100 results on '"Westendorp B"'
Search Results
2. Mesenchymal stromal/stem cells promote intestinal epithelium regeneration after chemotherapy-induced damage
- Author
-
Yetkin-Arik, B, Jansen, S A, Varderidou-Minasian, S, Westendorp, B, Skarp, K-P, Altelaar, M, Lindemans, C A, Lorenowicz, M J, Yetkin-Arik, B, Jansen, S A, Varderidou-Minasian, S, Westendorp, B, Skarp, K-P, Altelaar, M, Lindemans, C A, and Lorenowicz, M J
- Abstract
BACKGROUND: Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for leukemia and a range of non-malignant disorders. The success of the therapy is hampered by occurrence of acute graft-versus-host disease (aGvHD); an inflammatory response damaging recipient organs, with gut, liver, and skin being the most susceptible. Intestinal GvHD injury is often a life-threatening complication in patients unresponsive to steroid treatment. Allogeneic mesenchymal stromal/stem cell (MSC) infusions are a promising potential treatment for steroid-resistant aGvHD. Data from our institution and others demonstrate rescue of approximately 40-50% of aGvHD patients with MSCs in Phase I, II studies and minor side effects. Although promising, better understanding of MSC mode of action and patient response to MSC-based therapy is essential to improve this lifesaving treatment.METHODS: Single cell human small intestine organoids were embedded in Matrigel, grown for 5 days and treated with busulfan for 48 h. Organoids damaged by treatment with busulfan or control organoids were co-cultured with 5000, 10,000, and 50,000 MSCs for 24 h, 48 h or 7 days and the analyses such as surface area determination, proliferation and apoptosis assessment, RNA sequencing and proteomics were performed.RESULTS: Here, we developed a 3D co-culture model of human small intestinal organoids and MSCs, which allows to study the regenerative effects of MSCs on intestinal epithelium in a more physiologically relevant setting than existing in vitro systems. Using this model we mimicked chemotherapy-mediated damage of the intestinal epithelium. The treatment with busulfan, the chemotherapeutic commonly used as conditioning regiment before the HSCT, affected pathways regulating epithelial to mesenchymal transition, proliferation, and apoptosis in small intestinal organoids, as shown by transcriptomic and proteomic analysis. The co-culture of busulfan-treated intestinal organoi
- Published
- 2024
3. Mesenchymal stromal/stem cells promote intestinal epithelium regeneration after chemotherapy-induced damage
- Author
-
MS Algemene Pediatrie Onderzoek 2, MS Neonatologie, SCT patientenzorg, Child Health, Infection & Immunity, Regenerative Medicine and Stem Cells, Cancer, CMM Groep Coffer, CMM, Yetkin-Arik, B., Jansen, S. A., Varderidou-Minasian, S., Westendorp, B., Skarp, K. P., Altelaar, M., Lindemans, C. A., Lorenowicz, M. J., MS Algemene Pediatrie Onderzoek 2, MS Neonatologie, SCT patientenzorg, Child Health, Infection & Immunity, Regenerative Medicine and Stem Cells, Cancer, CMM Groep Coffer, CMM, Yetkin-Arik, B., Jansen, S. A., Varderidou-Minasian, S., Westendorp, B., Skarp, K. P., Altelaar, M., Lindemans, C. A., and Lorenowicz, M. J.
- Published
- 2024
4. Mesenchymal stromal/stem cells promote intestinal epithelium regeneration after chemotherapy-induced damage
- Author
-
Pathobiologie, Cell Biology, Metabolism and Cancer, Afd Biomol.Mass Spect. and Proteomics, Cell Biology, Metabolism & Cancer - Cancer, Yetkin-Arik, B, Jansen, S A, Varderidou-Minasian, S, Westendorp, B, Skarp, K-P, Altelaar, M, Lindemans, C A, Lorenowicz, M J, Pathobiologie, Cell Biology, Metabolism and Cancer, Afd Biomol.Mass Spect. and Proteomics, Cell Biology, Metabolism & Cancer - Cancer, Yetkin-Arik, B, Jansen, S A, Varderidou-Minasian, S, Westendorp, B, Skarp, K-P, Altelaar, M, Lindemans, C A, and Lorenowicz, M J
- Published
- 2024
5. Indian Hedgehog Suppresses a Stromal Cell–Driven Intestinal Immune Response
- Author
-
Westendorp, B. Florien, Büller, Nikè V.J.A., Karpus, Olga N., van Dop, Willemijn A., Koster, Jan, Versteeg, Rogier, Koelink, Pim J., Snel, Clinton Y., Meisner, Sander, Roelofs, Joris J.T.H., Uhmann, Anja, Ver Loren van Themaat, Emiel, Heijmans, Jarom, Hahn, Heidi, Muncan, Vanesa, Wildenberg, Manon E., and van den Brink, Gijs R.
- Published
- 2018
- Full Text
- View/download PDF
6. Oncogenic K-Ras Turns Death Receptors Into Metastasis-Promoting Receptors in Human and Mouse Colorectal Cancer Cells
- Author
-
Hoogwater, Frederik J.H., Nijkamp, Maarten W., Smakman, Niels, Steller, Ernst J.A., Emmink, Benjamin L., Westendorp, B. Florien, Raats, Danielle A.E., Sprick, Martin R., Schaefer, Uta, Van Houdt, Winan J., De Bruijn, Menno T., Schackmann, Ron C.J., Derksen, Patrick W.B., Medema, Jan–Paul, Walczak, Henning, Borel Rinkes, Inne H.M., and Kranenburg, Onno
- Published
- 2010
- Full Text
- View/download PDF
7. Discovery of Tissue-Specific Functions of Atypical E2Fs in Cancer
- Author
-
Bruin, A. de, Westendorp, B., Moreno Iglesias, Eva, Bruin, A. de, Westendorp, B., and Moreno Iglesias, Eva
- Published
- 2021
8. Synergistic functions of E2F7 and E2F8 are critical to suppress stress-induced skin cancer
- Author
-
Thurlings, I, Martínez-López, L M, Westendorp, B, Zijp, M, Kuiper, R, Tooten, P, Kent, L N, Leone, G, Vos, H J, Burgering, B, de Bruin, A, LS Pathobiologie, Dep Gezondheidszorg Landbouwhuisdieren, dPB RMSC, LS Pathobiologie, Dep Gezondheidszorg Landbouwhuisdieren, and dPB RMSC
- Subjects
Keratinocytes ,0301 basic medicine ,Cancer Research ,Skin Neoplasms ,GENES ,DNA damage ,DNA-BINDING DOMAINS ,Apoptosis ,PROGRESSION ,Biology ,medicine.disease_cause ,CELL-PROLIFERATION ,Mice ,03 medical and health sciences ,0302 clinical medicine ,E2F7 Transcription Factor ,Genetics ,medicine ,Animals ,Humans ,E2F1 ,TRANSCRIPTION ,E2F ,Molecular Biology ,ATYPICAL E2FS ,Mice, Knockout ,DAMAGE ,CARCINOGENESIS ,Cell cycle ,medicine.disease ,Repressor Proteins ,030104 developmental biology ,medicine.anatomical_structure ,TARGET ,030220 oncology & carcinogenesis ,FAMILY-MEMBER ,Immunology ,Cancer research ,Original Article ,Skin cancer ,biological phenomena, cell phenomena, and immunity ,Keratinocyte ,Carcinogenesis ,E2F Transcription Factors ,DNA Damage - Abstract
E2F transcription factors are important regulators of the cell cycle, and unrestrained activation of E2F-dependent transcription is considered to be an important driver of tumor formation and progression. Although highly expressed in normal skin and skin cancer, the role of the atypical E2Fs, E2F7 and E2F8, in keratinocyte homeostasis, regeneration and tumorigenesis is unknown. Surprisingly, keratinocyte-specific deletion of E2F7 and E2F8 in mice did not interfere with skin development and wound healing. However, the rate for successful isolation and establishment of E2f7/8-deficient primary keratinocyte cultures was much higher than for wild-type keratinocytes. Moreover, E2f7/8-deficient primary keratinocytes proliferate more efficiently under stress conditions, such as low/high confluence or DNA damage. Application of in vivo stress using the DMBA/TPA skin carcinogenesis protocol revealed that combined inactivation of E2f7/8 enhanced tumorigenesis and accelerated malignant progression. Loss of atypical E2Fs resulted in increased expression of E2F target genes, including E2f1. Additional loss of E2f1 did not rescue, but worsened skin tumorigenesis. We show that loss of E2F7/8 triggers apoptosis via induction of E2F1 in response to stress, indicating that the tumor-promoting effect of E2F7/8 inactivation can be partially compensated via E2F1-dependent apoptosis. Importantly, E2F7/8 repressed a large set of E2F target genes that are highly expressed in human patients with skin cancer. Together, our studies demonstrate that atypical E2Fs act as tumor suppressors, most likely via transcriptional repression of cell cycle genes in response to stress.
