• No results found

University of Groningen Functions of the C/EBPβ isoforms in breast cancer Sterken, Britt

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Functions of the C/EBPβ isoforms in breast cancer Sterken, Britt"

Copied!
17
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Functions of the C/EBPβ isoforms in breast cancer

Sterken, Britt

DOI:

10.33612/diss.172465560

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2021

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Sterken, B. (2021). Functions of the C/EBPβ isoforms in breast cancer. University of Groningen. https://doi.org/10.33612/diss.172465560

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter VI

(3)

The transcription factor C/EBPβ is a member of the CCAAT/enhancer binding protein family and is expressed in all tissues in the body (https://www.proteinatlas.org/ENSG00000172216-CEBPB). It is a single exon gene, but comes in different protein isoforms (LAP*, LAP and LIP) that are alternatively translated from a single CEBPB-mRNA. Translation into LAP and LAP* requires recognition of regular translation initiation sites. However, translation into LIP requires the translation of a uORF within the CEBPB-mRNA, and genetic ablation of the uORF results in a LIP-deficiency 1. A large number of studies have demonstrated that C/EBPβ is a mediator of proliferation and differentiation in a variety of tissues 2–13. Its protein isoforms LAP and LIP are widely expressed, but structurally and functionally different, as LIP lacks the N-terminal transactivation domains but competes for the same DNA binding sites as LAP. Therefore, the transcriptional activity of C/EBPβ is determined by the ratio between the LIP and LAP isoforms.

Previously, we established a translational control model for C/EBPβ, where the genetic ablation of the uORF resulted in systemic LIP-deficiency in the mouse 12. The phenotypes of these mice resemble the phenotypes of mice upon caloric restriction or mTORC1 inhibition, showing an increased health-and lifespan and a reduced overall tumour incidence 4,14. In addition, knock-in of LIP in at the Cebpb locus results in a reduced lifespan and increased overall tumour incidence, proposing that LIP has oncogenic functions 13. Integration of LIP under the whey acidic protein (WAP) promotor results in mammary epithelial specific overexpression of LIP and development of hyperplasias and less frequently neoplasias 15. C/EBPβ is a key transcriptional regulator in mammary gland development, its absence resulting in deficient mammary gland development (reviewed in 16,17). Given the low mutation rate, copy number variation and

mRNA overexpression of CEBPB in breast cancer

(https://cancer.sanger.ac.uk/cosmic), likely, the translational control into its different isoforms is involved in mediating the oncogenic functions in cancer development. In addition, a variety of studies have linked growth signalling to a favoured translation into the LIP isoform 18–20. Moreover, Gene Set Enrichment Analysis (GSEA) identified enriched gene expression patterns in triple negative

(4)

breast cancer samples that share common promotor motifs for C/EBPβ 21. However, the exact role of C/EBPβ in TNBC remains so far unknown. In this thesis, we show that LAP inhibits cell migration and invasion of cell lines derived from TNBC. Moreover, we show a regulation of microenvironmental genes by C/EBPβ in BT20 cells and in specific a regulation of extracellular matrix genes. We show that endogenous variation of the LIP/LAP ratio in basal-like murine tumours does not correlate with tumour specific survival. In addition, we generated a translational control model for C/EBPα to study the role of the isoforms on metabolism and oncogenesis.

C/EBPβ as a potential regulator of breast cancer metastasis

Previously, it was revealed that high LIP expression in human breast cancer samples negatively correlates with estrogen receptor (ER) and progesterone (PR) expression, and correlates with proliferation, aneuploidy and poor prognosis 22. Later on, studies demonstrated that LIP is highly expressed in HER2+ cancer and that LIP overexpressing tumours showed resistance to treatment with the ERBB2 inhibitor trastuzumab 18. In chapter III, we demonstrated that cell lines derived from TNBC display a high expression of LIP and a high LIP/LAP ratio. We observe that lowering the LIP/LAP ratio by overexpression of LAP reduces the migration and invasion of TNBC cells, and that C/EBPβ regulates migration-related and microenvironment migration-related genes in BT20 cells. Therefore, the C/EBPβ isoforms are potentially an interesting therapeutic target in treating cancer cell dissemination.

