zurück zum Zitat Bar-Ad V, Palmer J, Yang H, Cognetti D, Curry J, Luginbuhl A, Tuluc M, Campling B, Axelrod R. Current management of locally advanced head and neck cancer: the combination of chemotherapy with locoregional treatments. Semin Oncol. 2014;41(6):798–806.CrossRefPubMed Bar-Ad V, Palmer J, Yang H, Cognetti D, Curry J, Luginbuhl A, Tuluc M, Campling B, Axelrod R. Current management of locally advanced head and neck cancer: the combination of chemotherapy with locoregional treatments. Semin Oncol. 2014;41(6):798–806.CrossRefPubMed
8.
Zurück zum Zitat Weichselbaum RR, Beckett MA, Schwartz JL, Dritschilo A. Radioresistant tumor cells are present in head and neck carcinomas that recur after radiotherapy. Int J Radiat Oncol Biol Phys. 1988;15(3):575–9.CrossRefPubMed Weichselbaum RR, Beckett MA, Schwartz JL, Dritschilo A. Radioresistant tumor cells are present in head and neck carcinomas that recur after radiotherapy. Int J Radiat Oncol Biol Phys. 1988;15(3):575–9.CrossRefPubMed
9.
Zurück zum Zitat Perri F, Pacelli R, Della Vittoria Scarpati G, Cella L, Giuliano M, Caponigro F, Pepe S: Radioresistance in head and neck squamous cell carcinoma: biological bases and therapeutic implications. Head Neck. 2015;37(5):763–70.CrossRefPubMed Perri F, Pacelli R, Della Vittoria Scarpati G, Cella L, Giuliano M, Caponigro F, Pepe S: Radioresistance in head and neck squamous cell carcinoma: biological bases and therapeutic implications. Head Neck. 2015;37(5):763–70.CrossRefPubMed
10.
Zurück zum Zitat Gioanni J, Fischel JL, Lambert JC, Demard F, Mazeau C, Zanghellini E, Ettore F, Formento P, Chauvel P, Lalanne CM, et al. Two new human tumor cell lines derived from squamous cell carcinomas of the tongue: Establishment, characterization and response to cytotoxic treatment. Eur J Cancer Clin Oncol. 1988;24(9):1445–55.CrossRefPubMed Gioanni J, Fischel JL, Lambert JC, Demard F, Mazeau C, Zanghellini E, Ettore F, Formento P, Chauvel P, Lalanne CM, et al. Two new human tumor cell lines derived from squamous cell carcinomas of the tongue: Establishment, characterization and response to cytotoxic treatment. Eur J Cancer Clin Oncol. 1988;24(9):1445–55.CrossRefPubMed
11.
Zurück zum Zitat Ernst A, Anders H, Kapfhammer H, Orth M, Hennel R, Seidl K, Winssinger N, Belka C, Unkel S, Lauber K. HSP90 inhibition as a means of radiosensitizing resistant, aggressive soft tissue sarcomas. Cancer Lett. 2015;365(2):211–22.CrossRefPubMed Ernst A, Anders H, Kapfhammer H, Orth M, Hennel R, Seidl K, Winssinger N, Belka C, Unkel S, Lauber K. HSP90 inhibition as a means of radiosensitizing resistant, aggressive soft tissue sarcomas. Cancer Lett. 2015;365(2):211–22.CrossRefPubMed
13.
Zurück zum Zitat Kinzel L, Ernst A, Orth M, Albrecht V, Hennel R, Brix N, Frey B, Gaipl US, Zuchtriegel G, Reichel CA, et al. A novel HSP90 inhibitor with reduced hepatotoxicity synergizes with radiotherapy to induce apoptosis, abrogate clonogenic survival, and improve tumor control in models of colorectal cancer. Oncotarget. 2016. doi:10.18632/oncotarget.9774.PubMed Kinzel L, Ernst A, Orth M, Albrecht V, Hennel R, Brix N, Frey B, Gaipl US, Zuchtriegel G, Reichel CA, et al. A novel HSP90 inhibitor with reduced hepatotoxicity synergizes with radiotherapy to induce apoptosis, abrogate clonogenic survival, and improve tumor control in models of colorectal cancer. Oncotarget. 2016. doi:10.​18632/​oncotarget.​9774.PubMed
14.