- Published
- 2017
9. Chk1 and 14‐3‐3 proteins inhibit atypical E2Fs to prevent a permanent cell cycle arrest
- Author
-
Yuan, R., Vos, Harmjan, van Es, Robert M, Chen, J., Burgering, B.M.T., Westendorp, B., de Bruin, A., LS Pathobiologie, Afd Pharmacology, dPB RMSC, LS Pathobiologie, Afd Pharmacology, and dPB RMSC
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,RECOMBINATION ,Apoptosis ,PROGRESSION ,medicine.disease_cause ,0302 clinical medicine ,E2F7 Transcription Factor ,Neoplasms ,TRANSCRIPTION ,Phosphorylation ,S-CHECKPOINT ,Promoter Regions, Genetic ,DNAdamage ,General Neuroscience ,Articles ,Cell cycle ,3. Good health ,Cell biology ,030220 oncology & carcinogenesis ,cell cycle ,biological phenomena, cell phenomena, and immunity ,Protein Binding ,DNA Replication ,Cell Survival ,Cdc25 ,14‐3‐3 proteins ,DNA-DAMAGE RESPONSE ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,REPLICATION-STRESS ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Humans ,BREAST-CANCER ,CHEK1 ,E2F ,Molecular Biology ,REPAIR ,CDC25 ,General Immunology and Microbiology ,checkpoint kinase 1 ,DNA Replication, Repair & Recombination ,14-3-3 proteins ,Promoter ,Cell Cycle Checkpoints ,atypical E2Fs ,Repressor Proteins ,HEK293 Cells ,030104 developmental biology ,Protein Biosynthesis ,Cancer cell ,biology.protein ,DNA damage ,Carcinogenesis ,HeLa Cells - Abstract
The atypical E2Fs, E2F7 and E2F8, act as potent transcriptional repressors of DNA replication genes providing them with the ability to induce a permanent S‐phase arrest and suppress tumorigenesis. Surprisingly in human cancer, transcript levels of atypical E2Fs are frequently elevated in proliferating cancer cells, suggesting that the tumor suppressor functions of atypical E2Fs might be inhibited through unknown post‐translational mechanisms. Here, we show that atypical E2Fs can be directly phosphorylated by checkpoint kinase 1 (Chk1) to prevent a permanent cell cycle arrest. We found that 14‐3‐3 protein isoforms interact with both E2Fs in a Chk1‐dependent manner. Strikingly, Chk1 phosphorylation and 14‐3‐3‐binding did not relocate or degrade atypical E2Fs, but instead, 14‐3‐3 is recruited to E2F7/8 target gene promoters to possibly interfere with transcription. We observed that high levels of 14‐3‐3 strongly correlate with upregulated transcription of atypical E2F target genes in human cancer. Thus, we reveal that Chk1 and 14‐3‐3 proteins cooperate to inactivate the transcriptional repressor functions of atypical E2Fs. This mechanism might be of particular importance to cancer cells, since they are exposed frequently to DNA‐damaging therapeutic reagents.
- Published
- 2018
10. Colonic CD90+ Crypt Fibroblasts Secrete Semaphorins to Support Epithelial Growth
- Author
-
Karpus, Olga N., primary, Westendorp, B. Florien, additional, Vermeulen, Jacqueline L.M., additional, Meisner, Sander, additional, Koster, Jan, additional, Muncan, Vanesa, additional, Wildenberg, Manon E., additional, and van den Brink, Gijs R., additional
- Published
- 2019
- Full Text
- View/download PDF
11. Atypical E2Fs inhibit tumor angiogenesis
- Author
-
Weijts, B G M W, Westendorp, B, Hien, B T, Martínez-López, L M, Zijp, M, Thurlings, I, Thomas, R E, Schulte-Merker, Stefan, Bakker, W J, de Bruin, A, Weijts, B G M W, Westendorp, B, Hien, B T, Martínez-López, L M, Zijp, M, Thurlings, I, Thomas, R E, Schulte-Merker, Stefan, Bakker, W J, and de Bruin, A
- Abstract
Atypical E2F transcription factors (E2F7 and E2F8) function as key regulators of cell cycle progression and their inactivation leads to spontaneous cancer formation in mice. However, the mechanism of the tumor suppressor functions of E2F7/8 remain obscure. In this study we discovered that atypical E2Fs control tumor angiogenesis, one of the hallmarks of cancer. We genetically inactivated atypical E2Fs in epithelial and mesenchymal neoplasm and analyzed blood vessel formation in three different animal models of cancer. Tumor formation was either induced by application of 7,12-Dimethylbenz(a)anthracene/12-O-Tetradecanoylphorbol-13-acetate or by Myc/Ras overexpression. To our surprise, atypical E2Fs suppressed tumor angiogenesis in all three cancer models, which is in a sharp contrast to previous findings showing that atypical E2Fs promote angiogenesis during fetal development in mice and zebrafish. Real-time imaging in zebrafish displayed that fluorescent-labeled blood vessels showed enhanced intratumoral branching in xenografted E2f7/8-deficient neoplasms compared with E2f7/8-proficient neoplasms. DLL4 expression, a key negative inhibitor of vascular branching, was decreased in E2f7/8-deficient neoplastic cells, indicating that E2F7/8 might inhibit intratumoral vessel branching via induction of DLL4.Oncogene advance online publication, 18 September 2017; doi:10.1038/onc.2017.336.
- Published
- 2018
12. Atypical E2Fs inhibit tumor angiogenesis
- Author
-
Weijts, B.G.M.W., Westendorp, B., Hien, B. T., Martínez-López, L. M., van Zijp, H.M., Thurlings, I., Thomas, R. E., Schulte-Merker, S., Bakker, W. J., de Bruin, A., Weijts, B.G.M.W., Westendorp, B., Hien, B. T., Martínez-López, L. M., van Zijp, H.M., Thurlings, I., Thomas, R. E., Schulte-Merker, S., Bakker, W. J., and de Bruin, A.
- Published
- 2018
13. Chk1 and 14-3-3 proteins inhibit atypical E2Fs to prevent a permanent cell cycle arrest
- Author
-
LS Pathobiologie, Afd Pharmacology, dPB RMSC, Yuan, R., Vos, Harmjan, van Es, Robert M, Chen, J., Burgering, B.M.T., Westendorp, B., de Bruin, A., LS Pathobiologie, Afd Pharmacology, dPB RMSC, Yuan, R., Vos, Harmjan, van Es, Robert M, Chen, J., Burgering, B.M.T., Westendorp, B., and de Bruin, A.
- Published
- 2018
14. Atypical E2Fs inhibit tumor angiogenesis
- Author
-
LS Pathobiologie, dPB RMSC, Weijts, B G M W, Westendorp, B, Hien, B T, Martínez-López, L M, Zijp, M, Thurlings, I, Thomas, R E, Schulte-Merker, Stefan, Bakker, W J, de Bruin, A, LS Pathobiologie, dPB RMSC, Weijts, B G M W, Westendorp, B, Hien, B T, Martínez-López, L M, Zijp, M, Thurlings, I, Thomas, R E, Schulte-Merker, Stefan, Bakker, W J, and de Bruin, A
- Published
- 2018
15. Atypical E2Fs inhibit tumor angiogenesis.
- Author
-
Weijts, BGMW, Westendorp, B, Hien, BT, Martínez-López, LM, Zijp, M, Thurlings, I, Thomas, RE, Schulte-Merker, S, Bakker, WJ, de Bruin, A, Weijts, BGMW, Westendorp, B, Hien, BT, Martínez-López, LM, Zijp, M, Thurlings, I, Thomas, RE, Schulte-Merker, S, Bakker, WJ, and de Bruin, A
- Abstract
Atypical E2F transcription factors (E2F7 and E2F8) function as key regulators of cell cycle progression and their inactivation leads to spontaneous cancer formation in mice. However, the mechanism of the tumor suppressor functions of E2F7/8 remain obscure. In this study we discovered that atypical E2Fs control tumor angiogenesis, one of the hallmarks of cancer. We genetically inactivated atypical E2Fs in epithelial and mesenchymal neoplasm and analyzed blood vessel formation in three different animal models of cancer. Tumor formation was either induced by application of 7,12-Dimethylbenz(a)anthracene/12-O-Tetradecanoylphorbol-13-acetate or by Myc/Ras overexpression. To our surprise, atypical E2Fs suppressed tumor angiogenesis in all three cancer models, which is in a sharp contrast to previous findings showing that atypical E2Fs promote angiogenesis during fetal development in mice and zebrafish. Real-time imaging in zebrafish displayed that fluorescent-labeled blood vessels showed enhanced intratumoral branching in xenografted E2f7/8-deficient neoplasms compared with E2f7/8-proficient neoplasms. DLL4 expression, a key negative inhibitor of vascular branching, was decreased in E2f7/8-deficient neoplastic cells, indicating that E2F7/8 might inhibit intratumoral vessel branching via induction of DLL4.