Breast cancer metastasis can be summarised as a series of events including the migration, invasion, extravasation, intravasation and colonisation at distant target sites 23. The metastatic process involves a complex interplay between the cancer cells and its surrounding microenvironment. During cancer progression, tumour cells often become plastic and undergo morphological and phenotypical conversions, which are often induced by an Epithelial to Mesenchymal Transition (EMT) 24. The EMT is characterised by a downregulation of epithelial markers

(5)

and a gain of mesenchymal markers, and is a highly conserved and reversible process during which immotile epithelial cells give rise to polarised migrating mesenchymal cells. Previous studies have demonstrated a regulation of EMT genes by C/EBPβ, however, evidence is largely contradictory. One study provides evidence that LAP induces the EMT and transforms MCF10a cells 25, whereas the majority of studies reveals oncogenic functions for LIP (reviewed in Chapter II). Moreover, multiple studies have investigated the role of C/EBPβ in the EMT, but not the isoform specific functions. For instance, it was revealed that loss of C/EBPβ is required for TGFβ-induced EMT 26. In this study, we revealed a downregulation of E-cadherin and an upregulation of N-cadherin and Vimentin upon LIP overexpression in MCF10a cells, however, other mesenchymal markers were downregulated. Moreover, Gene Set Enrichment Analysis (GSEA) revealed a mixed regulation of EMT genes, indicating that C/EBPβ and its isoforms regulate EMT genes, but not in a one-directional manner. Whereas during the EMT in embryonic development epithelial cells lose their epithelial origin and become fully mesenchymal, cancer cells are often characterised by a “ hybrid” E/M phenotype. Also referred to as a partial EMT, this intermediate status between epithelial and mesenchymal phenotypes is thought to enhance invasiveness as reviewed in 27. Often, this partial EMT is characterised by collective cell migration, where the cells with epithelial phenotypes remain attached to other cells and mesenchymal leader cells interact with the ECM. Therefore, more detailed analysis of the role of the C/EBPβ isoforms in the EMT is required. Analysis of the RNA-seq data obtained from BT20 CEBPB-knockout cells revealed a downregulation of pro-oncogenic extracellular matrix genes and remodellers. Previously it was revealed that C/EBPβ drives the malignancy of mammary epithelial cells by promoting the expression of α5 integrin transcription, putting C/EBPβ forward as a regulator of integrin-dependent stromal–epithelial interactions 28. α5 integrin and its ligand fibronectin (FN) are frequently upregulated in metastatic tumours, and the study showed that inhibition of ligation of FN to α5 integrin reverted the phenotype towards non-malignant tissue. Here, we observe a downregulation of Tenascin C (TNC), Collagen 5A1 (COL5A1), Fibronectin 1 (FN1), and Thrombospondin 1 (THBS1)

(6)

(all ECM organisation) and MMP2, Lysyl Oxidase Like 1 (LOXL1) and Procollagen C-Endopeptidase Enhancer 2 (PCOLCE2) (all ECM remodelling) upon CEBPB-ko in BT20 cells. Moreover, a majority of those genes demonstrated to be upregulated by LAP and not LIP, indicating that potentially LAP promotes metastasis by contributing to remodelling of and interaction with the ECM during breast cancer progression. To understand how exactly LIP and LAP regulate cell migration and invasion, its interactions with the ECM and deposition and remodelling of the ECM should be analysed. Furthermore, expression patterns of the isoforms in different types, stages, and regions within the tumours could contribute to understanding the isoform-specific roles in breast cancer progression. One major challenge in this field is the lack of a LIP-specific antibody. Whereas LAP can LIP-specifically be targeted by the use of a N-terminal specific antibody, LIP is identical to the C-N-terminal part of LAP and even though multiple groups have put effort into generating a LIP specific antibody, it so far remained unsuccessful. Therefore, studying the expression of LIP and LAP in different types and stages of human breast tumours, and modelling of LIP and LAP in GEMM models for breast cancer, is required to establish the functions of the isoforms in breast cancer progression and metastasis.

In addition to the regulation of extracellular matrix related genes, DAVID functional clustering analysis revealed an upregulation of MHCI and MHCII and antigen presentation related genes upon CEBPB-knockout in BT20 cells. As LIP is the predominantly expressed C/EBPβ isoform in the parental BT20 cell line, one could hypothesise that LIP downregulates MHCI and MHCII molecules. Immune-escape strategies are commonly found in transformed cells, which include the loss/reduction of MHCI and MHCII expression to avoid T-cell recognition 29. Therefore, our RNA-seq data propose a potential additional role in the evasion of immune cells for the C/EBPβ-isoforms.