Zurück zum Zitat Hieber L, Huber R, Bauer V, Schäffner Q, Braselmann H, Thomas G, Bogdanova T, Zitzelsberger H. Chromosomal rearrangements in post-Chernobyl papillary thyroid carcinomas: evaluation by spectral karyotyping and automated interphase FISH. J Biomed Biotechnol. 2011;2011:693691.CrossRefPubMedPubMedCentral Hieber L, Huber R, Bauer V, Schäffner Q, Braselmann H, Thomas G, Bogdanova T, Zitzelsberger H. Chromosomal rearrangements in post-Chernobyl papillary thyroid carcinomas: evaluation by spectral karyotyping and automated interphase FISH. J Biomed Biotechnol. 2011;2011:693691.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Neuvial P, Hupe P. MANOR: CGH Micro-Array NORmalization. R package version 1.42.0. Neuvial P, Hupe P. MANOR: CGH Micro-Array NORmalization. R package version 1.42.0.
16.
Zurück zum Zitat Neuvial P, Hupe P, Brito I, Liva S, Manie E, Brennetot C, Radvanyi F, Aurias A, Barillot E. Spatial normalization of array-CGH data. BMC Bioinformatics. 2006;7(264):1–20. Neuvial P, Hupe P, Brito I, Liva S, Manie E, Brennetot C, Radvanyi F, Aurias A, Barillot E. Spatial normalization of array-CGH data. BMC Bioinformatics. 2006;7(264):1–20.
17.
Zurück zum Zitat van de Wiel MA, Brosens R, Eilers PHC, Kumps C, Meijer GA, Menten B, Sistermans E, Speleman F, Timmerman ME, Ylstra B. Smoothing waves in array CGH tumor profiles. Bioinformatics. 2009;25(9):1099–104.CrossRefPubMed van de Wiel MA, Brosens R, Eilers PHC, Kumps C, Meijer GA, Menten B, Sistermans E, Speleman F, Timmerman ME, Ylstra B. Smoothing waves in array CGH tumor profiles. Bioinformatics. 2009;25(9):1099–104.CrossRefPubMed
18.
Zurück zum Zitat Seshan VE, Olshen A. DNAcopy: DNA copy number data analysis. R package version 1.44.0. Seshan VE, Olshen A. DNAcopy: DNA copy number data analysis. R package version 1.44.0.
19.
Zurück zum Zitat Olshen AB, Venkatraman ES, Lucito R, Wigler M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics. 2004;5(4):557–72.CrossRefPubMed Olshen AB, Venkatraman ES, Lucito R, Wigler M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics. 2004;5(4):557–72.CrossRefPubMed
20.
Zurück zum Zitat van de Wiel MA, Kim KI, Vosse SJ, van Wieringen WN, Wilting SM, Ylstra B. CGHcall: calling aberrations for array CGH tumor profiles. Bioinformatics. 2007;23(7):892–4.CrossRefPubMed van de Wiel MA, Kim KI, Vosse SJ, van Wieringen WN, Wilting SM, Ylstra B. CGHcall: calling aberrations for array CGH tumor profiles. Bioinformatics. 2007;23(7):892–4.CrossRefPubMed
21.
Zurück zum Zitat van de Wiel MA, van Wieringen WN. CGHregions: dimension reduction for array CGH data with minimal information loss. Cancer Informatics. 2007;3:55–63.PubMedPubMedCentral van de Wiel MA, van Wieringen WN. CGHregions: dimension reduction for array CGH data with minimal information loss. Cancer Informatics. 2007;3:55–63.PubMedPubMedCentral
22.
Zurück zum Zitat R Core Team: R: A language and environment for statistical computing 2013. R Core Team: R: A language and environment for statistical computing 2013.
23.