- Published
- 2018
16. Atypical E2Fs inhibit tumor angiogenesis
- Author
-
Cancer, Hubrecht Institute with UMC, Weijts, B.G.M.W., Westendorp, B., Hien, B. T., Martínez-López, L. M., van Zijp, H.M., Thurlings, I., Thomas, R. E., Schulte-Merker, S., Bakker, W. J., de Bruin, A., Cancer, Hubrecht Institute with UMC, Weijts, B.G.M.W., Westendorp, B., Hien, B. T., Martínez-López, L. M., van Zijp, H.M., Thurlings, I., Thomas, R. E., Schulte-Merker, S., Bakker, W. J., and de Bruin, A.
- Published
- 2018
17. Heterozygosity of Chaperone Grp78 Reduces Intestinal Stem Cell Regeneration Potential and Protects against Adenoma Formation
- Author
-
van Lidth de Jeude, Jooske F., primary, Spaan, Claudia N., additional, Meijer, Bartolomeus J., additional, Smit, Wouter L., additional, Soeratram, Tanya T.D., additional, Wielenga, Mattheus C.B., additional, Westendorp, B. Florien, additional, Lee, Amy S., additional, Meisner, Sander, additional, Vermeulen, Jacqueline L.M., additional, Wildenberg, Manon E., additional, van den Brink, Gijs R., additional, Muncan, Vanesa, additional, and Heijmans, Jarom, additional
- Published
- 2018
- Full Text
- View/download PDF
18. Physiological significance of polyploidization in mammalian cells
- Author
-
Pandit, S.K., Westendorp, B., de Bruin, A., Tissue Repair, Dep Pathobiologie, Tissue Repair, and Dep Pathobiologie
- Subjects
Genetics ,Physiological significance ,genetic processes ,fungi ,food and beverages ,pathological conditions, signs and symptoms ,Cell Biology ,Disease ,Chronic injury ,Biology ,Phenotype ,Cell size ,Cell biology ,Polyploidy ,Mice ,Biological property ,Hepatocytes ,Animals ,Humans ,Adaptation ,E2F ,Cell Size - Abstract
Programmed polyploidization occurs in all mammalian species during development and aging in selected tissues, but the biological properties of polyploid cells remain obscure. Spontaneous polyploidization arises during stress and has been observed in a variety of pathological conditions, such as cancer and degenerative diseases. A major challenge in the field is to test the predicted functions of polyploidization in vivo. However, recent genetic mouse models with diminished polyploidization phenotypes represent novel, powerful tools to unravel the biological function of polyploidization. Contrary to a longstanding hypothesis, polyploidization appears to not be required for differentiation and has no obvious impact on proliferation. Instead, polyploidization leads to increased cell size and genetic diversity, which could promote better adaptation to chronic injury or stress. We discuss here the consequences of reducing polyploidization in mice and review which stress responses and molecular signals trigger polyploidization during development and disease.
- Published
- 2013
19. Synergistic functions of E2F7 and E2F8 are critical to suppress stress-induced skin cancer
- Author
-
LS Pathobiologie, Dep Gezondheidszorg Landbouwhuisdieren, dPB RMSC, Thurlings, I, Martínez-López, L M, Westendorp, B, Zijp, M, Kuiper, R, Tooten, P, Kent, L N, Leone, G, Vos, H J, Burgering, B, de Bruin, A, LS Pathobiologie, Dep Gezondheidszorg Landbouwhuisdieren, dPB RMSC, Thurlings, I, Martínez-López, L M, Westendorp, B, Zijp, M, Kuiper, R, Tooten, P, Kent, L N, Leone, G, Vos, H J, Burgering, B, and de Bruin, A
- Published
- 2017
20. Atypical E2Fs inhibit tumor angiogenesis
- Author
-
Weijts, B G M W, primary, Westendorp, B, additional, Hien, B T, additional, Martínez-López, L M, additional, Zijp, M, additional, Thurlings, I, additional, Thomas, R E, additional, Schulte-Merker, S, additional, Bakker, W J, additional, and de Bruin, A, additional
- Published
- 2017
- Full Text
- View/download PDF
21. Synergistic functions of E2F7 and E2F8 are critical to suppress stress-induced skin cancer
- Author
-
Thurlings, I, primary, Martínez-López, L M, additional, Westendorp, B, additional, Zijp, M, additional, Kuiper, R, additional, Tooten, P, additional, Kent, L N, additional, Leone, G, additional, Vos, H J, additional, Burgering, B, additional, and de Bruin, A, additional
- Published
- 2016
- Full Text
- View/download PDF
22. Stromal Indian hedgehog signaling is required for intestinal adenoma formation in mice
- Author
-
Büller, Nikè V J A, Rosekrans, Sanne L., Metcalfe, Ciara, Heijmans, Jarom, Van Dop, Willemijn A., Fessler, Evelyn, Jansen, Marnix, Ahn, Christina, Vermeulen, Jacqueline L M, Westendorp, B. Florien, Robanus-Maandag, Els C., Offerhaus, G. Johan, Medema, Jan Paul, D'Haens, Geert R A M, Wildenberg, Manon E., De Sauvage, Frederic J., Muncan, Vanesa, Van Den Brink, Gijs R., Büller, Nikè V J A, Rosekrans, Sanne L., Metcalfe, Ciara, Heijmans, Jarom, Van Dop, Willemijn A., Fessler, Evelyn, Jansen, Marnix, Ahn, Christina, Vermeulen, Jacqueline L M, Westendorp, B. Florien, Robanus-Maandag, Els C., Offerhaus, G. Johan, Medema, Jan Paul, D'Haens, Geert R A M, Wildenberg, Manon E., De Sauvage, Frederic J., Muncan, Vanesa, and Van Den Brink, Gijs R.
- Published
- 2015
23. Stromal Indian hedgehog signaling is required for intestinal adenoma formation in mice
- Author
-
Pathologie Pathologen staf, Cancer, Büller, Nikè V J A, Rosekrans, Sanne L., Metcalfe, Ciara, Heijmans, Jarom, Van Dop, Willemijn A., Fessler, Evelyn, Jansen, Marnix, Ahn, Christina, Vermeulen, Jacqueline L M, Westendorp, B. Florien, Robanus-Maandag, Els C., Offerhaus, G. Johan, Medema, Jan Paul, D'Haens, Geert R A M, Wildenberg, Manon E., De Sauvage, Frederic J., Muncan, Vanesa, Van Den Brink, Gijs R., Pathologie Pathologen staf, Cancer, Büller, Nikè V J A, Rosekrans, Sanne L., Metcalfe, Ciara, Heijmans, Jarom, Van Dop, Willemijn A., Fessler, Evelyn, Jansen, Marnix, Ahn, Christina, Vermeulen, Jacqueline L M, Westendorp, B. Florien, Robanus-Maandag, Els C., Offerhaus, G. Johan, Medema, Jan Paul, D'Haens, Geert R A M, Wildenberg, Manon E., De Sauvage, Frederic J., Muncan, Vanesa, and Van Den Brink, Gijs R.
- Published
- 2015
24. E2F7 and E2F8 promote angiogenesis through transcriptional activation of VEGFA in cooperation with HIF1
- Author
-
Weijts, B.G.M.W., Bakker, W.J., Cornelissen, P.W.A., Liang, K., Schaftenaar, F.H., Westendorp, B., de Wolf, C.A.C.M.T., Paciejewska, M., Scheele, C.L.G.J., Kent, L., Leone, G., Schulte-Merker, S., de Bruin, A., and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
embryonic-development ,proliferation ,growth-factor gene ,zebrafish ,hif-1-alpha ,vascular development ,o-2 homeostasis ,hypoxia-inducible factor-1 ,Experimentele Zoologie ,WIAS ,cancer ,Experimental Zoology ,factor 1-alpha - Abstract
The E2F family of transcription factors plays an important role in controlling cell-cycle progression. While this is their best-known function, we report here novel functions for the newest members of the E2F family, E2F7 and E2F8 (E2F7/8). We show that simultaneous deletion of E2F7/8 in zebrafish and mice leads to severe vascular defects during embryonic development. Using a panel of transgenic zebrafish with fluorescent-labelled blood vessels, we demonstrate that E2F7/8 are essential for proper formation of blood vessels. Despite their classification as transcriptional repressors, we provide evidence for a molecular mechanism through which E2F7/8 activate the transcription of the vascular endothelial growth factor A (VEGFA), a key factor in guiding angiogenesis. We show that E2F7/8 directly bind and stimulate the VEGFA promoter independent of canonical E2F binding elements. Instead, E2F7/8 form a transcriptional complex with the hypoxia inducible factor 1 (HIF1) to stimulate VEGFA promoter activity. These results uncover an unexpected link between E2F7/8 and the HIF1-VEGFA pathway providing a molecular mechanism by which E2F7/8 control angiogenesis.
