• No results found

Transcriptome profiling of a cell line model for malignant transformation in response to moderate hypoxia

N/A
N/A
Protected

Academic year: 2022

Share "Transcriptome profiling of a cell line model for malignant transformation in response to moderate hypoxia"

Copied!
18
0
0

Loading.... (view fulltext now)

Full text

(1)

Transcriptome profiling of a cell line model for malignant transformation in response to moderate hypoxia

Frida Danielsson 1 , Erik Fasterius 2 , Kemal Sanli 1 , Cheng Zhang 1 , Adil Mardinoglu 1 , Cristina Al-Khalili 2 , Mikael Huss 3 , Mathias Uhlén 1 , Emma Lundberg 1

1. Science for Life Laboratory, KTH-Royal Institute of Technology, 171 21 Stockholm, Sweden

2. School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden 3. Science for Life Laboratory, Department of Biochemistry and

Biophysics, Stockholm University, Box 1031, SE-171 21 Solna, Sweden

Abstract

The four-step BJ model for malignant transformation enables defined steps of

tumorigenesis to be studied, and is separated into normal, immortalized, transformed and metastasizing stages 1 . In human tumor tissues, hypoxia is a commonly

observed feature, as a result of rapidly proliferating cancer cells outgrowing their

surrounding vasculature network. Transformed cancer cells are known to exhibit

phenotypic alterations, enabling continuous proliferation despite a limited oxygen

supply. Here, we used RNA-sequencing (RNA-seq) to explore differences in gene

expression across the BJ model during cultivation in atmospheric oxygen compared

to a moderate hypoxic condition (3% O 2 ). We identified 909, 1090, 1247 and 794

significantly differentially expressed genes (DEGs) due to decreased oxygen supply,

in the four stages respectively. A majority of DEGs are downregulated in moderate

hypoxia in all stages except for the SV40-transformed stage. Several genes related

to proliferation and angiogenesis are upregulated across all stages in low oxygen,

such as E2F, BRCA1 and ANGPTL4. Even though the cell lines across the BJ model

are highly similar from a global view, the introduction of SV40 Large-T renders a

distinct adaptation to the altered condition. This is mainly manifested in altered lipid

metabolism and reversed expression patterns for several genes compared to the

previous stages within the model, such as the antioxidant SOD2 and VEGFA with its

up-stream regulator CXCL8.

(2)

Introduction

The transformation of primary cells into malignant counterparts capable of forming tumors is a multistep process where changes in the genome give rise to new characteristics 2 . These include increased rate of proliferation, the ability to escape survival

restrictions, increased vascularization and rewiring of the energy

metabolism 3 . To enable the study of defined steps of tumorigenesis, an isogenic model system for malignant transformation was generated by introducing genetic changes in the human fibroblast cell line BJ, thereby establishing an important foundation for future cancer research 1 . The model was generated by ectopic expression of the telomerase catalytic subunit (hTERT) followed by stepwise additions of two oncogenes, the Simian virus 40 Large-T and the oncogenic and hyperactive version of H-ras. With these sequentially added alterations, a model consisting of four cell lines was created, starting with the primary stage followed by an

immortalized stage (active telomerase, BJ hTERT), a transformed tumor forming stage (BJ hTERT SV40) and finally a fully transformed stage where cells are capable of metastasizing (BJ

hTERT+ SV40 RASG12V). Previous analysis of the transcriptome changes within this model showed that a

majority of genes are downregulated in the course of increased malignancy, and that most of the upregulated genes are directly related to

proliferation, whereas downregulation mainly affects a more functionally diverse set of extracellular proteins.

Analysis of differential gene

expression within the model revealed a number of interesting protein

candidates, for example BDH1, ANXA1, ANPEP and ANLN whose expression were compared in tumor tissues of matching malignancy

grades, indicating that the BJ model is a highly relevant model system for the study of human tumorigenesis and proliferative capability 4 .