Zurück zum Zitat Smyth GK. Limma: linear models for microarray data. In: Bioinformatics and Computational Biology Solutions Using {R} and Bioconductor. New York: Springer; 2005. p. 397–420.CrossRef Smyth GK. Limma: linear models for microarray data. In: Bioinformatics and Computational Biology Solutions Using {R} and Bioconductor. New York: Springer; 2005. p. 397–420.CrossRef
24.
Zurück zum Zitat Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5(10):R80.CrossRefPubMedPubMedCentral Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5(10):R80.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Lopez-Romero P: Agi4x44PreProcess: PreProcessing of Agilent 4x44 array data. R package version 1.16.0. Lopez-Romero P: Agi4x44PreProcess: PreProcessing of Agilent 4x44 array data. R package version 1.16.0.
26.
Zurück zum Zitat Li Q, Birkbak NJ, Gyorffy B, Szallasi Z, Eklund AC. Jetset: selecting the optimal microarray probe set to represent a gene. BMC Bioinformatics. 2011;12:474.CrossRefPubMedPubMedCentral Li Q, Birkbak NJ, Gyorffy B, Szallasi Z, Eklund AC. Jetset: selecting the optimal microarray probe set to represent a gene. BMC Bioinformatics. 2011;12:474.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Michna A. splineTimeR: Time-course differential gene expression data analysis using spline regression models followed by gene association network reconstruction. R package version 0995 2015. Michna A. splineTimeR: Time-course differential gene expression data analysis using spline regression models followed by gene association network reconstruction. R package version 0995 2015.
28.
Zurück zum Zitat Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Royal Stat Soc. 1995;57:289–300. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Royal Stat Soc. 1995;57:289–300.
29.
Zurück zum Zitat Opgen-Rhein R, Strimmer K. Using regularized dynamic correlation to infer gene dependency networks from time-series microarray data. The 4th International Workshop on Computational Systems Biology, WCSB 2006. Opgen-Rhein R, Strimmer K. Using regularized dynamic correlation to infer gene dependency networks from time-series microarray data. The 4th International Workshop on Computational Systems Biology, WCSB 2006.
30.
Zurück zum Zitat Opgen-Rhein R, Strimmer K. From correlation to causation networks: a simple approximate learning algorithm and its application to high-dimensional plant gene expression data. BMC Syst Biol. 2007;1:37.CrossRefPubMedPubMedCentral Opgen-Rhein R, Strimmer K. From correlation to causation networks: a simple approximate learning algorithm and its application to high-dimensional plant gene expression data. BMC Syst Biol. 2007;1:37.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Koschützki D, Schreiber F. Centrality analysis methods for biological networks and their application to gene regulatory networks. Gene Regulation Syst Biol. 2008;2:193–201. Koschützki D, Schreiber F. Centrality analysis methods for biological networks and their application to gene regulatory networks. Gene Regulation Syst Biol. 2008;2:193–201.
32.
Zurück zum Zitat Abbasi A, Hossain L. Hybrid Centrality Measures for Binary and Weighted Networks. In: Menezes R, Evsukoff A, González MC, editors. Complex Networks. Volume 424, edn. Berlin: Springer; 2013. p. 1–7. Abbasi A, Hossain L. Hybrid Centrality Measures for Binary and Weighted Networks. In: Menezes R, Evsukoff A, González MC, editors. Complex Networks. Volume 424, edn. Berlin: Springer; 2013. p. 1–7.
33.
Zurück zum Zitat Matthews L, Gopinath G, Gillespie M, Caudy M, Croft D, de Bono B, Garapati P, Hemish J, Hermjakob H, Jassal B, et al. Reactome knowledgebase of human biological pathways and processes. Nucleic Acids Res. 2009;37(Database issue):D619–22.CrossRefPubMed Matthews L, Gopinath G, Gillespie M, Caudy M, Croft D, de Bono B, Garapati P, Hemish J, Hermjakob H, Jassal B, et al. Reactome knowledgebase of human biological pathways and processes. Nucleic Acids Res. 2009;37(Database issue):D619–22.CrossRefPubMed
35.