- Published
- 2012
25. E2F7 represses a network of oscillating cell cycle genes to control S-phase progression
- Author
-
Westendorp, B., Mokry, M., Groot Koerkamp, M.J., Holstege, F.C.P., Cuppen, E.P.J.G., de Bruin, A., Tissue Repair, Dep Pathobiologie, Tissue Repair, Dep Pathobiologie, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Periodicity ,Transcription, Genetic ,DNA repair ,Biology ,S Phase ,Mice ,E2F7 Transcription Factor ,Transcription (biology) ,Consensus Sequence ,Genetics ,Animals ,Gene Regulatory Networks ,E2F ,Gene ,Binding Sites ,Base Sequence ,DNA replication ,G1 Phase ,Promoter ,Genomics ,Cell Cycle Gene ,Molecular biology ,Genes, cdc ,Repressor Proteins ,Gene Expression Regulation ,S Phase Cell Cycle Checkpoints ,E2F Transcription Factors ,DNA Damage - Abstract
E2F transcription factors are known to be important for timely activation of G 1 /S and G 2 /M genes required for cell cycle progression, but transcriptional mechanisms for deactivation of cell cycle-regulated genes are unknown. Here, we show that E2F7 is highly expressed during mid to late S-phase, occupies promoters of G 1 /S-regulated genes and represses their transcription. ChIP-seq analysis revealed that E2F7 binds preferentially to genomic sites containing the TTCCCGCC motif, which closely resembles the E2F consensus site. We identified 89 target genes that carry E2F7 binding sites close to the transcriptional start site and that are directly repressed by short-term induction of E2F7. Most of these target genes are known to be activated by E2Fs and are involved in DNA replication, metabolism and DNA repair. Importantly, induction of E2F7 during G 0 -G 1 /S resulted in S-phase arrest and DNA damage, whereas expression of E2F7 during G 2 /M failed to disturb cell cycle progression. These findings provide strong evidence that E2F7 directly controls the downswing of oscillating G 1 /S genes during S-phase progression.
- Published
- 2012
26. Stromal Indian Hedgehog Signaling Is Required for Intestinal Adenoma Formation in Mice
- Author
-
Büller, Nikè V.J.A., primary, Rosekrans, Sanne L., additional, Metcalfe, Ciara, additional, Heijmans, Jarom, additional, van Dop, Willemijn A., additional, Fessler, Evelyn, additional, Jansen, Marnix, additional, Ahn, Christina, additional, Vermeulen, Jacqueline L.M., additional, Westendorp, B. Florien, additional, Robanus-Maandag, Els C., additional, Offerhaus, G. Johan, additional, Medema, Jan Paul, additional, D’Haens, Geert R.A.M., additional, Wildenberg, Manon E., additional, de Sauvage, Frederic J., additional, Muncan, Vanesa, additional, and van den Brink, Gijs R., additional
- Published
- 2015
- Full Text
- View/download PDF
27. Physiological significance of polyploidization in mammalian cells
- Author
-
Tissue Repair, Dep Pathobiologie, Pandit, S.K., Westendorp, B., de Bruin, A., Tissue Repair, Dep Pathobiologie, Pandit, S.K., Westendorp, B., and de Bruin, A.
- Published
- 2013
28. HIF proteins connect the RB-E2F factors to angiogenesis
- Author
-
Tissue Repair, Dep Pathobiologie, Bakker, W.J., Weijts, B.G.M.W., Westendorp, B., de Bruin, A., Tissue Repair, Dep Pathobiologie, Bakker, W.J., Weijts, B.G.M.W., Westendorp, B., and de Bruin, A.
- Published
- 2013
29. The E2F6 repressor activates gene expression in myocardium resulting in dilated cardiomyopathy
- Author
-
Tissue Repair, Dep Pathobiologie, Westendorp, B., Major, J.L., Nader, M., Salih, M., Leenen, F.H., Tuana, B.S., Tissue Repair, Dep Pathobiologie, Westendorp, B., Major, J.L., Nader, M., Salih, M., Leenen, F.H., and Tuana, B.S.
- Published
- 2012
30. E2F7 represses a network of oscillating cell cycle genes to control S-phase progression.
- Author
-
Tissue Repair, Dep Pathobiologie, Westendorp, B., Mokry, M., Groot Koerkamp, M.J., Holstege, F.C.P., Cuppen, E.P.J.G., de Bruin, A., Tissue Repair, Dep Pathobiologie, Westendorp, B., Mokry, M., Groot Koerkamp, M.J., Holstege, F.C.P., Cuppen, E.P.J.G., and de Bruin, A.
- Published
- 2012
31. E2F7 and E2F8 promote angiogenesis through transcriptional activation of VEGFA in cooperation with HIF1
- Author
-
Weijts, B.G., Bakker, W.J., Cornelissen, P.W., Liang, K.H., Schaftenaar, F.H., Westendorp, B., de Wolf, C.A., Paciejewska, M., Scheele, C.L., Kent, L., Leone, G., Schulte-Merker, S., de Bruin, A., Weijts, B.G., Bakker, W.J., Cornelissen, P.W., Liang, K.H., Schaftenaar, F.H., Westendorp, B., de Wolf, C.A., Paciejewska, M., Scheele, C.L., Kent, L., Leone, G., Schulte-Merker, S., and de Bruin, A.
- Abstract
The E2F family of transcription factors plays an important role in controlling cell-cycle progression. While this is their best-known function, we report here novel functions for the newest members of the E2F family, E2F7 and E2F8 (E2F7/8). We show that simultaneous deletion of E2F7/8 in zebrafish and mice leads to severe vascular defects during embryonic development. Using a panel of transgenic zebrafish with fluorescent-labelled blood vessels, we demonstrate that E2F7/8 are essential for proper formation of blood vessels. Despite their classification as transcriptional repressors, we provide evidence for a molecular mechanism through which E2F7/8 activate the transcription of the vascular endothelial growth factor A (VEGFA), a key factor in guiding angiogenesis. We show that E2F7/8 directly bind and stimulate the VEGFA promoter independent of canonical E2F binding elements. Instead, E2F7/8 form a transcriptional complex with the hypoxia inducible factor 1 (HIF1) to stimulate VEGFA promoter activity. These results uncover an unexpected link between E2F7/8 and the HIF1-VEGFA pathway providing a molecular mechanism by which E2F7/8 control angiogenesis., The E2F family of transcription factors plays an important role in controlling cell-cycle progression. While this is their best-known function, we report here novel functions for the newest members of the E2F family, E2F7 and E2F8 (E2F7/8). We show that simultaneous deletion of E2F7/8 in zebrafish and mice leads to severe vascular defects during embryonic development. Using a panel of transgenic zebrafish with fluorescent-labelled blood vessels, we demonstrate that E2F7/8 are essential for proper formation of blood vessels. Despite their classification as transcriptional repressors, we provide evidence for a molecular mechanism through which E2F7/8 activate the transcription of the vascular endothelial growth factor A (VEGFA), a key factor in guiding angiogenesis. We show that E2F7/8 directly bind and stimulate the VEGFA promoter independent of canonical E2F binding elements. Instead, E2F7/8 form a transcriptional complex with the hypoxia inducible factor 1 (HIF1) to stimulate VEGFA promoter activity. These results uncover an unexpected link between E2F7/8 and the HIF1-VEGFA pathway providing a molecular mechanism by which E2F7/8 control angiogenesis.
- Published
- 2012
32. HIF proteins connect the RB-E2F factors to angiogenesis
- Author
-
Bakker, W.J., Weijts, B.G.M.W., Westendorp, B., de Bruin, A., Tissue Repair, and Dep Pathobiologie
- Subjects
Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Angiogenesis ,VEGF receptors ,Biology ,Biochemistry ,angiogenesis ,E2F ,Neoplasms ,Internal medicine ,Genetics ,medicine ,Transcriptional regulation ,HIF ,cancer ,Animals ,Humans ,Point of View ,Binding Sites ,Neovascularization, Pathologic ,hypoxia ,Cancer ,Hypoxia (medical) ,medicine.disease ,VEGF ,Embryonic stem cell ,E2F Transcription Factors ,Repressor Proteins ,Vascular endothelial growth factor A ,Receptors, Vascular Endothelial Growth Factor ,Endocrinology ,Cancer research ,biology.protein ,Hypoxia-Inducible Factor 1 ,medicine.symptom ,RB ,Biotechnology - Abstract
Recently, we showed that E2F7 and E2F8 (E2F7/8) are critical regulators of angiogenesis through transcriptional control of VEGFA in cooperation with HIF. (1) Here we investigate the existence of other novel putative angiogenic E2F7/8-HIF targets, and discuss the role of the RB-E2F pathway in regulating angiogenesis during embryonic and tumor development.