One of the most important elements for the life of mammalian cells is oxygen. Oxygen is involved in almost all cellular functions and during normal oxygen conditions mammalian cells generate the energy that they require for molecular processes by oxidative phosphorylation in the mitochondria.

As first demonstrated by Otto Warburg in the 1920’s and thereafter confirmed in numerous different studies,

transformed cancer cells prefer to

(3)

generate energy through glycolysis instead of oxidative phosphorylation, even under aerobic conditions and regardless of the fact that it is relatively inefficient in terms of ATP

generation 5,6 . This change in cellular energetics is today known as one of the hallmarks of cancer and solid tumors often contain areas where no oxygen is present as a result of the tumor outgrowing the capacity of its surrounding vasculatory network.

Creation of oxygen-deprived microenvironments promotes

malignant progression through clonal selection of aggressive phenotypes and subsequent poor prognosis 7 . On a molecular level, decreased oxygen levels in tumors are associated with increased hypermethylation events with consequences on gene

expression 8 . Promoter regions of genes involved in cell-cycle arrest, DNA repair and apoptosis, glycolysis, metastasis and angiogenesis have been shown to be targets for hypoxia- induced hypermethylation.

To investigate the transcriptional changes induced by decreased

oxygen supply, and how the response differ between the four isogenically matched cell lines at different degree of malignancy, we evaluated the whole

transcriptome using RNA-seq after cultivation in both the standard in vitro setup of atmospheric oxygen and at moderate hypoxia (3% O 2 ) for six passages. By comparing differential gene expression across the model, we explored how the different cell lines, despite their overall resemblance, respond differently to cultivation in 3%

oxygen. The SV40-transformation appears again 4 to induce a relatively dramatic phenotypic shift, reflected in upregulation of several genes that are downregulated in the previous stages, as well as increased lipid metabolism.

Materials & Methods

Cell cultivation

All four cell lines in the model were cultivated in Dulbecco’s modified Eagle’s Medium (Sigma-Aldrich) supplemented with 10% Fetal Bovine Serum (Sigma-Aldrich) for six

passages at 37 °C. Cultivation was performed in a humidified atmosphere containing 5% CO 2 and both under atmospheric oxygen pressure and 3%

oxygen for six passages. The cells were grown up to 80% confluence and counted with a Scepter 2.0 Cell

Counter (Merck Millipore, Billerica, MA,

(4)

USA). Cells were cultivated in

duplicate plates in parallel for each cell line and oxygen condition.

RNA sequencing

RNA was extracted from the cells using the RNeasy kit (Qiagen),

generating high quality total RNA (i.e.

RIN>8) that was used as input material for library construction with Illumina TruSeq Stranded mRNA reagents (Illumina). Duplicate samples for each cell line were sequenced on the Illumina HiSeq2500 platform.

Raw sequences were mapped to the Human reference genome GrCh37 and further quantified using the Kallisto software 9 to generate normalized Transcript Per Million (TPM) values.

TPM values for genes were generated by summing up TPM values for the corresponding transcripts generated by Kallisto. Genes with a TPM value greater than 1 were considered as expressed. Differential expression analysis was performed with the Kallisto software (v0.42.1) 9 on the raw FASTQ files with default parameters, followed by differential expression analysis using the TXimport (v1.0.3) 10 and edgeR (v3.14.0) 11 R packages.

Detailed code is available as an RMarkdown document in the supplementary data. The GRCh37

human reference genome assembly was used in all steps of the analyses.

Cell authentication

Cell line authenticity and mutational analysis was performed as previously described in 12 . Briefly, the raw RNA- seq data was aligned using the 2-pass method of the STAR (v2.5.1b) aligner, followed by de-duplication, re-

calibration and variant calling with the Genome Analysis Toolkit (GATK) Best Practices workflow (v3.5.0) 13 . The resulting variant calls were annotated using SnpEff and SnpSift (v4.2)

followed by filtering and analysis using in-house Python and R scripts 14,15 .