Zurück zum Zitat Wang J, Hu L, Gupta N, Shamseldin T, Ozawa T, Klem J, Cardell M, Deen DF. Induction and characterization of human glioma clones with different radiosensitivities. Neoplasia. 1999;1(2):138–44.CrossRefPubMedPubMedCentral Wang J, Hu L, Gupta N, Shamseldin T, Ozawa T, Klem J, Cardell M, Deen DF. Induction and characterization of human glioma clones with different radiosensitivities. Neoplasia. 1999;1(2):138–44.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Bentzen SM, Atasoy BM, Daley FM, Dische S, Richman PI, Saunders MI, Trott KR, Wilson GD. Epidermal growth factor receptor expression in pretreatment biopsies from head and neck squamous cell carcinoma as a predictive factor for a benefit from accelerated radiation therapy in a randomized controlled trial. J Clin Oncol. 2005;23(24):5560.CrossRefPubMed Bentzen SM, Atasoy BM, Daley FM, Dische S, Richman PI, Saunders MI, Trott KR, Wilson GD. Epidermal growth factor receptor expression in pretreatment biopsies from head and neck squamous cell carcinoma as a predictive factor for a benefit from accelerated radiation therapy in a randomized controlled trial. J Clin Oncol. 2005;23(24):5560.CrossRefPubMed
37.
Zurück zum Zitat Kalyankrishna S, Grandis JR. Epidermal growth factor receptor biology in head and neck cancer. J Clin Oncol. 2006;24(17):2666–72.CrossRefPubMed Kalyankrishna S, Grandis JR. Epidermal growth factor receptor biology in head and neck cancer. J Clin Oncol. 2006;24(17):2666–72.CrossRefPubMed
38.
Zurück zum Zitat Stadler ME, Patel MR, Couch ME, Hayes DN. Molecular biology of head and neck cancer: risks and pathways. Hematol Oncol Clin North Am. 2008;22(6):1099–124.CrossRefPubMedPubMedCentral Stadler ME, Patel MR, Couch ME, Hayes DN. Molecular biology of head and neck cancer: risks and pathways. Hematol Oncol Clin North Am. 2008;22(6):1099–124.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Yang H, Cai YC, Cao Y, Song M, An X, Xia Y, Wei J, Jiang WQ, Shi YX. The prognostic value of Tiam1 protein expression in head and neck squamous cell carcinoma: a retrospective study. Chin J Cancer. 2015;34(12):614–21.PubMed Yang H, Cai YC, Cao Y, Song M, An X, Xia Y, Wei J, Jiang WQ, Shi YX. The prognostic value of Tiam1 protein expression in head and neck squamous cell carcinoma: a retrospective study. Chin J Cancer. 2015;34(12):614–21.PubMed
40.
Zurück zum Zitat Lui VW, Hedberg ML, Li H, Vangara BS, Pendleton K, Zeng Y, Lu Y, Zhang Q, Du Y, Gilbert BR, et al. Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers. Cancer Discov. 2013;3(7):761–9.CrossRefPubMedPubMedCentral Lui VW, Hedberg ML, Li H, Vangara BS, Pendleton K, Zeng Y, Lu Y, Zhang Q, Du Y, Gilbert BR, et al. Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers. Cancer Discov. 2013;3(7):761–9.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Szczepanski MJ, Czystowska M, Szajnik M, Harasymczuk M, Boyiadzis M, Kruk-Zagajewska A, Szyfter W, Zeromski J, Whiteside TL. Triggering of Toll-like receptor 4 expressed on human head and neck squamous cell carcinoma promotes tumor development and protects the tumor from immune attack. Cancer Res. 2009;69(7):3105–13.CrossRefPubMedPubMedCentral Szczepanski MJ, Czystowska M, Szajnik M, Harasymczuk M, Boyiadzis M, Kruk-Zagajewska A, Szyfter W, Zeromski J, Whiteside TL. Triggering of Toll-like receptor 4 expressed on human head and neck squamous cell carcinoma promotes tumor development and protects the tumor from immune attack. Cancer Res. 2009;69(7):3105–13.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Overgaard J. Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck - a systematic review and meta-analysis. Radiother Oncol. 2011;100(1):22–32.CrossRefPubMed Overgaard J. Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck - a systematic review and meta-analysis. Radiother Oncol. 2011;100(1):22–32.CrossRefPubMed
43.