We previously reported that LIP reprograms cellular metabolism by inducing the stem cell factor Lin28b via repression of let-7 microRNAs. In order for cancer cells to establish uncontrolled proliferation it requires adjustments in cellular metabolism. Under aerobic conditions, cells use glucose in the glycolysis which

(7)

gets processed to pyruvate, which in the mitochondria is processed to carbon dioxide. Upon anaerobic conditions, little of this pyruvate is dispatched to the mitochondria and lactate production is favoured. Otto Warburg demonstrated that cancer cells often acquire a different metabolism known as “aerobic glycolysis”, during which the cancer cells, even upon aerobic conditions, limit their metabolism largely to glycolysis, even though oxygen is available 30,31. More recent research revealed also an important role of mitochondrial metabolism – the TCA cycle and oxidative Phosphorylation (OXPHOS) – in cancer growth, with tumour cells displaying increased flux through mitochondrial pathways 32. More recently, it was revealed that metastatic breast cancer cell lines display an enhanced metabolic activity, by upregulating both the glycolysis and OXPHOS (reviewed in 33). Moreover, derivation of metastatic breast cancer cells with preferential colonisation sites revealed different metabolic profiles, with cells prone to liver metastasis upregulating glycolysis, whereas cells prone to bone and lung metastasis favour OXPHOS 34. Interestingly, we find that LIP boosts both basal and maximal extracellular acidification rate (ECAR) as a measure for glycolysis and oxygen consumption rate (OCR) as a measure for OXPHOS, and that LAP only boosts the OCR. Furthermore, we demonstrate that this metabolic induction by LIP is dependent on RNA-binding protein Lin28b 5, which is known to regulate the translation of mRNAs for glycolytic and mitochondrial enzymes35,36. Therefore, further studies should investigate how potential metabolic alterations induced by LIP/LAP might influence where cancer cells colonise, but also whether potential regulation of translation into LIP/LAP might be a way to adjust or adapt to metastasising tumour cells’ needs. Furthermore, the regulation of metabolic enzymes by LIP was demonstrated to be regulated in a post-translational fashion. Therefore, in addition to our existing RNA-seq data obtained from BT20 CEBPB-knockout cells, proteomics might reveal a potential additional regulation level mediated by C/EBPβ in TNBC cells and could aid the search for targets that promote breast cancer progression.

Previously we have successfully reduced the LIP/LAP ratio in vitro in a screen using the ENZO library of FDA-approved drugs, and for future experiments it will be interesting to test these drugs in vivo and study their effects on breast cancer metastasis 37. The role of the LIP and LAP isoforms in cell invasion, their partial regulation of the EMT and their effect on the microenvironment have to

(8)

be further researched in in vitro and in vivo models to reveal whether the LIP/LAP ratio can be put forward as a therapeutic target in breast cancer.

Generation of C/EBPβ-isoform specific Genetically Engineered Mouse Models (GEMMs) for breast cancer

The lack of proper models to investigate the role of LIP and LAP in breast tumourigenesis is one major limitation in the field of C/EBPβ. Whereas studies previously have used allografts to maintain intact immune systems, it often does not reflect the mutational heterogeneity and intratumoural clonal heterogeneity found in mammary tumours. Given this tumour complexity, the use of genetically engineered mouse models (GEMMs) has become one of the most accurate ways to model the disease, its de novo tumourigenesis allowing analysis of cause-effect relations, rather than correlations 38. In addition, given the regulation of antigen presentation-related and extracellular matrix-like genes as described in Chapter III, modelling of the role of the C/EBPβ-isoforms in an intact breast cancer microenvironment seems essential.

For these reasons, we chose to model LIP and LAP in a GEMM model for breast cancer. Given the high mutational rate of p53 in basal-like breast cancer ( ~80 % of all basal-like cancers), we chose to make use of the existing Wap-Cre;Trp53F/F (WP) model 39–41, where we selectively overexpressed LIP-luciferase and LAP-luciferase. Based on data obtained from MMECs isolated from the WP-LIP and WP-LAP cohorts, we concluded that LIP and LAP expression is silenced by DNA-methylation, which corresponded to a lack of luminescence signal in the majority of WP-LIP and WP-LAP tumours. Whereas in previous studies c-Myc and Cas9 have been successfully integrated and overexpressed from the Col1a1 locus in the same mouse model 42, similar to our study, the overexpression of esterogen receptor (ER) α is silenced over time 43. Given previous reports that obtaining ERα positive mammary tumours in mice has proven to be difficult (most GEMMs developing ERα -negative tumours and the few cases that do develop ER+ tumours do not respond to estrogen or endocrine therapy) 44, this