Metabolic profilingMetabolic profiling of the BJ model was elaborated by following a top-down analysis

approach within the broad category of metabolism in the KEGG Orthology database 16 . The program PACFM (v.

0.2) 17 was used to plot the log fold

change data between the atmospheric

and 3% oxygen treatments of the cell

lines at different hierarchy levels within

metabolism. Data was plotted by

preserving the absent functional

categories in each cancer cell line, in

order to facilitate intuitive comparison

(5)

between the different stages of the BJ model.

Results

Different model stages show varying responses to moderate hypoxia

In order to generate a complete overview of the transcriptome at the different stages in the BJ model and under atmospheric versus 3% oxygen levels, we performed RNA-seq

analysis. An overview of the isogenic model system and the experimental setup can be seen in Figure 1A. Cell authentication was performed as previously described to confirm the isogenic nature of the cells and confirm the model’s stage-specific mutations 12 . The results corroborate both the identity of the cells and the presence of mutations only in expected cell lines (see Supplementary Data). Gene

expression was estimated using the Kallisto software 9 , followed by hierarchical clustering of Spearman correlations for all expressed protein coding genes across the model (N=14,994). While there are high correlations between all the samples (Spearman correlation coefficients >

0.9, Figure 1B), there is a significant pattern within the model itself: the primary and immortalized samples cluster by model stage whereas the two transformed cell lines cluster according to oxygen level, indicating a higher degree of adaptation to the change in oxygen supply. To investigate this adaptation further, differential gene expression was analyzed with edgeR, using a cutoff of 0.01 for adjusted p-value, a fold

change of 2 and expression of 1 TPM in at least one of the samples being compared. Going from the primary cells to the immortalized cells, to the transformed cells and finally to the metastasizing cells, we identified 909, 1090, 1247 and 794 differentially expressed genes (DEGs), respectively between the two oxygen levels (Figure 2A). The largest difference is observed in the SV40-transformed cells,

indicating that this change promotes

alteration of gene expression as a

consequence of low oxygen. The

transcriptional response to oxygen

depravation is more accentuated in the

transformed tumor-forming cells (BJ

SV40) than in any of the other isogenic

cells analyzed. A majority of DEGs are

downregulated at low levels of oxygen

in all cell lines within the model, except

(6)

for the SV40-transformed cell line (Figure 2B).

Functional enrichment reveals changes in proliferation and angiogenesis as response to hypoxia

Functional enrichment analysis was performed using the Database for Annotation, Visualization and

Integrative Discovery (DAVID) tool, for upregulated and downregulated genes separately 18 . The three functional clusters with highest significance, involving upregulated genes in the primary cell line, are related to angiogenesis, proliferation and response to decrease in

oxygen/hypoxia respectively.

Interestingly, functions related to angiogenesis and proliferation are also among the top three functions

involving downregulated genes in the primary cell line (Figure 3). This reflects the generalized view provided by functional enrichment analysis, and the complexity of the genetic circuitry affected by the change in oxygen supply. The upregulated genes in the primary cell line that are known

mediators of hypoxic response include PLOD2 that is a known prognostic factor upregulated in several tumors and PDK1 that is known to be critical

in the adaptation to hypoxia, by attenuation of mitochondrial ROS production 19,20 . PDK1 is significantly upregulated in low oxygen in the three first steps in the BJ model, where it reaches the same expression level as in the metastasizing cell under

atmospheric oxygen pressure. Genes

involved in angiogenesis such as E2F,

BRCA1 and ANGPTL4 are also among

the significantly upregulated genes in

the primary cell line. The top three

upregulated clusters in the SV40-

transformed cell line are mainly related

to responses to external stimuli and

hypoxia, while the corresponding

clusters of the last stage of the model

are more related to cell migration,

proliferation and apoptosis. The

downregulated clusters of the third

stage are related to various RNA

processing functions, as well as

regulation of apoptosis, while the

fourth stage has downregulated

clusters related to interferon signaling

and the immune response. Taken

together, the enrichment analyses

indicates that the four-stage model not

only acquires important cancer-related

milestones with decreased degree of

differentiation as previously shown, but

also that these have a large effect on

the cells’ response to lower oxygen

levels.