Zurück zum Zitat Li JZ, Gao W, Chan JY, Ho WK, Wong TS. Hypoxia in head and neck squamous cell carcinoma. ISRN Otolaryngol. 2012;2012:708974.PubMedPubMedCentral Li JZ, Gao W, Chan JY, Ho WK, Wong TS. Hypoxia in head and neck squamous cell carcinoma. ISRN Otolaryngol. 2012;2012:708974.PubMedPubMedCentral
44.
Zurück zum Zitat Meijer TW, Kaanders JH, Span PN, Bussink J. Targeting hypoxia, HIF-1, and tumor glucose metabolism to improve radiotherapy efficacy. Clin Cancer Res. 2012;18(20):5585–94.CrossRefPubMed Meijer TW, Kaanders JH, Span PN, Bussink J. Targeting hypoxia, HIF-1, and tumor glucose metabolism to improve radiotherapy efficacy. Clin Cancer Res. 2012;18(20):5585–94.CrossRefPubMed
45.
Zurück zum Zitat Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55(2):74–108.CrossRefPubMed Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55(2):74–108.CrossRefPubMed
46.
Zurück zum Zitat Vokes EE, Weichselbaum RR, Lippman SM, Hong WK. Head and neck cancer. N Engl J Med. 1993;328(3):184–94.CrossRefPubMed Vokes EE, Weichselbaum RR, Lippman SM, Hong WK. Head and neck cancer. N Engl J Med. 1993;328(3):184–94.CrossRefPubMed
47.
Zurück zum Zitat Sharafinski ME, Ferris RL, Ferrone S, Grandis JR. Epidermal growth factor receptor targeted therapy of squamous cell carcinoma of the head and neck. Head Neck. 2010;32(10):1412–21.CrossRefPubMedPubMedCentral Sharafinski ME, Ferris RL, Ferrone S, Grandis JR. Epidermal growth factor receptor targeted therapy of squamous cell carcinoma of the head and neck. Head Neck. 2010;32(10):1412–21.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Ang KK, Berkey BA, Tu X, Zhang HZ, Katz R, Hammond EH, Fu KK, Milas L. Impact of epidermal growth factor receptor expression on survival and pattern of relapse in patients with advanced head and neck carcinoma. Cancer Res. 2002;62(24):7350–6.PubMed Ang KK, Berkey BA, Tu X, Zhang HZ, Katz R, Hammond EH, Fu KK, Milas L. Impact of epidermal growth factor receptor expression on survival and pattern of relapse in patients with advanced head and neck carcinoma. Cancer Res. 2002;62(24):7350–6.PubMed
49.
Zurück zum Zitat Nijkamp MM, Span PN, Bussink J, Kaanders JH. Interaction of EGFR with the tumour microenvironment: implications for radiation treatment. Radiother Oncol. 2013;108(1):17–23.CrossRefPubMed Nijkamp MM, Span PN, Bussink J, Kaanders JH. Interaction of EGFR with the tumour microenvironment: implications for radiation treatment. Radiother Oncol. 2013;108(1):17–23.CrossRefPubMed
50.
Zurück zum Zitat Ibrahim SO, Vasstrand EN, Liavaag PG, Johannessen AC, Lillehaug JR. Expression of c-erbB proto-oncogene family members in squamous cell carcinoma of the head and neck. Anticancer Res. 1997;17(6D):4539–46.PubMed Ibrahim SO, Vasstrand EN, Liavaag PG, Johannessen AC, Lillehaug JR. Expression of c-erbB proto-oncogene family members in squamous cell carcinoma of the head and neck. Anticancer Res. 1997;17(6D):4539–46.PubMed
51.
Zurück zum Zitat Xia W, Lau YK, Zhang HZ, Liu AR, Li L, Kiyokawa N, Clayman GL, Katz RL, Hung MC. Strong correlation between c-erbB-2 overexpression and overall survival of patients with oral squamous cell carcinoma. Clin Cancer Res. 1997;3(1):3–9.PubMed Xia W, Lau YK, Zhang HZ, Liu AR, Li L, Kiyokawa N, Clayman GL, Katz RL, Hung MC. Strong correlation between c-erbB-2 overexpression and overall survival of patients with oral squamous cell carcinoma. Clin Cancer Res. 1997;3(1):3–9.PubMed
52.