(9)

indicates that transgene expression of genes that do not favour tumour growth are prone to epigenetic silencing in this mouse model. Even though LIP proves to promote hyperplasia formation in the mammary gland and predisposes to oncogenesis 13,15, this study indicates that perhaps only strong tumour drivers are successfully expressed in this p53-deficient breast cancer model.

Analysis of tumours obtained from WP, WP-LIP and WP-LAP cohorts revealed consistent expression of C/EBPβ in all tumours and a high variability in LIP/LAP ratios amongst the tumours. With an average LIP/LAP ratio of ~1 in the tumours from all cohorts, this ratio proved to be higher than in normal differentiated tissues 45, but lower than we observed in cell lines derived from TNBC (LIP:LAP ratios of ~3). This corresponds to a relatively low LIP/LAP ratio observed in the murine basal-like 4T1 cell line 26. Even though LIP transgene expression increases hyperproliferation in the mammary gland 15, the observed differences in LIP/LAP ratios between human and mouse basal-like breast cancer might involve different C/EBPβ isoform functions between the two species. The variation in the LIP/LAP ratio in the p53-deficient mouse mammary tumours allowed for correlational studies between LIP/LAP ratio and survival. Even though we found no correlation between LIP/LAP ratio and mammary tumour-specific survival, more drastic changes in LIP/LAP ratio might be required to influence tumour development, as is reported and observed in our CebpbΔuORF model 45. Given the relatively high expression of C/EBPβ in the mammary tumours obtained from our cohorts, loss-of-function studies might reveal more about the functions of the C/EBPβ-isoforms. Our existing CebpbΔuORF model drastically decreases the

LIP/LAP ratio systemically, and might be a suitable model to study the loss of function of LIP in mammary tumourigenesis. Therefore, we propose crossing of the CebpbΔuORF model with the existing Wap-Cre:Trp53F/F model, followed by surgical resection and transplantation of tumours into WT recipient mice to prevent potential systemic effects within the CebpbΔuORF mice. In addition, to

model the LIP overexpression, our R26-LIP mice can be crossed with the Wap-Cre:Trp53F/F model, as LIP transgene expression from the R26 locus has been proven not to be epigenetically silenced 5.

(10)

Generation of the C/EBPα

-∆uORF mouse

Previously, we published that mutation of the CEBPB-uORF results in a LIP-deficiency which increases health-and lifespan in mice that display a reduced overall tumour incidence, resembling the phenotype of mice kept under caloric restriction or mTORC1 inhibition4,12,14. Both C/EBPα and C/EBPβ translation is controlled via mTORC1 signalling, with mTORC1 signalling favouring translation into the truncated isoforms LIP and p30 respectively (reviewed in Chapter II, Chapter V and published in 1). To investigate the importance of uORF-mediated translational control of C/EBPα in vivo, we generated the C/EBPα-∆uORF mouse model. Both C/EBPα and C/EBPβ are established regulators of metabolism, proliferation, and differentiation. In particular, C/EBPα is known for mediating adipocyte differentiation46–49. To test the involvement of the C/EBPα uORF-mediated translational control in the regulation of energy homeostasis, mice will be subjected to a series of tests to measure insulin sensitivity, glucose tolerance, leptin sensitivity, and energy expenditure and body weight and composition. Cebpa-/- mice die a few hours after birth due to hypoglycemia, and show defects in lung and liver development and a lack of mature neutrophils and eosinophils. Crossing of heterozygous C/EBPα-∆uORF mice reveals offspring generation at regular Mendelian ratios, indicating that p30 is dispensible for normal development, or potentially another truncated isoform of CEBPs takes over during development – even though preliminary results show no upregulation of C/EBPβ-LIP upon depletion of p30. Moreover, preliminary data obtained from young cohorts (3-6 months of age) reveals no clear differences in mouse weight, which might show larger differences in weight at higher age as described for the C/EBPβ-∆uORF mice4.