- Published
- 2013
33. Discovery of Tissue-Specific Functions of Atypical E2Fs in Cancer
- Author
-
Eva Moreno Iglesias, Bruin, A. de, Westendorp, B., and University Utrecht
- Subjects
Genetically modified mouse ,stomatognathic diseases ,business.industry ,medicine ,Cancer research ,Tissue specific ,Cancer ,biological phenomena, cell phenomena, and immunity ,Liver cancer ,medicine.disease ,business ,atypical E2Fs ,RB ,transgenic mice ,Polyploidization ,liver cancer ,NAFLD - Abstract
The cell cycle control system ensures that the genome is properly duplicated and evenly distributed over the daughter cells. This system is highly conserved and regulated by a plethora of genes. Among this regulatory system is the Rb-E2F network, which controls the expression of key cell cycle genes. Thus, it is not surprising that Rb is often lost or mutated in cancer, resulting in hyperactivated E2F transcription and enhanced tumorigenesis. To counteract enhanced E2F transcription, alternative mechanisms that inhibit E2F transcription to efficiently block proliferation of cancer cells are needed. In this thesis, we manipulated E2F-dependent transcription by altering the expression of E2F7 and E2F8 in mouse models. E2F7/8 are atypical cell cycle inhibitors belonging to the E2F family that possess Rb-independent repressive function. We hypothesized that overactivation of atypical E2F repressors efficiently blocks cancer cell proliferation. The studies from this thesis demonstrated that downregulation of E2F transcription by boosting E2F7/8 expression impaired the ability of liver cancer cells to progress through the cell cycle and suppressed tumor growth. In addition, unrestrained E2F dependent transcription due to loss of atypical E2Fs and RB, induced tumorigenesis in a tissue cell-type specific manner via a mechanism that involves maintenance of genomic instability. Lastly, we unravel novel functions of atypical E2Fs in lipid metabolism by controlling liver polyploidization. Taken together, this thesis has contributed to our understanding of the role of atypical E2Fs and Rb in cancer and provides scientific evidence that inhibition of E2F-depedent transcription could be a novel therapeutic option in oncology .
- Published
- 2021
34. Assessment of Kinome-Wide Activity Remodeling upon Picornavirus Infection.
- Author
-
Veth TS, Nouwen LV, Zwaagstra M, Lyoo H, Wierenga KA, Westendorp B, Altelaar MAFM, Berkers C, van Kuppeveld FJM, and Heck AJR
- Subjects
- Humans, HeLa Cells, Proteome metabolism, Protein Kinases metabolism, Virus Replication, Phosphorylation, Picornaviridae physiology, Picornaviridae enzymology, Picornaviridae Infections virology, Picornaviridae Infections metabolism
- Abstract
Picornaviridae represent a large family of single-stranded positive RNA viruses of which different members can infect both humans and animals. These include the enteroviruses (e.g., poliovirus, coxsackievirus, and rhinoviruses) as well as the cardioviruses (e.g., encephalomyocarditis virus). Picornaviruses have evolved to interact with, use, and/or evade cellular host systems to create the optimal environment for replication and spreading. It is known that viruses modify kinase activity during infection, but a proteome-wide overview of the (de)regulation of cellular kinases during picornavirus infection is lacking. To study the kinase activity landscape during picornavirus infection, we here applied dedicated targeted mass spectrometry-based assays covering ∼40% of the human kinome. Our data show that upon infection, kinases of the MAPK pathways become activated (e.g., ERK1/2, RSK1/2, JNK1/2/3, and p38), while kinases involved in regulating the cell cycle (e.g., CDK1/2, GWL, and DYRK3) become inactivated. Additionally, we observed the activation of CHK2, an important kinase involved in the DNA damage response. Using pharmacological kinase inhibitors, we demonstrate that several of these activated kinases are essential for the replication of encephalomyocarditis virus. Altogether, the data provide a quantitative understanding of the regulation of kinome activity induced by picornavirus infection, providing a resource important for developing novel antiviral therapeutic interventions., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
35. MUC13 negatively regulates tight junction proteins and intestinal epithelial barrier integrity via protein kinase C.
- Author
-
Segui-Perez C, Stapels DAC, Ma Z, Su J, Passchier E, Westendorp B, Wubbolts RW, Wu W, van Putten JPM, and Strijbis K
- Subjects
- Humans, Intestines, Intestinal Mucosa metabolism, Tight Junctions metabolism, Occludin, Mucins metabolism, Epithelial Cells metabolism, Tight Junction Proteins metabolism, Protein Kinase C metabolism
- Abstract
Glycosylated mucin proteins contribute to the essential barrier function of the intestinal epithelium. The transmembrane mucin MUC13 is an abundant intestinal glycoprotein with important functions for mucosal maintenance that are not yet completely understood. We demonstrate that in human intestinal epithelial monolayers, MUC13 localized to both the apical surface and the tight junction (TJ) region on the lateral membrane. MUC13 deletion resulted in increased transepithelial resistance (TEER) and reduced translocation of small solutes. TEER buildup in ΔMUC13 cells could be prevented by addition of MLCK, ROCK or protein kinase C (PKC) inhibitors. The levels of TJ proteins including claudins and occludin were highly increased in membrane fractions of MUC13 knockout cells. Removal of the MUC13 cytoplasmic tail (CT) also altered TJ composition but did not affect TEER. The increased buildup of TJ complexes in ΔMUC13 and MUC13-ΔCT cells was dependent on PKC. The responsible PKC member might be PKCδ (or PRKCD) based on elevated protein levels in the absence of full-length MUC13. Our results demonstrate for the first time that a mucin protein can negatively regulate TJ function and stimulate intestinal barrier permeability., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2024. Published by The Company of Biologists Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
36. Editorial: The role of E2F transcription factors in cancer.
- Author
-
Iglesias-Ara A and Westendorp B
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
37. Glycosylated extracellular mucin domains protect against SARS-CoV-2 infection at the respiratory surface.
- Author
-
Chatterjee M, Huang LZX, Mykytyn AZ, Wang C, Lamers MM, Westendorp B, Wubbolts RW, van Putten JPM, Bosch BJ, Haagmans BL, and Strijbis K
- Subjects
- Humans, Angiotensin-Converting Enzyme 2, SARS-CoV-2 metabolism, CA-125 Antigen metabolism, Lung metabolism, Polysaccharides, Mucins metabolism, COVID-19
- Abstract
Mucins play an essential role in protecting the respiratory tract against microbial infections while also acting as binding sites for bacterial and viral adhesins. The heavily O-glycosylated gel-forming mucins MUC5AC and MUC5B eliminate pathogens by mucociliary clearance. Transmembrane mucins MUC1, MUC4, and MUC16 can restrict microbial invasion at the apical surface of the epithelium. In this study, we determined the impact of host mucins and mucin glycans on epithelial entry of SARS-CoV-2. Human lung epithelial Calu-3 cells express the SARS-CoV-2 entry receptor ACE2 and high levels of glycosylated MUC1, but not MUC4 and MUC16, on their cell surface. The O-glycan-specific mucinase StcE specifically removed the glycosylated part of the MUC1 extracellular domain while leaving the underlying SEA domain and cytoplasmic tail intact. StcE treatment of Calu-3 cells significantly enhanced infection with SARS-CoV-2 pseudovirus and authentic virus, while removal of terminal mucin glycans sialic acid and fucose from the epithelial surface did not impact viral entry. In Calu-3 cells, the transmembrane mucin MUC1 and ACE2 are located to the apical surface in close proximity and StcE treatment results in enhanced binding of purified spike protein. Both MUC1 and MUC16 are expressed on the surface of human organoid-derived air-liquid interface (ALI) differentiated airway cultures and StcE treatment led to mucin removal and increased levels of SARS-CoV-2 replication. In these cultures, MUC1 was highly expressed in non-ciliated cells while MUC16 was enriched in goblet cells. In conclusion, the glycosylated extracellular domains of different transmembrane mucins might have similar protective functions in different respiratory cell types by restricting SARS-CoV-2 binding and entry., Competing Interests: The authors declare no competing interests., (Copyright: © 2023 Chatterjee et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
38. Single cell analysis of docosahexaenoic acid suppression of sequential LPS-induced proinflammatory and interferon-regulated gene expression in the macrophage.