Zurück zum Zitat Shintani S, Funayama T, Yoshihama Y, Alcalde RE, Matsumura T. Prognostic significance of ERBB3 overexpression in oral squamous cell carcinoma. Cancer Lett. 1995;95(1–2):79–83.CrossRefPubMed Shintani S, Funayama T, Yoshihama Y, Alcalde RE, Matsumura T. Prognostic significance of ERBB3 overexpression in oral squamous cell carcinoma. Cancer Lett. 1995;95(1–2):79–83.CrossRefPubMed
53.
Zurück zum Zitat Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, Gilardi MC, Bravatà V. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond). 2015;12:14.CrossRef Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, Gilardi MC, Bravatà V. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond). 2015;12:14.CrossRef
54.
Zurück zum Zitat Roses RE, Xu M, Koski GK, Czerniecki BJ. Radiation therapy and Toll-like receptor signaling: implications for the treatment of cancer. Oncogene. 2008;27(2):200–7.CrossRefPubMed Roses RE, Xu M, Koski GK, Czerniecki BJ. Radiation therapy and Toll-like receptor signaling: implications for the treatment of cancer. Oncogene. 2008;27(2):200–7.CrossRefPubMed
55.
Zurück zum Zitat Koschützki D. Network Centralities. In: Junker BH, Schreiber F, editors. Analysis of Biological Networks. Hoboken: Wiley; 2007. p. 65–84. Koschützki D. Network Centralities. In: Junker BH, Schreiber F, editors. Analysis of Biological Networks. Hoboken: Wiley; 2007. p. 65–84.
56.
Zurück zum Zitat Acosta JC, O'Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, Fumagalli M, Da Costa M, Brown C, Popov N, et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008;133(6):1006–18.CrossRefPubMed Acosta JC, O'Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, Fumagalli M, Da Costa M, Brown C, Popov N, et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008;133(6):1006–18.CrossRefPubMed
57.
Zurück zum Zitat Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, et al. Initial sequencing and analysis of the human genome. Nature. 2001;409(6822):860–921.CrossRefPubMed Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, et al. Initial sequencing and analysis of the human genome. Nature. 2001;409(6822):860–921.CrossRefPubMed
58.
Zurück zum Zitat Löwer R, Löwer J, Kurth R. The viruses in all of us: characteristics and biological significance of human endogenous retrovirus sequences. Proc Natl Acad Sci U S A. 1996;93(11):5177–84.CrossRefPubMedPubMedCentral Löwer R, Löwer J, Kurth R. The viruses in all of us: characteristics and biological significance of human endogenous retrovirus sequences. Proc Natl Acad Sci U S A. 1996;93(11):5177–84.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Villesen P, Aagaard L, Wiuf C, Pedersen FS. Identification of endogenous retroviral reading frames in the human genome. Retrovirology. 2004;1:32.CrossRefPubMedPubMedCentral Villesen P, Aagaard L, Wiuf C, Pedersen FS. Identification of endogenous retroviral reading frames in the human genome. Retrovirology. 2004;1:32.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Hughes JF, Coffin JM. Evidence for genomic rearrangements mediated by human endogenous retroviruses during primate evolution. Nat Genet. 2001;29(4):487–9.CrossRefPubMed Hughes JF, Coffin JM. Evidence for genomic rearrangements mediated by human endogenous retroviruses during primate evolution. Nat Genet. 2001;29(4):487–9.CrossRefPubMed
61.
Zurück zum Zitat Whitelaw E, Martin DI. Retrotransposons as epigenetic mediators of phenotypic variation in mammals. Nat Genet. 2001;27(4):361–5.CrossRefPubMed Whitelaw E, Martin DI. Retrotransposons as epigenetic mediators of phenotypic variation in mammals. Nat Genet. 2001;27(4):361–5.CrossRefPubMed
62.
