255 results on '"Medema RH"'
Search Results
2. 3D genomics across the tree of life reveals condensin II as a determinant of architecture type
- Author
-
Hoencamp, C, Dudchenko, O, Elbatsh, AMO, Brahmachari, S, Raaijmakers, JA, van Schaik, T, Cacciatore, AS, Contessoto, VG, van Heesbeen, RGHP, van den Broek, B, Mhaskar, AN, Teunissen, H, St Hilaire, BG, Weisz, D, Omer, AD, Pham, M, Colaric, Z, Yang, Z, Rao, SSP, Mitra, N, Lui, C, Yao, W, Khan, R, Moroz, LL, Kohn, A, St Leger, J, Mena, A, Holcroft, K, Gambetta, MC, Lim, F, Farley, E, Stein, N, Haddad, A, Chauss, D, Mutlu, AS, Wang, MC, Young, ND, Hildebrandt, E, Cheng, HH, Knight, CJ, Burnham, TLU, Hovel, KA, Beel, AJ, Mattei, P-J, Kornberg, RD, Warren, WC, Cary, G, Gomez-Skarmeta, JL, Hinman, V, Lindblad-Toh, K, Di Palma, F, Maeshima, K, Multani, AS, Sen, P, Nel-Themaat, L, Behringer, RR, Kaur, P, Medema, RH, van Steensel, B, de Wit, E, Onuchic, JN, Di Pierro, M, Aiden, EL, Rowland, BD, Hoencamp, C, Dudchenko, O, Elbatsh, AMO, Brahmachari, S, Raaijmakers, JA, van Schaik, T, Cacciatore, AS, Contessoto, VG, van Heesbeen, RGHP, van den Broek, B, Mhaskar, AN, Teunissen, H, St Hilaire, BG, Weisz, D, Omer, AD, Pham, M, Colaric, Z, Yang, Z, Rao, SSP, Mitra, N, Lui, C, Yao, W, Khan, R, Moroz, LL, Kohn, A, St Leger, J, Mena, A, Holcroft, K, Gambetta, MC, Lim, F, Farley, E, Stein, N, Haddad, A, Chauss, D, Mutlu, AS, Wang, MC, Young, ND, Hildebrandt, E, Cheng, HH, Knight, CJ, Burnham, TLU, Hovel, KA, Beel, AJ, Mattei, P-J, Kornberg, RD, Warren, WC, Cary, G, Gomez-Skarmeta, JL, Hinman, V, Lindblad-Toh, K, Di Palma, F, Maeshima, K, Multani, AS, Sen, P, Nel-Themaat, L, Behringer, RR, Kaur, P, Medema, RH, van Steensel, B, de Wit, E, Onuchic, JN, Di Pierro, M, Aiden, EL, and Rowland, BD
- Abstract
We investigated genome folding across the eukaryotic tree of life. We find two types of three-dimensional (3D) genome architectures at the chromosome scale. Each type appears and disappears repeatedly during eukaryotic evolution. The type of genome architecture that an organism exhibits correlates with the absence of condensin II subunits. Moreover, condensin II depletion converts the architecture of the human genome to a state resembling that seen in organisms such as fungi or mosquitoes. In this state, centromeres cluster together at nucleoli, and heterochromatin domains merge. We propose a physical model in which lengthwise compaction of chromosomes by condensin II during mitosis determines chromosome-scale genome architecture, with effects that are retained during the subsequent interphase. This mechanism likely has been conserved since the last common ancestor of all eukaryotes.