(7)

SV40-transformation leads to increased adaptation to lowered oxygen supply

By comparing DEGs shared across the model stages, the impact of the Large- T oncogene becomes even clearer:

not only are more genes differentially regulated in the transformed stage than in the others, the DEGs shared with the previous immortalized stage are also more often regulated in the opposite direction (Figure 4a). Out of the 216 DEGs that are shared

between the immortalized and transformed cells, 157 genes are regulated in opposite directions, and a majority of these are upregulated in the SV40-transformed cells. For example, Interleukin 8 (CXCL8) is upregulated more than five-fold in the SV40 transformed cells (31 TPM to 788 TPM) but is downregulated in the immortalized cells (84 TPM to 65 TPM). This protein is known to promote the expression of VEGFA which mediates homeostatic adaptation to hypoxic condition by promoting vascularization to compensate for the decrease in oxygen supply 21 . VEGFA is differentially expressed and

upregulated in the SV40 transformed cells and displays expression levels

changing from 30 TPM in atmospheric oxygen to 150 TPM in 3% oxygen.

Another interesting protein,

upregulated from 74 TPM to 345 TPM in the SV40-transformed cells, and downregulated from 1994 TPM to 166 TPM in the immortalized cells, is the mitochondrial superoxide dismutase, SOD2. This enzyme catalyzes the transformation of superoxide radicals (O 2 -) into either oxygen (O 2 ) or peroxide (H 2 O 2 ), and has been

considered one of the most important regulators of cellular redox state in normal and cancer cells. Due to its anti-apoptotic effect, overexpression of SOD2 has been linked to increased invasiveness of tumor metastases 22,23 . Increased levels of SOD2 has been linked to increased metastatic

capability by enabling cancer cells to maintain an increased growth potential and stay protected from excess ROS that would otherwise lead to apoptosis and necrosis 24 .

The dramatic change due to the SV40-

transformation is further shown among

the number of DEGs by comparing

expression within the model, rather

than between oxygen levels for the

same stages. Here, we observe 1330,

2976 and 732 DEGs between primary

versus immortalized, immortalized

(8)

versus transformed and transformed versus metastasizing, respectively, at atmospheric oxygen concentration, under which the model was initially created. Interestingly, the

corresponding numbers at low oxygen levels are 1072, 2000 and 288. All stages of the model show a larger amount of variation in terms of number of DEGs for atmospheric oxygen levels, while the differences become less pronounced in low oxygen. Again, the comparison between the last two stages of the model is clearly different from the first two, showing very few DEGs. This is in line with the

hypothesis that the model achieves most of its oxygen adaptations in the third stage. This could be explained by the fact that both of the two stages affect highly interconnected

oncogenes p53 and HRAS. Among the 264 DEGs across oxygen conditions that are shared between the primary and the immortalized cells, 89%

(N=236) are regulated in the same direction. Several of the upregulated genes are related to proliferation, for example the E2F receptor, MCM10 and the most commonly used marker for proliferation used in clinical

settings, MKI67 (Figure 4b). Among DEGs shared between the

transformed and metastasizing cells, a

large majority (78%) is regulated in the same direction. Several genes related to lipid metabolism and increased motility are upregulated, for example several chemokines and colony stimulating factors CSF2 and CSF3 (Figure 3c).