- Author
-
Wierenga KA, Riemers FM, Westendorp B, Harkema JR, and Pestka JJ
- Subjects
- Mice, Animals, Lipopolysaccharides pharmacology, Interferons metabolism, NF-kappa B metabolism, Single-Cell Analysis, Toll-Like Receptor 4 metabolism, Macrophages, Cytokines metabolism, Gene Expression, Docosahexaenoic Acids pharmacology, Docosahexaenoic Acids metabolism, Fatty Acids, Omega-3 metabolism
- Abstract
Preclinical and clinical studies suggest that consumption of long chain omega-3 polyunsaturated fatty acids (PUFAs) reduces severity of chronic inflammatory and autoimmune diseases. While these ameliorative effects are conventionally associated with downregulated expression of proinflammatory cytokine and chemokine genes, our laboratory has recently identified Type 1 interferon (IFN1)-regulated gene expression to be another key target of omega-3 PUFAs. Here we used single cell RNA sequencing (scRNAseq) to gain new mechanistic perspectives on how the omega-3 PUFA docosahexaenoic acid (DHA) influences TLR4-driven proinflammatory and IFN1-regulated gene expression in a novel self-renewing murine fetal liver-derived macrophage (FLM) model. FLMs were cultured with 25 µM DHA or vehicle for 24 h, treated with modest concentration of LPS (20 ng/ml) for 1 and 4 h, and then subjected to scRNAseq using the 10X Chromium System. At 0 h (i.e., in the absence of LPS), DHA increased expression of genes associated with the NRF2 antioxidant response (e.g. Sqstm1 , Hmox1, Chchd10 ) and metal homeostasis (e.g. Mt1 , Mt2, Ftl1, Fth1 ), both of which are consistent with DHA-induced polarization of FLMs to a more anti-inflammatory phenotype. At 1 h post-LPS treatment, DHA inhibited LPS-induced cholesterol synthesis genes (e.g. Scd1, Scd2 , Pmvk, Cyp51, Hmgcs1 , and Fdps) which potentially could contribute to interference with TLR4-mediated inflammatory signaling. At 4 h post-LPS treatment, LPS-treated FLMs reflected a more robust inflammatory response including upregulation of proinflammatory cytokine (e.g. Il1a, Il1b, Tnf ) and chemokine (e.g. Ccl2, Ccl3, Ccl4, Ccl7 ) genes as well as IFN1-regulated genes (e.g. Irf7, Mx1, Oasl1, Ifit1 ), many of which were suppressed by DHA. Using single-cell regulatory network inference and clustering (SCENIC) to identify gene expression networks, we found DHA modestly downregulated LPS-induced expression of NF-κB-target genes. Importantly, LPS induced a subset of FLMs simultaneously expressing NF-κB- and IRF7/STAT1/STAT2-target genes that were conspicuously absent in DHA-pretreated FLMs. Thus, DHA potently targeted both the NF-κB and the IFN1 responses. Altogether, scRNAseq generated a valuable dataset that provides new insights into multiple overlapping mechanisms by which DHA may transcriptionally or post-transcriptionally regulate LPS-induced proinflammatory and IFN1-driven responses in macrophages., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Wierenga, Riemers, Westendorp, Harkema and Pestka.)
- Published
- 2022
- Full Text
- View/download PDF
39. Collection of cells for single-cell RNA sequencing using high-resolution fluorescence microscopy.
- Author
-
Segeren HA, Andree KC, Oomens L, and Westendorp B
- Subjects
- Base Sequence genetics, Flow Cytometry methods, Gene Expression Profiling methods, High-Throughput Nucleotide Sequencing methods, Humans, RNA genetics, Sequence Analysis, RNA methods, Transcriptome genetics, Exome Sequencing methods, Workflow, Cell Separation methods, Microscopy, Fluorescence methods, Single-Cell Analysis methods
- Abstract
FACS sorting followed by single-cell RNA-sequencing (SORT-Seq) is a popular procedure to select cells of interest for single-cell transcriptomics. However, FACS is not suitable for measurement of subcellular distribution of fluorescence or for small samples (<1,000 cells). The VYCAP puncher system overcomes these limitations. Here, we describe a workflow to capture, image, and collect fluorescent human retina pigment epithelium cells for SORT-Seq using this system. The workflow can be used for any cell type with a diameter of ∼5-50 μm. For complete details on the use and execution of this protocol, please refer to Segeren et al. (2020)., Competing Interests: H.A.S. and B.W. declare no competing interests. L.O. and K.C.A. are employed by VYCAP B.V., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
40. Atypical E2Fs either Counteract or Cooperate with RB during Tumorigenesis Depending on Tissue Context.
- Author
-
Moreno E, Pandit SK, Toussaint MJM, Bongiovanni L, Harkema L, van Essen SC, van Liere EA, Westendorp B, and de Bruin A
- Abstract
E2F-transcription factors activate many genes involved in cell cycle progression, DNA repair, and apoptosis. Hence, E2F-dependent transcription must be tightly regulated to prevent tumorigenesis, and therefore metazoan cells possess multiple E2F regulation mechanisms. The best-known is the Retinoblastoma protein (RB), which is mutated in many cancers. Atypical E2Fs (E2F7 and -8) can repress E2F-target gene expression independently of RB and are rarely mutated in cancer. Therefore, they may act as emergency brakes in RB-mutated cells to suppress tumor growth. Currently, it is unknown if and how RB and atypical E2Fs functionally interact in vivo . Here, we demonstrate that mice with liver-specific combinatorial deletion of Rb and E2f7/8 have reduced life-spans compared to E2f7/8 or Rb deletion alone. This was associated with increased proliferation and enhanced malignant progression of liver tumors. Hence, atypical repressor E2Fs and RB cooperatively act as tumor suppressors in hepatocytes. In contrast, loss of either E2f7 or E2f8 largely prevented the formation of pituitary tumors in Rb
+/- mice. To test whether atypical E2Fs can also function as oncogenes independent of RB loss, we induced long-term overexpression of E2f7 or E2f8 in mice. E2F7 and -8 overexpression increased the incidence of tumors in the lungs, but not in other tissues. Collectively, these data show that atypical E2Fs can promote but also inhibit tumorigenesis depending on tissue type and RB status. We propose that the complex interactions between atypical E2Fs and RB on maintenance of genetic stability underlie this context-dependency.- Published
- 2021
- Full Text
- View/download PDF
41. PIDDosome-induced p53-dependent ploidy restriction facilitates hepatocarcinogenesis.
- Author
-
Sladky VC, Knapp K, Szabo TG, Braun VZ, Bongiovanni L, van den Bos H, Spierings DC, Westendorp B, Curinha A, Stojakovic T, Scharnagl H, Timelthaler G, Tsuchia K, Pinter M, Semmler G, Foijer F, de Bruin A, Reiberger T, Rohr-Udilova N, and Villunger A
- Subjects
- Animals, Carcinogenesis genetics, Humans, Mice, Ploidies, Tumor Suppressor Protein p53 genetics, Carcinoma, Hepatocellular genetics, Liver Neoplasms genetics
- Abstract
Polyploidization frequently precedes tumorigenesis but also occurs during normal development in several tissues. Hepatocyte ploidy is controlled by the PIDDosome during development and regeneration. This multi-protein complex is activated by supernumerary centrosomes to induce p53 and restrict proliferation of polyploid cells, otherwise prone for chromosomal instability. PIDDosome deficiency in the liver results in drastically increased polyploidy. To investigate PIDDosome-induced p53-activation in the pathogenesis of liver cancer, we chemically induced hepatocellular carcinoma (HCC) in mice. Strikingly, PIDDosome deficiency reduced tumor number and burden, despite the inability to activate p53 in polyploid cells. Liver tumors arise primarily from cells with low ploidy, indicating an intrinsic pro-tumorigenic effect of PIDDosome-mediated ploidy restriction. These data suggest that hyperpolyploidization caused by PIDDosome deficiency protects from HCC. Moreover, high tumor cell density, as a surrogate marker of low ploidy, predicts poor survival of HCC patients receiving liver transplantation. Together, we show that the PIDDosome is a potential therapeutic target to manipulate hepatocyte polyploidization for HCC prevention and that tumor cell density may serve as a novel prognostic marker for recurrence-free survival in HCC patients., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
42. Excessive E2F Transcription in Single Cancer Cells Precludes Transient Cell-Cycle Exit after DNA Damage.
- Author
-
Segeren HA, van Rijnberk LM, Moreno E, Riemers FM, van Liere EA, Yuan R, Wubbolts R, de Bruin A, and Westendorp B
- Subjects
- Cell Cycle, Humans, DNA Damage genetics, E2F Transcription Factors metabolism, Sequence Analysis, RNA methods
- Abstract
E2F transcription factors control the expression of cell-cycle genes. Cancers often demonstrate enhanced E2F target gene expression, which can be explained by increased percentages of replicating cells. However, we demonstrate in human cancer biopsy specimens that individual neoplastic cells display abnormally high levels of E2F-dependent transcription. To mimic this situation, we delete the atypical E2F repressors (E2F7/8) or overexpress the E2F3 activator in untransformed cells. Cells with elevated E2F activity during S/G2 phase fail to exit the cell cycle after DNA damage and undergo mitosis. In contrast, wild-type cells complete S phase and then exit the cell cycle by activating the APC/C
Cdh1 via repression of the E2F target Emi1. Many arrested wild-type cells eventually inactivate APC/CCdh1 to execute a second round of DNA replication and mitosis, thereby becoming tetraploid. Cells with elevated E2F transcription fail to exit the cell cycle after DNA damage, which potentially causes genomic instability, promotes malignant progression, and reduces drug sensitivity., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