- Published
- 2021
3. Activation of nuclear factor-kappa B signalling promotes cellular senescence
- Author
-
Rovillain, E, Mansfield, L, Caetano, C, Alvarez-Fernandez, M, Caballero, OL, Medema, RH, Hummerich, H, and Jat, PS
- Published
- 2011
- Full Text
- View/download PDF
4. Consequences of chromosome segregation errors
- Author
-
CMM Sectie Molecular Cancer Research, Cancer, Medema, RH, Raaijmakers, J.A., Soto Ruiz De La Torre, Mar, CMM Sectie Molecular Cancer Research, Cancer, Medema, RH, Raaijmakers, J.A., and Soto Ruiz De La Torre, Mar
- Published
- 2019
5. Product of the surrounding: Location dependent effects on DNA double strand break responses
- Author
-
Medema, RH, van den Berg, Jeroen, Medema, RH, and van den Berg, Jeroen
- Published
- 2019
6. Cohesin Releases DNA through Asymmetric ATPase-Driven Ring Opening
- Author
-
Elbatsh, AMO, Haarhuis, JHI, Petela, N, Chapard, C, Fish, A, Celie, PH, Stadnik, M, Ristic, D., Wyman, C.L., Medema, RH, Nasmyth, K, Rowland, BD, Molecular Genetics, and Radiotherapy
- Subjects
Adenosine Triphosphatases ,Saccharomyces cerevisiae Proteins ,Chromosomal Proteins, Non-Histone ,Cell Cycle ,Nuclear Proteins ,Acetylation ,Cell Cycle Proteins ,DNA ,Saccharomyces cerevisiae ,Cell Biology ,Article ,Chromatin ,Catalytic Domain ,Humans ,biological phenomena, cell phenomena, and immunity ,Molecular Biology - Abstract
Summary Cohesin stably holds together the sister chromatids from S phase until mitosis. To do so, cohesin must be protected against its cellular antagonist Wapl. Eco1 acetylates cohesin’s Smc3 subunit, which locks together the sister DNAs. We used yeast genetics to dissect how Wapl drives cohesin from chromatin and identified mutants of cohesin that are impaired in ATPase activity but remarkably confer robust cohesion that bypasses the need for the cohesin protectors Eco1 in yeast and Sororin in human cells. We uncover a functional asymmetry within the heart of cohesin’s highly conserved ABC-like ATPase machinery and find that both ATPase sites contribute to DNA loading, whereas DNA release is controlled specifically by one site. We propose that Smc3 acetylation locks cohesin rings around the sister chromatids by counteracting an activity associated with one of cohesin’s two ATPase sites., Graphical Abstract, Highlights • Cohesin’s DNA release involves an asymmetric activity within its ATPase machinery • DNA release is driven by the ATPase site proximal to the locking acetylation marks • Cohesin’s DNA release is ATPase driven in yeast and humans • Cohesin’s loading onto DNA is controlled by both of its ATPase sites, Tight regulation of DNA entrapment and release by the cohesin complex is crucial for its multiple cellular functions. Elbatsh et al. find that cohesin’s release from DNA requires an activity associated with one of its ATPase sites, whereas both sites control cohesin’s loading onto DNA.
- Published
- 2016
- Full Text
- View/download PDF
7. Cell fate decisions after DNA damage: Withdraw the risky cases
- Author
-
Medema, RH, Feringa, Femke M., Medema, RH, and Feringa, Femke M.
- Published
- 2018
8. Keeping it togehter: DNA entrapment and release by cohesin
- Author
-
Cancer, CMM USEQ Facility, Medema, RH, Rowland, B.D., Haarhuis, J.H.I., Cancer, CMM USEQ Facility, Medema, RH, Rowland, B.D., and Haarhuis, J.H.I.
- Published
- 2016
9. Mitotic spindle perturbations
- Author
-
CMM Sectie Molecular Cancer Research, Medema, RH, Tame, Mihoko Amy, CMM Sectie Molecular Cancer Research, Medema, RH, and Tame, Mihoko Amy
- Published
- 2016
10. FoxM1 promotes beta-Catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis
- Author
-
Zhang, N, Wei, P, Gong, A, Chiu, WT, Lee, HT, Colman, H, Huang, H, Xue, J, Liu, M, Wang, Y, Sawaya, R, Xie, K, Yung, WKA, Medema, RH, He, X, Huang, S, Zhang, N, Wei, P, Gong, A, Chiu, WT, Lee, HT, Colman, H, Huang, H, Xue, J, Liu, M, Wang, Y, Sawaya, R, Xie, K, Yung, WKA, Medema, RH, He, X, and Huang, S
- Published
- 2013
11. Mutations in LRRC50 Predispose Zebrafish and Humans to Seminomas
- Author
-