Transcriptome changes in low oxygen resembles the process of malignant transformation

Of all the 909 DEGs identified in the primary cell line, over a third are also DE within the model during the

atmospheric condition under which the model system was created, and a majority of these are regulated in the same direction. Several genes involved in proliferation and vasculature development that are upregulated due to the introduction of hTERT, are also up-regulated in 3%

oxygen, such as E2F receptors and several chemochines. ALDH1A1 that was previously shown, both on transcript and protein level, to completely disappear after

immortalization in atmospheric oxygen,

is also downregulated in 3% oxygen,

going from 429 TPM in atmospheric

condition to 54 TPM in 3% oxygen. For

the immortalized cell line this is even

clearer, sharing 60% of the DEGs

(9)

across oxygen conditions with DEGs that are differentially expressed due to the SV40 transformation. Several genes display similar patterns, for example upregulation of ANLN, AURKA and ADAM19.

SV40 triggers capability of metabolic switching

Among the DEGs observed across the oxygen conditions, the totality of

enzymes related to metabolic pathways exhibit an overall upregulation in low oxygen concentration compared to the atmospheric conditions (Figure 5).

Across the different stages of the model, the transformed tumor forming state (SV40-transformed) displays a relatively higher number of

upregulated metabolic genes

compared to the rest. Downregulation of genes belonging to lipid,

carbohydrate and amino acid metabolism in the immortalized cell line under moderate hypoxia is conspicuous, as shown in Figure 5b.

As shown in Figure 5c-d, upregulation of biosynthetic pathways during low oxygen supply in the transformed and metastasizing cell lines is evident. The downregulated enzymes in the

immortalized cell line are almost entirely reversed in favor of lipid,

carbohydrate and amino acid

metabolic pathways in the transformed cell lines. For example the fatty acid desaturase FADS1 is downregulated from 155 TPM to 71 TPM in the immortalized cells, but upregulated from 65 TPM to 137 TPM in the transformed cells. This protein regulated the unsaturation of fatty acids and in a recent study by others, reduced expression of this protein was shown to correlate with poor prognosis in small lung cancer, contradictive to our results and indicating a more complex role of this protein 25 . The aldo-keto reductase AKR1B1, which is involved in glycerolipid metabolism and often over-expressed in human cancers, show dramatic upregulation from 237 TPM to 1156 TPM in the SV40-transformed cells, whereas it is downregulated from 2490 TPM to 546 TPM in the immortalized cells.

Stearoyl-CoA (SCD), involved in the metabolism of fatty acids is another example of a protein that shows reversed regulation between the immortalized and transformed cells.

Knock down of this protein has

previously shown to induce apoptosis

in tumor cells grown in vitro, indicating

the ability for the SV40-transformed

cells to evade apoptosis through

(10)

upregulation of this metabolic process 26,27 . Another prominent

difference between the different stages of the model is the downregulation of energy metabolism in metastasizing cell lines (Figure 5d). Figure 5 also shows that enzymes taking role in lipid, carbohydrate and amino-acid metabolic pathways are largely shared within these pathways, resulting in the dissemination of the up or

downregulation to the entirety of these broad groups of pathways throughout the functional hierarchy (see the central lines in Figures 5 a-d).

Discussion

By profiling transcriptome changes under atmospheric and 3% oxygen conditions, we observe that even though the four cell lines in the BJ model for malignant transformation are highly similar, they display striking differences in their response to decreased oxygen supply. These differences are most prominent after the introduction of SV40 Large-T. The transformation with the SV40 Large-T oncogene has a large effect on the RNA expression of the cells, while the addition of the mutated HRAS has a smaller, relative effect. This separation

of the BJ model system into two groups representing pre and post transformation states is in accordance with our previous findings 4 .