In addition to the established functions in fat differentiation and metabolism, C/EBPα is a major regulator of proliferation. The p42 isoform is a well-known tumour suppressor and CEBPA is found mutated in ~15% of the acute myeloid leukemia (AML) patients, often harbouring nonsense mutations in the coding sequence specific for p42, thereby abolishing p42 expression while preserving p30 50,51. In addition, in vivo models harbouring mutations resulting in p42-deficiency

(11)

result in the development of AML 52. Next to its role in AML, C/EBPα is known to display decreased levels in solid tumours of breast, liver, skin, head and neck, lung, gastric, cervical, endometrial and liposarcoma. 53–61. Interestingly, recently C/EBPα has been reported to influence the metastatic process by regulating the EMT, where it was shown that C/EBPα is downregulated via TGFβ signalling directly via SMAD3 62. Overexpression of C/EBPα, after EMT induction by stimulation with TGFβ, induces the Mesenchymal to Epithelial Transition (MET) in breast epithelial cells, and they therefore speculate that C/EBPα is an epithelial gatekeeper and that re-expression of C/EBPα might be favourable in the prevention of breast cancer metastasis. Even though the authors do not elaborate on isoform-specific functions, there are certain parallels with our study described in Chapter III, where overexpression of LIP in untransformed mammary epithelial cells (partially) induces the EMT, hypothesising that this might inhibit the function of LAP. The redundant functions described earlier for C/EBPα and C/EBPβ might explain why LIP induces the EMT, by either inhibiting the epithelial functions of LAP and p42. However, the overexpression of LAP in TNBC cell lines (data not shown) does not result in a regulation of EMT genes, and similarly, Lourenço et al do not show the regulation of EMT markers by C/EBPα in breast cancer models 62. Possibly, the CEBP interaction partners that are required for the regulation of EMT genes, are present in untransformed epithelial cells but not cancer cells. To test the involvement of the C/EBPα and C/EBPβ isoforms in EMT, the p30-deficient C/EBPα-∆uORF model could be crossed with existing breast cancer mouse models, to study whether breast cancer cells without p30 are able to undergo EMT. Moreover, one could test whether the short isoforms of C/EBPα (p30) and C/EBPβ (LIP) inhibit the function of C/EBPα (p42) and its epithelial induction, by making a double mutant with p30 and LIP deficiency, and crossing this with an existing metastatic mouse model. Potentially, this regulation of C/EBPα and C/EBPβ influencing breast cancer progression by (partly) regulating the EMT, is regulated at later stages by translational control mediated by oncogenes, such as mTORC1, or c-MYC which is characteristically amplified in TNBC.

(12)

Overall conclusion

In this thesis, we show that LAP overexpression decreases migration and invasion of triple-negative breast cancer cells and LIP promotes migration in breast epithelial cells, which we find associated with a mixed EMT signature. We propose a role for the LIP/LAP ratio in the regulation of breast cancer cell migration and ECM remodelling, two key characteristics that are associated with the aggressive phenotype of TNBC cells (Chapter III). We followed up to study the effects of LIP and LAP in in vivo using GEMMs for basal-like breast cancer in Chapter IV, where we showed that the exogenous LIP and LAP expression are silenced by methylation, and that endogenous variation in LIP/LAP ratio does not correlate to tumour onset and survival. In chapter V, we designed a C/EBPα-∆uORF model to study the effects of p30 ablation on health, ageing and cancer development, which potentially can be used to study the EMT in breast cancer. Our studies propose that both LIP and LAP isoforms might contribute to breast tumour development by altering cell invasion, metabolism and the microenvironment. However, future GEMM models for breast cancer containing uORF mutations and LIP transgene expression will be required to investigate how LIP and LAP contribute to tumour progression and metastasis.

(13)

References

1. Calkhoven, C. F., Müller, C. & Leutz, A. Translational control of C/EBPalpha and C/EBPbeta isoform expression. Genes Dev. 14, 1920–32 (2000).

2. Screpanti, I. et al. Lymphoproliferative disorder and imbalanced T-helper response in C/EBP beta-deficient mice. EMBO J. 14, 1932–41 (1995).

3. Greenbaum, L. E. et al. CCAAT enhancer-binding protein β is required for normal hepatocyte proliferation in mice after partial hepatectomy. J. Clin. Invest.

102, 996–1007 (1998).