Basten, SG, Davis, EE, Gillis, Ad, van Rooijen, E, Stoop, Hans, Babala, N, Logister, I, Heath, ZG, Jonges, TN, Katsanis, N, Voest, EE, van Eeden, FJ, Medema, RH, Ketting, RF, Schulte-Merker, S, Looijenga, LHJ (Leendert), Giles, RH, Basten, SG, Davis, EE, Gillis, Ad, van Rooijen, E, Stoop, Hans, Babala, N, Logister, I, Heath, ZG, Jonges, TN, Katsanis, N, Voest, EE, van Eeden, FJ, Medema, RH, Ketting, RF, Schulte-Merker, S, Looijenga, LHJ (Leendert), and Giles, RH
- Published
- 2013
12. Inhibition of polo-like kinase-1 by DNA damage occurs in an ATM- or ATR-dependent fashion
- Author
-
van Vugt, MATM, Smits, VAJ, Klompmaker, R, Medema, RH, Damage and Repair in Cancer Development and Cancer Treatment (DARE), and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
CAFFEINE ,CELL-CYCLE REGULATION ,CDC25 ,CHK1 ,RADIOSENSITIZING AGENT ,ATAXIA-TELANGIECTASIA ,IN-VITRO ,biological phenomena, cell phenomena, and immunity ,PROTEIN-KINASE ,PHOSPHORYLATION ,CHECKPOINT KINASE - Abstract
Polo-like kinases play multiple roles in different phases of mitosis. We have recently shown that the mammalian polo-like kinase, Plk1, is inhibited in response to DNA damage and that this inhibition may lead to cell cycle arrests at multiple points in mitosis. Here we have investigated the role of the checkpoint kinases ATM ((a) under bar taxia (t) under bar elangiectasia (m) under bar utated) and ATR (ATM- and Rad3-related) in DNA damage-induced inhibition of Plk1. We show that inhibition of Plk1 kinase activity is efficiently blocked by the radio-sensitizing agent caffeine. Using ATM(-/-) cells we show that under certain circumstances, inhibition of Plk1 by DNA-damaging agents critically depends on ATM. In addition, we show that LN radiation also causes inhibition of Plk1, and we present evidence that this inhibition is mediated by ATR. Taken together, our data demonstrate that ATM and ATR can regulate Plk1 kinase activity in response to a variety of DNA-damaging agents.
- Published
- 2001
13. Bicaudal D2, Dynein, and Kinesin-1 Associate with Nuclear Pore Complexes and Regulate Centrosome and Nuclear Positioning during Mitotic Entry
- Author
-
Splinter, EC, Tanenbaum, ME, Lindqvist, A, Jaarsma, Dick, Flotho, A, Yu, Carol, Grigoriev, I, Engelsma, D, Haasdijk, Elize, Keijzer, Nanda, Demmers, Jeroen, Fornerod, M, Melchior, F, Hoogenraad, CC, Medema, RH, Akhmanova, Anna, Splinter, EC, Tanenbaum, ME, Lindqvist, A, Jaarsma, Dick, Flotho, A, Yu, Carol, Grigoriev, I, Engelsma, D, Haasdijk, Elize, Keijzer, Nanda, Demmers, Jeroen, Fornerod, M, Melchior, F, Hoogenraad, CC, Medema, RH, and Akhmanova, Anna
- Abstract
BICD2 is one of the two mammalian homologues of the Drosophila Bicaudal D, an evolutionarily conserved adaptor between microtubule motors and their cargo that was previously shown to link vesicles and mRNP complexes to the dynein motor. Here, we identified a G2-specific role for BICD2 in the relative positioning of the nucleus and centrosomes in dividing cells. By combining mass spectrometry, biochemical and cell biological approaches, we show that the nuclear pore complex (NPC) component RanBP2 directly binds to BICD2 and recruits it to NPCs specifically in G2 phase of the cell cycle. BICD2, in turn, recruits dynein-dynactin to NPCs and as such is needed to keep centrosomes closely tethered to the nucleus prior to mitotic entry. When dynein function is suppressed by RNA interference-mediated depletion or antibody microinjection, centrosomes and nuclei are actively pushed apart in late G2 and we show that this is due to the action of kinesin-1. Surprisingly, depletion of BICD2 inhibits both dynein and kinesin-1-dependent movements of the nucleus and cytoplasmic NPCs, demonstrating that BICD2 is needed not only for the dynein function at the nuclear pores but also for the antagonistic activity of kinesin-1. Our study demonstrates that the nucleus is subject to opposing activities of dynein and kinesin-1 motors and that BICD2 contributes to nuclear and centrosomal positioning prior to mitotic entry through regulation of both dynein and kinesin-1.