Zurück zum Zitat Stoye JP. Studies of endogenous retroviruses reveal a continuing evolutionary saga. Nat Rev Microbiol. 2012;10(6):395–406.PubMed Stoye JP. Studies of endogenous retroviruses reveal a continuing evolutionary saga. Nat Rev Microbiol. 2012;10(6):395–406.PubMed
63.
Zurück zum Zitat Wang-Johanning F, Rycaj K, Plummer JB, Li M, Yin B, Frerich K, Garza JG, Shen J, Lin K, Yan P, et al. Immunotherapeutic potential of anti-human endogenous retrovirus-K envelope protein antibodies in targeting breast tumors. J Natl Cancer Inst. 2012;104(3):189–210.CrossRefPubMedPubMedCentral Wang-Johanning F, Rycaj K, Plummer JB, Li M, Yin B, Frerich K, Garza JG, Shen J, Lin K, Yan P, et al. Immunotherapeutic potential of anti-human endogenous retrovirus-K envelope protein antibodies in targeting breast tumors. J Natl Cancer Inst. 2012;104(3):189–210.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Wang-Johanning F, Liu J, Rycaj K, Huang M, Tsai K, Rosen DG, Chen DT, Lu DW, Barnhart KF, Johanning GL. Expression of multiple human endogenous retrovirus surface envelope proteins in ovarian cancer. Int J Cancer. 2007;120(1):81–90.CrossRefPubMed Wang-Johanning F, Liu J, Rycaj K, Huang M, Tsai K, Rosen DG, Chen DT, Lu DW, Barnhart KF, Johanning GL. Expression of multiple human endogenous retrovirus surface envelope proteins in ovarian cancer. Int J Cancer. 2007;120(1):81–90.CrossRefPubMed
65.
Zurück zum Zitat Wang-Johanning F, Frost AR, Jian B, Azerou R, Lu DW, Chen DT, Johanning GL. Detecting the expression of human endogenous retrovirus E envelope transcripts in human prostate adenocarcinoma. Cancer. 2003;98(1):187–97.CrossRefPubMed Wang-Johanning F, Frost AR, Jian B, Azerou R, Lu DW, Chen DT, Johanning GL. Detecting the expression of human endogenous retrovirus E envelope transcripts in human prostate adenocarcinoma. Cancer. 2003;98(1):187–97.CrossRefPubMed
66.
Zurück zum Zitat Li Z, Sheng T, Wan X, Liu T, Wu H, Dong J. Expression of HERV-K correlates with status of MEK-ERK and p16INK4A-CDK4 pathways in melanoma cells. Cancer Invest. 2010;28(10):1031–7.CrossRefPubMed Li Z, Sheng T, Wan X, Liu T, Wu H, Dong J. Expression of HERV-K correlates with status of MEK-ERK and p16INK4A-CDK4 pathways in melanoma cells. Cancer Invest. 2010;28(10):1031–7.CrossRefPubMed
67.
Zurück zum Zitat Lee JR, Jung YD, Kim YH, Park SJ, Huh JW, Kim HS. Effects of HERV-R env knockdown in combination with ionizing radiation on apoptosis-related gene expression in A549 lung cancer cells. Biochem Physiol. 2016;5:1. Lee JR, Jung YD, Kim YH, Park SJ, Huh JW, Kim HS. Effects of HERV-R env knockdown in combination with ionizing radiation on apoptosis-related gene expression in A549 lung cancer cells. Biochem Physiol. 2016;5:1.
Metadaten
Titel
Transcriptomic analyses of the radiation response in head and neck squamous cell carcinoma subclones with different radiation sensitivity: time-course gene expression profiles and gene association networks
verfasst von
Agata Michna
Ulrike Schötz
Martin Selmansberger
Horst Zitzelsberger
Kirsten Lauber
Kristian Unger
Julia Hess
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Radiation Oncology / Ausgabe 1/2016
Elektronische ISSN: 1748-717X
DOI
https://doi.org/10.1186/s13014-016-0672-0

Weitere Artikel der Ausgabe 1/2016

Radiation Oncology 1/2016 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.