43. E2F-Family Members Engage the PIDDosome to Limit Hepatocyte Ploidy in Liver Development and Regeneration.
- Author
-
Sladky VC, Knapp K, Soratroi C, Heppke J, Eichin F, Rocamora-Reverte L, Szabo TG, Bongiovanni L, Westendorp B, Moreno E, van Liere EA, Bakker B, Spierings DCJ, Wardenaar R, Pereyra D, Starlinger P, Schultze S, Trauner M, Stojakovic T, Scharnagl H, Fava LL, Foijer F, de Bruin A, and Villunger A
- Subjects
- Aneuploidy, Animals, CRADD Signaling Adaptor Protein physiology, Centrosome, Cyclin-Dependent Kinase Inhibitor p21 physiology, Cytokinesis, Female, Hepatocytes metabolism, Humans, Male, Mice, Mice, Knockout, Caspase 2 physiology, Death Domain Receptor Signaling Adaptor Proteins physiology, E2F Transcription Factors physiology, Hepatocytes cytology, Liver Regeneration, Polyploidy, Tumor Suppressor Protein p53 physiology
- Abstract
E2F transcription factors control the cytokinesis machinery and thereby ploidy in hepatocytes. If or how these proteins limit proliferation of polyploid cells with extra centrosomes remains unknown. Here, we show that the PIDDosome, a signaling platform essential for caspase-2-activation, limits hepatocyte ploidy and is instructed by the E2F network to control p53 in the developing as well as regenerating liver. Casp2 and Pidd1 act as direct transcriptional targets of E2F1 and its antagonists, E2F7 and E2F8, that together co-regulate PIDDosome expression during juvenile liver growth and regeneration. Of note, whereas hepatocyte aneuploidy correlates with the basal ploidy state, the degree of aneuploidy itself is not limited by PIDDosome-dependent p53 activation. Finally, we provide evidence that the same signaling network is engaged to control ploidy in the human liver after resection. Our study defines the PIDDosome as a primary target to manipulate hepatocyte ploidy and proliferation rates in the regenerating liver., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
44. Cyclin F-dependent degradation of E2F7 is critical for DNA repair and G2-phase progression.
- Author
-
Yuan R, Liu Q, Segeren HA, Yuniati L, Guardavaccaro D, Lebbink RJ, Westendorp B, and de Bruin A
- Subjects
- Cell Cycle Checkpoints, Cyclins genetics, DNA Damage, DNA Replication, E2F7 Transcription Factor genetics, HeLa Cells, Humans, Protein Binding, Repressor Proteins genetics, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Ubiquitination, Cyclins metabolism, DNA Repair, E2F7 Transcription Factor metabolism, G2 Phase physiology, Proteolysis, Repressor Proteins metabolism
- Abstract
E2F7 and E2F8 act as tumor suppressors via transcriptional repression of genes involved in S-phase entry and progression. Previously, we demonstrated that these atypical E2Fs are degraded by APC/C
C dh1 during G1 phase of the cell cycle. However, the mechanism driving the downregulation of atypical E2Fs during G2 phase is unknown. Here, we show that E2F7 is targeted for degradation by the E3 ubiquitin ligase SCFcyclin F during G2. Cyclin F binds via its cyclin domain to a conserved C-terminal CY motif on E2F7. An E2F7 mutant unable to interact with SCFcyclin F remains stable during G2. Furthermore, SCFcyclin F can also interact and induce degradation of E2F8. However, this does not require the cyclin domain of SCFcyclin F nor the CY motifs in the C-terminus of E2F8, implying a different regulatory mechanism than for E2F7. Importantly, depletion of cyclin F causes an atypical-E2F-dependent delay of the G2/M transition, accompanied by reduced expression of E2F target genes involved in DNA repair. Live cell imaging of DNA damage revealed that cyclin F-dependent regulation of atypical E2Fs is critical for efficient DNA repair and cell cycle progression., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2019
- Full Text
- View/download PDF
45. Atypical E2Fs inhibit tumor angiogenesis.
- Author
-
Weijts BGMW, Westendorp B, Hien BT, Martínez-López LM, Zijp M, Thurlings I, Thomas RE, Schulte-Merker S, Bakker WJ, and de Bruin A
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Calcium-Binding Proteins, Carcinogens toxicity, Cell Line, Tumor, E2F7 Transcription Factor genetics, Fibroblasts, Gene Expression Regulation, Neoplastic, Humans, Keratinocytes, Mice, Mice, Knockout, Mice, Nude, Neoplasms blood supply, Neoplasms chemically induced, Neoplasms genetics, Neoplasms, Experimental blood supply, Neoplasms, Experimental chemically induced, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Neovascularization, Pathologic genetics, Primary Cell Culture, Repressor Proteins genetics, Xenograft Model Antitumor Assays, Zebrafish, E2F7 Transcription Factor metabolism, Intracellular Signaling Peptides and Proteins metabolism, Membrane Proteins metabolism, Neoplasms pathology, Neovascularization, Pathologic pathology, Repressor Proteins metabolism
- Abstract
Atypical E2F transcription factors (E2F7 and E2F8) function as key regulators of cell cycle progression and their inactivation leads to spontaneous cancer formation in mice. However, the mechanism of the tumor suppressor functions of E2F7/8 remain obscure. In this study we discovered that atypical E2Fs control tumor angiogenesis, one of the hallmarks of cancer. We genetically inactivated atypical E2Fs in epithelial and mesenchymal neoplasm and analyzed blood vessel formation in three different animal models of cancer. Tumor formation was either induced by application of 7,12-Dimethylbenz(a)anthracene/12-O-Tetradecanoylphorbol-13-acetate or by Myc/Ras overexpression. To our surprise, atypical E2Fs suppressed tumor angiogenesis in all three cancer models, which is in a sharp contrast to previous findings showing that atypical E2Fs promote angiogenesis during fetal development in mice and zebrafish. Real-time imaging in zebrafish displayed that fluorescent-labeled blood vessels showed enhanced intratumoral branching in xenografted E2f7/8-deficient neoplasms compared with E2f7/8-proficient neoplasms. DLL4 expression, a key negative inhibitor of vascular branching, was decreased in E2f7/8-deficient neoplastic cells, indicating that E2F7/8 might inhibit intratumoral vessel branching via induction of DLL4.
- Published
- 2018
- Full Text
- View/download PDF
46. Indian Hedgehog Suppresses a Stromal Cell-Driven Intestinal Immune Response.
- Author
-
Westendorp BF, Büller NVJA, Karpus ON, van Dop WA, Koster J, Versteeg R, Koelink PJ, Snel CY, Meisner S, Roelofs JJTH, Uhmann A, Ver Loren van Themaat E, Heijmans J, Hahn H, Muncan V, Wildenberg ME, and van den Brink GR
- Abstract
Background & Aims: Upon intestinal epithelial damage a complex wound healing response is initiated to restore epithelial integrity and defend against pathogenic invasion. Epithelium-derived Indian Hedgehog (Ihh) functions as a critical sensor in this process. Signaling occurs in a paracrine manner because the receptor for Ihh is expressed only in the mesenchyme, but the exact Hedgehog target cell has remained elusive. The aim of this study was to elucidate further the nature of this target cell in the context of intestinal inflammation., Methods: Hedgehog activity was modulated genetically in both cell type-specific and body-wide models and the resulting animals were analyzed for gene expression profiles and sensitivity for dextran sodium sulfate (DSS) colitis. To characterize the Hedgehog target cell, Gli1-CreERT2-Rosa26-ZsGreen animals were generated, which express ZsGreen in all Hedgehog-responsive cells. These cells were characterized using flow cytometry and immunofluorescence., Results: Loss of Indian Hedgehog from the intestinal epithelium resulted in a rapid increase in expression of inflammation-related genes, accompanied by increased influx of immune cells. Animals with epithelium-specific deletion of Ihh or lacking the Hedgehog receptor Smoothened from Hedgehog target cells were more sensitive to DSS colitis. In contrast, specific deletion of Smoothened in the myeloid compartment did not alter the response to DSS. This suggests that Hedgehog signaling does not repress intestinal immunity through an effect on myeloid cells. Indeed, we found that Hedgehog-responsive cells expressed gp38, smooth muscle actin, and desmin, indicating a fibroblastic nature. Ihh signaling inhibited expression of C-X-C motif chemokine ligand 12 (CXCL12) in fibroblasts in vitro and in vivo, thereby impairing the recruitment of immune cells., Conclusions: We show that epithelium-derived Indian Hedgehog signals exclusively to fibroblasts in the intestine. Loss of Ihh leads to a rapid immune response with up-regulation of fibroblast-derived CXCL12, and migration of immune cells into the lamina propria.