The overall upregulation of the enzymes belonging to metabolic pathways throughout the different stages of cancer cell lines indicates that 3% oxygen supply, which is

considered moderate hypoxia, triggers metabolic activity. Relatively high upregulation of lipid, carbohydrate and amino acid metabolic pathways in the transformed tumor forming state in comparison to the previous cell lines may suggest a potential confirmation for the suggested mechanism, by which the cells increase the

consumption of ATP to trigger elevated fluxes of glycolytic pathways 28 . The metastasizing cell line expands the ATP consumption to other biosynthetic pathways, including glycan

biosynthesis and the metabolism of other amino acids in addition to a prominent increase in the

carbohydrate metabolism (Figure 5d).

The downregulation of oxidative

phosphorylation in metastasizing cells

during decreased oxygen supply

indicates the adaptation capability of

these cells to anaerobic conditions. In

contrast to the debated Warburg

effect, according to our results, the

(11)

metastasizing cells might prefer oxidative phosphorylation under

aerobic conditions whereas they down- regulate the genes taking role in oxidative phosphorylation favoring glycolytic pathways during low oxygen supply. Oxygen concentrations in human tumors are highly

heterogeneous, often containing regions where the oxygen

concentration reaches zero, known as anoxic regions 29 . Even though tumor hypoxia often manifests itself with lower oxygen concentrations than used in this study, we observe large differences in gene expression across the conditions. The BJ model has several inherent limitations in its ability to mimic the in vivo response to

hypoxia, mainly by being of fibroblast origin and created under atmospheric oxygen pressure. This greatly limits its use to studying the effects of oxygen levels on already transformed cells, and not the process in which oxygen levels may interplay in the process of

malignant transformation itself. With this study we show that the BJ model is not only a suitable model to study the mechanisms underlying malignant transformation. The potential of this model system expands beyond this and offers possibilities to expose the cells to perturbations such as drugs, under varying environmental

conditions to study the molecular effects and rewiring of signaling pathways. The BJ model offers a system to enable a deeper

understanding of the link between cancerous stage and response to environmental changes, which will add strength to the over-all mechanistic understanding of tumor development and behavior.

(12)

References

1. Hahn, W. C. et al. Creation of human tumour cells with defined genetic elements.

Nature 400, 464–468 (1999).

2. Vogelstein, B. & Kinzler, K.

W. The multistep nature of cancer. Trends Genet. 9, 138–141 (1993).

3. Hanahan, D. & Weinberg, R.

A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

4. Danielsson, F. et al. Majority of differentially expressed genes are down-regulated during malignant

transformation in a four-stage model. Proc. Natl. Acad. Sci.

U.S.A. 110, 6853–6858 (2013).

5. Gatenby, R. A. & Gillies, R. J.

Why do cancers have high aerobic glycolysis? Nat. Rev.

Cancer 4, 891–899 (2004).

6. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg Effect: The

Metabolic Requirements of Cell Proliferation. Science 324, 1029–1033 (2009).

7. Vaupel, P. Hypoxia and aggressive tumor phenotype:

implications for therapy and prognosis. Oncologist 13 Suppl 3, 21–26 (2008).

8. Thienpont, B. et al. Tumour hypoxia causes DNA

hypermethylation by reducing TET activity. Nature 537, 63–

68 (2016).

9. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L.

Near-optimal probabilistic

RNA-seq quantification. Nat.

Biotechnol. 34, 525–527 (2016).

10. Soneson, C., Love, M. I. &

Robinson, M. D. Differential analyses for RNA-seq:

transcript-level estimates improve gene-level inferences. F1000Res 4, 1521 (2015).

11. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data.

Bioinformatics 26, 139–140 (2010).

12. Fasterius, E. & Szigyarto, C.

A.-K. A Novel RNA

Sequencing Data Analysis Method for Cell Line Authentication .

13. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data.

Genome Res. 20, 1297–1303 (2010).

14. Cingolani, P. et al. A program for annotating and predicting the effects of single

nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin) 6, 80–92 (2012).

15. Cingolani, P. et al. Using Drosophila melanogaster as a Model for Genotoxic

Chemical Mutational Studies with a New Program, SnpSift.

Front Genet 3, 35 (2012).