- Published
- 2010
14. FOXM1 (forkhead box M1)
- Author
-
Laoukili, J, primary, Alvarez, Fernandez M, additional, and Medema, RH, additional
- Published
- 2011
- Full Text
- View/download PDF
15. Knockdown of Polo-like kinase-1 prevents centrosome separation and leads to a spindle checkpoint arrest
- Author
-
van Vugt, MATM, Medema, RH, Damage and Repair in Cancer Development and Cancer Treatment (DARE), and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
16. Reduction of chromosomal instability and inflammation is a common aspect of adaptation to aneuploidy.
- Author
-
Hintzen DC, Schubert M, Soto M, Medema RH, and Raaijmakers JA
- Subjects
- Humans, Adaptation, Physiological genetics, Cell Line, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Signal Transduction, Aneuploidy, Chromosomal Instability, Inflammation genetics, Inflammation pathology, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Cell Proliferation genetics
- Abstract
Aneuploidy, while detrimental to untransformed cells, is notably prevalent in cancer. Aneuploidy is found as an early event during tumorigenesis which indicates that cancer cells have the ability to surmount the initial stress responses associated with aneuploidy, enabling rapid proliferation despite aberrant karyotypes. To generate more insight into key cellular processes and requirements underlying adaptation to aneuploidy, we generated a panel of aneuploid clones in p53-deficient RPE-1 cells and studied their behavior over time. As expected, de novo-generated aneuploid clones initially display reduced fitness, enhanced levels of chromosomal instability (CIN), and an upregulated inflammatory response. Intriguingly, after prolonged culturing, aneuploid clones exhibit increased proliferation rates while maintaining aberrant karyotypes, indicative of an adaptive response to the aneuploid state. Interestingly, all adapted clones display reduced CIN and reduced inflammatory signaling, suggesting that these are common aspects of adaptation to aneuploidy. Collectively, our data suggests that CIN and concomitant inflammation are key processes that require correction to allow for fast proliferation in vitro. Finally, we provide evidence that amplification of oncogenic KRAS can promote adaptation., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. A novel lineage-tracing tool reveals that hypoxic tumor cells drive tumor relapse after radiotherapy.
- Author
-
Menegakis A, Vennin C, Ient J, Groot AJ, Krenning L, Klompmaker R, Friskes A, Ilic M, Yaromina A, Harkes R, van den Broek B, Jakob Sonke J, De Jong M, Piepers J, van Rheenen J, Vooijs MA, and Medema RH
- Abstract
Purpose: Tumor hypoxia imposes a main obstacle to the efficacy of anti-cancer therapy. Understanding the cellular dynamics of individual hypoxic cells before, during and post-treatment has been hampered by the technical inability to identify and trace these cells over time., Methods and Materials: Here, we present a novel lineage-tracing reporter for hypoxic cells based on the conditional expression of a HIF1a-CreER
T2 -UnaG biosensor that can visualize hypoxic cells in a time-dependent manner and trace the fate of hypoxic cells over time. We combine this system with multiphoton microscopy, flow cytometry, and immunofluorescence to characterize the role of hypoxic cells in tumor relapse after irradiation in H1299 tumor spheroids and in vivo xenografts., Results: We validate the reporter in monolayer cultures and we show that tagged cells colocalize in spheroids and human tumor xenografts with the hypoxic marker pimonidazole. We found that irradiation of H1299-HIFcreUnaG spheroids leads to preferential outgrowth of cells from the hypoxic core. Similarly, in xenografts tumors, although initially UnaG-positive-cells coincide with pimonidazole-positive tumor areas and they are merely quiescent, upon Irradiation UnaG-positive cells enrich in regrowing tumors and are mainly proliferative., Conclusions: Collectively, our data provide clear evidence that the hypoxic cells drive tumor relapse after irradiation., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier B.V.)- Published
- 2024
- Full Text
- View/download PDF
18. Chromatin context-dependent effects of epigenetic drugs on CRISPR-Cas9 editing.
- Author
-
Schep R, Trauernicht M, Vergara X, Friskes A, Morris B, Gregoricchio S, Manzo SG, Zwart W, Beijersbergen RL, Medema RH, and van Steensel B
- Subjects
- Humans, DNA Repair, CRISPR-Associated Protein 9 metabolism, CRISPR-Associated Protein 9 genetics, Heterochromatin metabolism, Heterochromatin genetics, Cell Line, Histones metabolism, Euchromatin genetics, DNA Breaks, Double-Stranded drug effects, CRISPR-Cas Systems, Gene Editing methods, Epigenesis, Genetic drug effects, Chromatin metabolism, Chromatin genetics, Histone Deacetylase Inhibitors pharmacology
- Abstract
The efficiency and outcome of CRISPR/Cas9 editing depends on the chromatin state at the cut site. It has been shown that changing the chromatin state can influence both the efficiency and repair outcome, and epigenetic drugs have been used to improve Cas9 editing. However, because the target proteins of these drugs are not homogeneously distributed across the genome, the efficacy of these drugs may be expected to vary from locus to locus. Here, we systematically analyzed this chromatin context-dependency for 160 epigenetic drugs. We used a human cell line with 19 stably integrated reporters to induce a double-stranded break in different chromatin environments. We then measured Cas9 editing efficiency and repair pathway usage by sequencing the mutational signatures. We identified 58 drugs that modulate Cas9 editing efficiency and/or repair outcome dependent on the local chromatin environment. For example, we find a subset of histone deacetylase inhibitors that improve Cas9 editing efficiency throughout all types of heterochromatin (e.g. PCI-24781), while others were only effective in euchromatin and H3K27me3-marked regions (e.g. apicidin). In summary, this study reveals that most epigenetic drugs alter CRISPR editing in a chromatin-dependent manner, and provides a resource to improve Cas9 editing more selectively at the desired location., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