- Published
- 2017
- Full Text
- View/download PDF
47. Dosage-dependent copy number gains in E2f1 and E2f3 drive hepatocellular carcinoma.
- Author
-
Kent LN, Bae S, Tsai SY, Tang X, Srivastava A, Koivisto C, Martin CK, Ridolfi E, Miller GC, Zorko SM, Plevris E, Hadjiyannis Y, Perez M, Nolan E, Kladney R, Westendorp B, de Bruin A, Fernandez S, Rosol TJ, Pohar KS, Pipas JM, and Leone G
- Subjects
- Animals, Humans, Mice, Mice, Knockout, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular metabolism, E2F1 Transcription Factor genetics, E2F1 Transcription Factor metabolism, E2F3 Transcription Factor genetics, E2F3 Transcription Factor metabolism, Gene Dosage, Genes, Neoplasm, Liver Neoplasms genetics, Liver Neoplasms metabolism, Neoplasm Proteins genetics, Neoplasm Proteins metabolism
- Abstract
Disruption of the retinoblastoma (RB) tumor suppressor pathway, either through genetic mutation of upstream regulatory components or mutation of RB1 itself, is believed to be a required event in cancer. However, genetic alterations in the RB-regulated E2F family of transcription factors are infrequent, casting doubt on a direct role for E2Fs in driving cancer. In this work, a mutation analysis of human cancer revealed subtle but impactful copy number gains in E2F1 and E2F3 in hepatocellular carcinoma (HCC). Using a series of loss- and gain-of-function alleles to dial E2F transcriptional output, we have shown that copy number gains in E2f1 or E2f3b resulted in dosage-dependent spontaneous HCC in mice without the involvement of additional organs. Conversely, germ-line loss of E2f1 or E2f3b, but not E2f3a, protected mice against HCC. Combinatorial mapping of chromatin occupancy and transcriptome profiling identified an E2F1- and E2F3B-driven transcriptional program that was associated with development and progression of HCC. These findings demonstrate a direct and cell-autonomous role for E2F activators in human cancer.
- Published
- 2017
- Full Text
- View/download PDF
48. E2f8 mediates tumor suppression in postnatal liver development.
- Author
-
Kent LN, Rakijas JB, Pandit SK, Westendorp B, Chen HZ, Huntington JT, Tang X, Bae S, Srivastava A, Senapati S, Koivisto C, Martin CK, Cuitino MC, Perez M, Clouse JM, Chokshi V, Shinde N, Kladney R, Sun D, Perez-Castro A, Matondo RB, Nantasanti S, Mokry M, Huang K, Machiraju R, Fernandez S, Rosol TJ, Coppola V, Pohar KS, Pipas JM, Schmidt CR, de Bruin A, and Leone G
- Subjects
- Alleles, Animals, Biopsy, Cell Proliferation, Cell Survival, DNA analysis, E2F7 Transcription Factor genetics, Female, Gene Deletion, Genotype, Hepatocytes cytology, Humans, Liver physiology, Male, Mice, Oligonucleotide Array Sequence Analysis, Protein Binding, Protein Domains, Repressor Proteins genetics, Sequence Analysis, RNA, Signal Transduction, Carcinoma, Hepatocellular metabolism, E2F7 Transcription Factor metabolism, Liver growth & development, Liver Neoplasms metabolism, Repressor Proteins metabolism
- Abstract
E2F-mediated transcriptional repression of cell cycle-dependent gene expression is critical for the control of cellular proliferation, survival, and development. E2F signaling also interacts with transcriptional programs that are downstream of genetic predictors for cancer development, including hepatocellular carcinoma (HCC). Here, we evaluated the function of the atypical repressor genes E2f7 and E2f8 in adult liver physiology. Using several loss-of-function alleles in mice, we determined that combined deletion of E2f7 and E2f8 in hepatocytes leads to HCC. Temporal-specific ablation strategies revealed that E2f8's tumor suppressor role is critical during the first 2 weeks of life, which correspond to a highly proliferative stage of postnatal liver development. Disruption of E2F8's DNA binding activity phenocopied the effects of an E2f8 null allele and led to HCC. Finally, a profile of chromatin occupancy and gene expression in young and tumor-bearing mice identified a set of shared targets for E2F7 and E2F8 whose increased expression during early postnatal liver development is associated with HCC progression in mice. Increased expression of E2F8-specific target genes was also observed in human liver biopsies from HCC patients compared to healthy patients. In summary, these studies suggest that E2F8-mediated transcriptional repression is a critical tumor suppressor mechanism during postnatal liver development.
- Published
- 2016
- Full Text
- View/download PDF
49. Feedback regulation between atypical E2Fs and APC/CCdh1 coordinates cell cycle progression.
- Author
-
Boekhout M, Yuan R, Wondergem AP, Segeren HA, van Liere EA, Awol N, Jansen I, Wolthuis RM, de Bruin A, and Westendorp B
- Subjects
- Animals, Cdh1 Proteins genetics, Cdh1 Proteins metabolism, Cells, Cultured, Cyclins metabolism, E2F Transcription Factors genetics, HEK293 Cells, HeLa Cells, Humans, Mice, Mice, Inbred C57BL, Protein Binding, Anaphase-Promoting Complex-Cyclosome metabolism, E2F Transcription Factors metabolism, Feedback, Physiological, S Phase
- Abstract
E2F transcription factors control the oscillating expression pattern of multiple target genes during the cell cycle. Activator E2Fs, E2F1-3, induce an upswing of E2F targets, which is essential for the G1-to-S phase transition, whereas atypical E2Fs, E2F7 and E2F8, mediate a downswing of the same targets during late S, G2, and M phases. Expression of atypical E2Fs is induced by E2F1-3, but it is unknown how atypical E2Fs are inactivated in a timely manner. Here, we demonstrate that E2F7 and E2F8 are substrates of the anaphase-promoting complex/cyclosome (APC/C). Removal of CDH1, or mutating the CDH1-interacting KEN boxes, stabilized E2F7/8 from anaphase onwards and during G1. Expressing KEN mutant E2F7 during G1 impairs S phase entry and eventually results in cell death. Furthermore, we show that E2F8, but not E2F7, interacts also with APC/C(C) (dc20). Importantly, atypical E2Fs can activate APC/C(C) (dh1) by repressing its inhibitors cyclin A, cyclin E, and Emi1. In conclusion, we discovered a feedback loop between atypical E2Fs and APC/C(C) (dh1), which ensures balanced expression of cell cycle genes and normal cell cycle progression., (© 2016 The Authors.)
- Published
- 2016
- Full Text
- View/download PDF
50. E2F7 and E2F8 promote angiogenesis through transcriptional activation of VEGFA in cooperation with HIF1.
- Author
-
Weijts BG, Bakker WJ, Cornelissen PW, Liang KH, Schaftenaar FH, Westendorp B, de Wolf CA, Paciejewska M, Scheele CL, Kent L, Leone G, Schulte-Merker S, and de Bruin A
- Subjects
- Animals, Animals, Genetically Modified, Cell Line, Tumor, E2F Transcription Factors genetics, Embryonic Development genetics, Embryonic Development physiology, Gene Deletion, Humans, Mice, Promoter Regions, Genetic, Zebrafish, E2F Transcription Factors metabolism, Hypoxia-Inducible Factor 1 metabolism, Neovascularization, Physiologic genetics, Transcriptional Activation, Vascular Endothelial Growth Factor A genetics
- Abstract
The E2F family of transcription factors plays an important role in controlling cell-cycle progression. While this is their best-known function, we report here novel functions for the newest members of the E2F family, E2F7 and E2F8 (E2F7/8). We show that simultaneous deletion of E2F7/8 in zebrafish and mice leads to severe vascular defects during embryonic development. Using a panel of transgenic zebrafish with fluorescent-labelled blood vessels, we demonstrate that E2F7/8 are essential for proper formation of blood vessels. Despite their classification as transcriptional repressors, we provide evidence for a molecular mechanism through which E2F7/8 activate the transcription of the vascular endothelial growth factor A (VEGFA), a key factor in guiding angiogenesis. We show that E2F7/8 directly bind and stimulate the VEGFA promoter independent of canonical E2F binding elements. Instead, E2F7/8 form a transcriptional complex with the hypoxia inducible factor 1 (HIF1) to stimulate VEGFA promoter activity. These results uncover an unexpected link between E2F7/8 and the HIF1-VEGFA pathway providing a molecular mechanism by which E2F7/8 control angiogenesis.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.