16. Kanehisa, M., Sato, Y.,

Kawashima, M., Furumichi,

M. & Tanabe, M. KEGG as a

reference resource for gene

and protein annotation.

(13)

Nucleic Acids Res. 44, D457–

62 (2016).

17. Sanli, K., Sinclair, L., Nilsson, R. H., Mardinoglu, A. & Eiler, A. PACFM: Pathway Analysis with Circos in Functional Metagenomics. Manuscript.

18. Huang, D. W., Sherman, B. T.

& Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat.

Protocols 4, 44–57 (2008).

19. Gilkes, D. M., Semenza, G. L.

& Wirtz, D. Hypoxia and the extracellular matrix: drivers of tumour metastasis. Nat. Rev.

Cancer 14, 430–439 (2014).

20. Kim, J.-W., Tchernyshyov, I., Semenza, G. L. & Dang, C.

V. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular

adaptation to hypoxia. Cell Metabolism 3, 177–185 (2006).

21. Martin, D., Galisteo, R. &

Gutkind, J. S. CXCL8/IL8 stimulates vascular endothelial growth factor (VEGF) expression and the autocrine activation of VEGFR2 in endothelial cells by activating NFkappaB through the CBM

(Carma3/Bcl10/Malt1)

complex. J. Biol. Chem. 284, 6038–6042 (2009).

22. Yan, T., Oberley, L. W., Zhong, W. & St Clair, D. K.

Manganese-containing superoxide dismutase overexpression causes phenotypic reversion in SV40-transformed human

lung fibroblasts. Cancer Res.

56, 2864–2871 (1996).

23. Hempel, N., Carrico, P. M. &

Melendez, J. A. Manganese superoxide dismutase (Sod2) and redox-control of signaling events that drive metastasis.

Anticancer Agents Med Chem 11, 191–201 (2011).

24. Liu, X. et al. Deregulation of manganese superoxide dismutase (SOD2)

expression and lymph node metastasis in tongue

squamous cell carcinoma.

BMC Cancer 10, 365 (2010).

25. Wang, D. et al. Reduced Expression of FADS1

Predicts Worse Prognosis in Non-Small-Cell Lung Cancer.

J Cancer 7, 1226–1232 (2016).

26. Roongta, U. V. et al. Cancer cell dependence on

unsaturated fatty acids implicates stearoyl-CoA desaturase as a target for cancer therapy. Mol. Cancer Res. 9, 1551–1561 (2011).

27. Valli, A. et al. Hypoxia induces a lipogenic cancer cell phenotype via HIF1α- dependent and -independent pathways. Oncotarget 6, 1920–1941 (2015).

28. Fang, M. et al. The ER UDPase ENTPD5 promotes protein N-glycosylation, the Warburg effect, and

proliferation in the PTEN pathway. Cell 143, 711–724 (2010).

29. Brown, J. M. & Wilson, W. R.

Exploiting tumour hypoxia in cancer treatment. Nat. Rev.

Cancer 4, 437–447 (2004).

(14)

Figure 1 A. Experimental setup B. Spearman correlations between all samples.

Figure 2 A. Number of DEGs across oxygen conditions in the four stages of the model respectively. B. Up- and downregulation across the four stages of the model, showed as percentage.

BJ BJ

hTERT+ BJ

hTERT+ SV40

hTERT+ BJ BJ hTERT+ SV40

RasG12V

Malignant transformation

Atmospheric oxygen 3% oxygen

RNA-seq

A B

Cell Oxygen

CellOxygen

Oxygen Low Atm Cell

Primary Immortalized Transformed Metastasizing

0.9 0.92 0.94 0.96 0.98 1

(15)

Figure 3 Top three significantly enriched clusters based on the Gene Ontology domain Biological Function in DAVID, for up- and downregulated genes separately at each stage in the BJ model.