19. Paradoxical Activation of Oncogenic Signaling as a Cancer Treatment Strategy.
- Author
-
Dias MH, Friskes A, Wang S, Fernandes Neto JM, van Gemert F, Mourragui S, Papagianni C, Kuiken HJ, Mainardi S, Alvarez-Villanueva D, Lieftink C, Morris B, Dekker A, van Dijk E, Wilms LHS, da Silva MS, Jansen RA, Mulero-Sánchez A, Malzer E, Vidal A, Santos C, Salazar R, Wailemann RAM, Torres TEP, De Conti G, Raaijmakers JA, Snaebjornsson P, Yuan S, Qin W, Kovach JS, Armelin HA, Te Riele H, van Oudenaarden A, Jin H, Beijersbergen RL, Villanueva A, Medema RH, and Bernards R
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Xenograft Model Antitumor Assays, Cell Cycle Proteins metabolism, Cell Cycle Proteins antagonists & inhibitors, Protein-Tyrosine Kinases antagonists & inhibitors, Protein-Tyrosine Kinases metabolism, Drug Resistance, Neoplasm, Nuclear Proteins metabolism, Nuclear Proteins genetics, DNA Replication, Signal Transduction, Protein Phosphatase 2 metabolism, Colonic Neoplasms drug therapy, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Colonic Neoplasms genetics
- Abstract
Cancer homeostasis depends on a balance between activated oncogenic pathways driving tumorigenesis and engagement of stress response programs that counteract the inherent toxicity of such aberrant signaling. Although inhibition of oncogenic signaling pathways has been explored extensively, there is increasing evidence that overactivation of the same pathways can also disrupt cancer homeostasis and cause lethality. We show here that inhibition of protein phosphatase 2A (PP2A) hyperactivates multiple oncogenic pathways and engages stress responses in colon cancer cells. Genetic and compound screens identify combined inhibition of PP2A and WEE1 as synergistic in multiple cancer models by collapsing DNA replication and triggering premature mitosis followed by cell death. This combination also suppressed the growth of patient-derived tumors in vivo. Remarkably, acquired resistance to this drug combination suppressed the ability of colon cancer cells to form tumors in vivo. Our data suggest that paradoxical activation of oncogenic signaling can result in tumor-suppressive resistance. Significance: A therapy consisting of deliberate hyperactivation of oncogenic signaling combined with perturbation of the stress responses that result from this is very effective in animal models of colon cancer. Resistance to this therapy is associated with loss of oncogenic signaling and reduced oncogenic capacity, indicative of tumor-suppressive drug resistance., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
20. Widespread chromatin context-dependencies of DNA double-strand break repair proteins.
- Author
-
Vergara X, Manjón AG, de Haas M, Morris B, Schep R, Leemans C, Friskes A, Beijersbergen RL, Sanders MA, Medema RH, and van Steensel B
- Subjects
- Humans, BRCA2 Protein genetics, BRCA2 Protein metabolism, Ataxia Telangiectasia Mutated Proteins metabolism, Ataxia Telangiectasia Mutated Proteins genetics, DNA Repair, DNA Breaks, Double-Stranded, DNA End-Joining Repair, Chromatin metabolism, Chromatin genetics, Heterochromatin metabolism, Heterochromatin genetics, Euchromatin metabolism, Euchromatin genetics
- Abstract
DNA double-strand breaks are repaired by multiple pathways, including non-homologous end-joining (NHEJ) and microhomology-mediated end-joining (MMEJ). The balance of these pathways is dependent on the local chromatin context, but the underlying mechanisms are poorly understood. By combining knockout screening with a dual MMEJ:NHEJ reporter inserted in 19 different chromatin environments, we identified dozens of DNA repair proteins that modulate pathway balance dependent on the local chromatin state. Proteins that favor NHEJ mostly synergize with euchromatin, while proteins that favor MMEJ generally synergize with distinct types of heterochromatin. Examples of the former are BRCA2 and POLL, and of the latter the FANC complex and ATM. Moreover, in a diversity of human cancer types, loss of several of these proteins alters the distribution of pathway-specific mutations between heterochromatin and euchromatin. Together, these results uncover a complex network of proteins that regulate MMEJ:NHEJ balance in a chromatin context-dependent manner., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