4. Müller, C. et al. Reduced expression of C/EBPβ-LIP extends health and lifespan in mice. Elife 7, e34985 (2018).

5. Ackermann, T. et al. C/EBPβ-LIP induces cancer-type metabolic reprogramming by regulating the let-7/LIN28B circuit in mice. Commun. Biol. 2, 208 (2019). 6. Sterneck, E., Tessarollo, L. & Johnson, P. F. An essential role for C/EBPβ in female

reproduction. Genes Dev. 11, 2153–2162 (1997).

7. Robinson, G. W., Johnson, P. F., Hennighausen, L. & Sterneck, E. The C/EBPbeta transcription factor regulates epithelial cell proliferation and differentiation in the mammary gland. Genes Dev. 12, 1907–16 (1998).

8. Zhu, S. et al. C/EBPβ Modulates the Early Events of Keratinocyte Differentiation Involving Growth Arrest and Keratin 1 and Keratin 10 Expression. Mol. Cell. Biol.

19, 7181–7190 (1999).

9. Müller, C., Kowenz-Leutz, E., Grieser-Ade, S., Graf, T. & Leutz, A. NF-M (chicken C/EBPβ) induces eosinophilic differentiation and apoptosis in a hematopoietic progenitor cell line. EMBO J. 14, 6127–6135 (1995).

10. Pall, M. et al. The transcription factor C/EBP-β and its role in ovarian function: Evidence for direct involvement in the ovulatory process. EMBO J. 16, 5273–5279 (1997).

11. Buck, M., Poli, V., Van Der Geer, P., Chojkier, M. & Hunter, T. Phosphorylation of rat serine 105 or mouse threonine 217 in C/EBPβ is required for hepatocyte proliferation induced by TGFα. Mol. Cell 4, 1087–1092 (1999).

12. Wethmar, K., Calkhoven, C. F. & Leutz, A. RESEARCH COMMUNICATION C / EBPb DuORF mice — a genetic model for uORF-mediated translational control in mammals. Genes Dev. 15–20 (2010).

13. Begay, V. et al. Deregulation of the endogenous C/EBPβ LIP isoform predisposes to tumorigenesis. J. Mol. Med. 93, 39–49 (2014).

14. Zidek, L. M. et al. Deficiency in mTORC1-controlled C/EBPβ-mRNA translation improves metabolic health in mice. EMBO Rep. 16, 1022–36 (2015).

15. Zahnow, C. A., Cardiff, R. D., Laucirica, R., Medina, D. & Rosen, J. M. A role for CCAAT/enhancer binding protein beta-liver-enriched inhibitory protein in mammary epithelial cell proliferation. Cancer Res. 61, 261–9 (2001).

(14)

16. Hennighausen, L. & Robinson, G. W. Signaling Pathways in Mammary Gland Development. Dev. Cell 1, 467–475 (2001).

17. Shillingford, J. M. & Hennighausen, L. Experimental mouse genetics - Answering fundamental questions about mammary gland biology. Trends Endocrinol. Metab. 12, 402–408 (2001).

18. Arnal-Estapé, A. et al. HER2 silences tumor suppression in breast cancer cells by switching expression of C/EBPβ isoforms. Cancer Res. 70, 9927–9936 (2010). 19. Baldwin, B. R., Timchenko, N. A. & Zahnow, C. A. Epidermal Growth Factor

Receptor Stimulation Activates the RNA Binding Protein CUG-BP1 and Increases Expression of C/EBP -LIP in Mammary Epithelial Cells. Mol. Cell. Biol.

24, 3682–3691 (2004).

20. Li, H., Baldwin, B. R. & Zahnow, C. a. LIP expression is regulated by IGF-1R signaling and participates in suppression of anoikis. Mol. Cancer 10, 100 (2011). 21. Willis, S. et al. Enriched transcription factor signatures in triple negative breast

cancer indicates possible targeted therapies with existing drugs. Meta Gene 4, 129–141 (2015).

22. Zahnow, C. a, Younes, P., Laucirica, R. & Rosen, J. M. Overexpression of C/EBPbeta-LIP, a naturally occurring, dominant-negative transcription factor, in human breast cancer. J. Natl. Cancer Inst. 89, 1887–1891 (1997).