BJ BJ

hTERT+ BJ

hTERT+ SV40 BJ

hTERT+ SV40 RasG12V

Proliferation Angiogenesis Differentiation

Angiogenesis Proliferation Hypoxia

Collagen Migration

Response to lipid Renal dev.

Angiogenesis Cell cycle

tRNA metabolism Apoptosis

ncRNA process External stimulus Hypoxia

Angiogenesis

Immune response IFNγ response Stress response Migration Proliferation Apoptosis Regulation

in 3% oxygen

(16)

Figure 4 Shared DEGs across oxygen conditions between the cell lines in the BJ model.

A. Shared DEGs between immortalized and SV40-transformed cells. B. Shared DEGs between primary and immortalized cells. C. Shared DEGs between SV40-transformed and metastasizing cells.

A

log2FC BJ primary

log2FC BJ hTERT

−6 −4 −2 2 4 6

−6−4−2246

AMPD3

CXCL1 CXCL8

●●

log2FC BJ hTERT

log2FC BJ SV40

−6 −4 −2 2 4 6

−6−4−2246

CD82 AKR1B1 SOD2

ID3 IL1B

ASS1

GPNMB IL6

IGFBP3 STC1

IL24

CXCL1 CXCL8

MME

log2FC BJ SV40

log2FC BJ Ras

−6 −4 −2 2 4 6

−6−4−2246

ACTG2 HRAS

δTPM <5 δTPM 5−50 δTPM 50−100 δTPM 100−500 δTPM >500

A B

C

(17)

B

C

D

A

(18)

Figure 5. Fold changes and linkages of metabolic functions in control vs. moderate hypoxic treatments of the cell lines across the BJ model. A. PACFM plot based on the metabolic profiling of BJ RNA-Seq data by using the KEGG Orthology database as reference representing the fold changes of hypoxic cell lines in comparison to

atmospheric oxygen treatment. The outer and the inner circular heat maps show the average log fold changes of the functional categories within metabolism and the second hierarchy level KEGG Orthology functions, respectively. The bars in the third level represent the average fold changes of pathway level categories. Upregulated pathways are represented with red, and blue color-coding represents downregulated pathways.

Similarly, the inner most circle shows the fold changes of individual enzymes bar three level color coding where red and blue represent up- and downregulation, respectively in addition to the green bars showing no change between control and hypoxic

treatments. Bezier curves in the middle of the plot represent the enzymes shared between the broad categories at the second hierarchy level. The lines start from the category that the color codes represent and end up at various categories at the

equivalent hierarchy level. Full names of the abbreviated KEGG categories shown in this plot can be found in Supplementary material. B. Immortalized cells C. SV40-transformed cells D. Metastasizing cells

References

Related documents

Since both cell lines arise from the same cancer form, pancreatic cancer, it can be seen that BxPC-3 exhibit similar results as seen in for Panc-1 in Figure 2 showing the

At Company D, the software division have adopted Scrum artifacts by having a Backlog, monitoring the Sprint and always trying to reach an Increment each Sprint hence the

[ 18 ] Radiances for the AMSU-B sounding channels were simulated by ARTS and RTTOV using the interpolated profiles as input, that means that the radiative transfer equation

By transcriptome profiling under atmospheric and moderate hypoxic (3% O 2 ) conditions, we observed that despite being highly similar, the four cell lines of the BJ model

After 1 month in culture, HPC LSK BCR/ABL p210 cells show reduced expression of stem cell markers (c-Kit, Sca-1) and differentiate into myeloid (CD11b, Gr-1), but not lymphoid

[13] , then used to assess the response of astrocytes to three typical examples of cell therapies that are currently under investigation for CNS repair: neu- ral crest stem

“participants are typically presented with one sentence or clause at a time and are instructed to comment on their understanding of the sentence or clause in the context of the

In accordance with Osterwalder and Pigneur (2010) and our case about “Innovative Logistics in Halland” we would like to argue that adaption of potential