23. Welch, D. R. & Hurst, D. R. De fi ning the Hallmarks of Metastasis Neoplastic progression Hallmarks of cancer. 1–18 (2019). doi:10.1158/0008-5472.CAN-19-0458

24. Kim, D. H. et al. Epithelial Mesenchymal Transition in Embryonic Development , Tissue Repair and Cancer : A Comprehensive Overview. 1–25 doi:10.3390/jcm7010001

25. Bundy, L. M. & Sealy, L. CCAAT/enhancer binding protein beta (C/EBPβ)-2 transforms normal mammary epithelial cells and induces epithelial to mesenchymal transition in culture. Oncogene 22, 869–883 (2003).

26. Johansson, J. et al. MiR-155-mediated loss of C/EBPβ shifts the TGF-β response from growth inhibition to epithelial-mesenchymal transition, invasion and metastasis in breast cancer. Oncogene 32, 5614–5624 (2013).

27. Saitoh, M. JB special review-cellular plasticity in epithelial homeostasis and diseases: Involvement of partial EMT in cancer progression. J. Biochem. 164, 257– 264 (2018).

28. Damiano, L. et al. Oncogenic targeting of BRM drives malignancy through C/EBPβ-dependent induction of ’5 integrin. Oncogene 33, 2441–2453 (2014). 29. Thibodeau, J. & Lapointe, R. Targeting the MHC Class II antigen presentation

pathway in cancer immunotherapy. 1, 908–916 (2012).

(15)

(1956).

31. Heiden, M. G. Vander et al. Understanding the Warburg Effect : Cell Proliferation. 324, 1029–1034 (2009).

32. Ahn, C. S. & Metallo, C. M. Mitochondria as biosynthetic factories for cancer proliferation. Cancer Metab. 3, 1–10 (2015).

33. Jia, D., Park, J., Jung, K., Levine, H. & Kaipparettu, B. Elucidating the Metabolic Plasticity of Cancer: Mitochondrial Reprogramming and Hybrid Metabolic States. Cells 7, 21 (2018).

34. Dupuy, F. et al. PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer. Cell Metab. 22, 577–589 (2015).

35. Ma, X. et al. Lin28/let-7 axis regulates aerobic glycolysis and cancer progression via PDK1. Nat. Commun. 5, 5212 (2014).

36. Zhang, J. et al. LIN28 Regulates Stem Cell Metabolism and Conversion to Primed Pluripotency. Cell Stem Cell 19, 66–80 (2016).

37. Zaini, M. A. et al. A screening strategy for the discovery of drugs that reduce C/EBPβ-LIP translation with potential calorie restriction mimetic properties. Sci. Rep. 7, 42603 (2017).

38. Annunziato, S., Barazas, M., Rottenberg, S. & Jonkers, J. Genetic dissection of cancer development, therapy response, and resistance in mouse models of breast cancer. Cold Spring Harb. Symp. Quant. Biol. 81, 141–150 (2016).

39. Jonkers, J. et al. Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nat. Genet. 29, 418–425 (2001). 40. Derksen, P. W. B. et al. Mammary-specific inactivation of E-cadherin and p53

impairs functional gland development and leads to pleomorphic invasive lobular carcinoma in mice. Dis. Model. Mech. 4, 347–58 (2011).

41. Huijbers, I. J. et al. Using the GEMM-ESC strategy to study gene function in mouse models. Nat. Protoc. 10, 1755–1785 (2015).

42. Annunziato, S. et al. Comparative oncogenomics identifies combinations of driver genes and drug targets in BRCA1-mutated breast cancer. Nat. Commun.

10, (2019).

43. Cornelissen, L. M. et al. Exogenous ERα Expression in the Mammary Epithelium Decreases Over Time and Does Not Contribute to p53-Deficient Mammary Tumor Formation in Mice. J. Mammary Gland Biol. Neoplasia 24, 305–321 (2019).

44. Özdemir, B. C., Sflomos, G. & Brisken, C. The challenges of modeling hormone receptor-positive breast cancer in mice. Endocr. Relat. Cancer 25, R319–R330 (2018).

45. Müller, C. et al. Reduced expression of C/EBPβ-LIP extends health and lifespan in mice. Elife 7, (2018).

(16)

adjacent binding on a genome-wide scale. Genes Dev. 22, 2941–2952 (2008). 47. Rosen, E. D. et al. RESEARCH COMMUNICATION C / EBPα induces

adipogenesis through PPARγ : a unified pathway. Genes Dev. 16, 22–26 (2002). 48. Pedersen, T. A. et al. Distinct C/EBPalpha motifs regulate lipogenic and

gluconeogenic gene expression in vivo. EMBO J. 26, 1081–93 (2007).

49. Siersbæk, R. & Mandrup, S. Transcriptional networks controlling adipocyte differentiation. Cold Spring Harb. Symp. Quant. Biol. 76, 247–255 (2011).

50. Pabst, T. et al. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-α (C/EBPα), in acute myeloid leukemia. Nat. Genet. 27, 263–270 (2001).

51. Snaddon, J. et al. Mutations of CEBPA in acute myeloid leukemia FAB types M1 and M2. Genes, Chromosom. Cancer 37, 72–78 (2003).

52. Kirstetter, P. et al. Modeling of C/EBPα Mutant Acute Myeloid Leukemia Reveals a Common Expression Signature of Committed Myeloid Leukemia-Initiating Cells. Cancer Cell 13, 299–310 (2008).

53. Bennett, K. L. et al. Tumor suppressor activity of CCAAT/enhancer binding protein a is epigenetically down-regulated in head and neck squamous cell carcinoma. Cancer Res. 67, 4657–4664 (2007).

54. Gery, S. et al. Down-regulation and growth inhibitory role of C/EBPα in breast cancer. Clin. Cancer Res. 11, 3184–3190 (2005).

55. Wu, Y. V. et al. Restoration of C/EBPα in dedifferentiated liposarcoma induces G2/M cell cycle arrest and apoptosis. Genes Chromosom. Cancer 51, 313–327 (2012).

56. Tseng, H. H. et al. Reduced expression of C/EBPα protein in hepatocellular carcinoma is associated with advanced tumor stage and shortened patient survival. J. Cancer Res. Clin. Oncol. 135, 241–247 (2009).

57. Shim, M., Powers, K. L., Ewing, S. J., Zhu, S. & Smart, R. C. Diminished expression of C/EBPα in skin carcinomas is linked to oncogenic ras and reexpression of C/EBPα in carcinoma cells inhibits proliferation. Cancer Res. 65, 861–867 (2005).

58. Costa, D. B. et al. tumors : a correlative study of E3590. 56, 97–103 (2012). 59. Pan, Z. et al. Down-regulation of the expression of CCAAT/enhancer binding

protein α gene in cervical squamous cell carcinoma. BMC Cancer 14, 1–9 (2014). 60. Shi, D.-B. et al. C/EBPα-induced miR-100 expression suppresses tumor

metastasis and growth by targeting ZBTB7A in gastric cancer. Cancer Lett. 369, 376–385 (2015).

61. Takai, N. et al. Discovery of epigenetically masked tumor suppressor genes in endometrial cancer. Mol. Cancer Res. 3, 261–269 (2005).

62. Lourenço, A. R. et al. C/EBPɑ is crucial determinant of epithelial maintenance by preventing epithelial-to-mesenchymal transition. Nat. Commun. 11, 1–18 (2020).

(17)

Referenties

GERELATEERDE DOCUMENTEN

Next we investigated whether ectopic expression of LIP in the low LIP-expressing T47D and MCF-7 Luminal A cell lines results in increased sensitivity to 2-DG, as measured by

Therefore, in connection with the lower cancer incidence in the female LIP-deficient C/EBPβ ΔuORF mice and the increased cancer incidence in mice with LIP overexpression,

Our data suggests C/EBPβ-LIP as an important mediator of metabolic signalling since its expression is responsive to nutrient levels (though mTORC1) and it controls cellular

Onze data draagt C/EBPβ-LIP voor als belangrijke regulator van metabole signalling omdat de expressie gereguleerd wordt door voedingsstoffen (via mTORC1) en het op cellulair

Therefore, we hypothesise that C/EBPβ-LIP is an important mediator of the diet to regulate body functions and a reduction of C/EBPβ-LIP level by pharmaceuticals might

Ich möchte dir auch danken für die vielen Diskussionen und spitzen Kommentare zu vielen verschieden Sachen (sowas wie „Wie du warst noch nie bei IKEA? Du weist aber

MTORC1 is known to be involved in the regulation of cancer cell survival, proliferation, protein biosynthesis and cellular metabolism 47.. In order to stimulate mRNA

1) C/EBPβ–LIP translation is controlled by the nutrient sensing kinase complex mTORC1 and deficiency in LIP expression results in metabolic improvements similar