21. EU-LIFE charter of independent life science research institutes.
- Author
-
Superti-Furga G, Agostinho M, Bury J, Cook S, Durinx C, Ender A, van Luenen H, Lund AH, Medema RH, Miączyńska M, Nickel D, Pelicci PG, Puisieux A, Ripatti S, Sander M, Schubeler D, Serrano L, Sommer T, Sonne-Hansen K, Tomančák P, Vives J, Vontas J, and Bettencourt-Dias M
- Subjects
- Academies and Institutes, Biological Science Disciplines, Biomedical Research
- Abstract
The diverse range of organizations contributing to the global research ecosystem is believed to enhance the overall quality and resilience of its output. Mid-sized autonomous research institutes, distinct from universities, play a crucial role in this landscape. They often lead the way in new research fields and experimental methods, including those in social and organizational domains, which are vital for driving innovation. The EU-LIFE alliance was established with the goal of fostering excellence by developing and disseminating best practices among European biomedical research institutes. As directors of the 15 EU-LIFE institutes, we have spent a decade comparing and refining our processes. Now, we are eager to share the insights we've gained. To this end, we have crafted this Charter, outlining 10 principles we deem essential for research institutes to flourish and achieve ground-breaking discoveries. These principles, detailed in the Charter, encompass excellence, independence, training, internationality and inclusivity, mission focus, technological advancement, administrative innovation, cooperation, societal impact, and public engagement. Our aim is to inspire the establishment of new institutes that adhere to these principles and to raise awareness about their significance. We are convinced that they should be viewed a crucial component of any national and international innovation strategies., (© 2024 Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
22. UNCAN.eu: Toward a European Federated Cancer Research Data Hub.
- Author
-
Boutros M, Baumann M, Bigas A, Chaabane L, Guérin J, Habermann JK, Jobard A, Pelicci PG, Stegle O, Tonon G, Valencia A, Winkler EC, Blanc P, De Maria R, Medema RH, Nagy P, Tabernero J, and Solary E
- Subjects
- Humans, Research, European Union, Neoplasms
- Abstract
To enable a collective effort that generates a new level of UNderstanding CANcer (UNCAN.eu) [Cancer Discov (2022) 12 (11): OF1], the European Union supports the creation of a sustainable platform that connects cancer research across Member States. A workshop hosted in Heidelberg gathered European cancer experts to identify ongoing initiatives that may contribute to building this platform and discuss the governance and long-term evolution of a European Federated Cancer Data Hub., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
23. Perturbations in 3D genome organization can promote acquired drug resistance.
- Author
-
Manjón AG, Manzo SG, Prekovic S, Potgeter L, van Schaik T, Liu NQ, Flach K, Peric-Hupkes D, Joosten S, Teunissen H, Friskes A, Ilic M, Hintzen D, Franceschini-Santos VH, Zwart W, de Wit E, van Steensel B, and Medema RH
- Subjects
- Humans, Paclitaxel pharmacology, Drug Resistance, Multiple genetics, DNA Methylation genetics, Cell Line, Tumor, Drug Resistance, Neoplasm genetics, Neoplasms genetics
- Abstract
Acquired drug resistance is a major problem in the treatment of cancer. hTERT-immortalized, untransformed RPE-1 cells can acquire resistance to Taxol by derepressing the ABCB1 gene, encoding for the multidrug transporter P-gP. Here, we investigate how the ABCB1 gene is derepressed. ABCB1 activation is associated with reduced H3K9 trimethylation, increased H3K27 acetylation, and ABCB1 displacement from the nuclear lamina. While altering DNA methylation and H3K27 methylation had no major impact on ABCB1 expression, nor did it promote resistance, disrupting the nuclear lamina component Lamin B Receptor did promote the acquisition of a Taxol-resistant phenotype in a subset of cells. CRISPRa-mediated gene activation supported the notion that lamina dissociation influences ABCB1 derepression. We propose a model in which nuclear lamina dissociation of a repressed gene allows for its activation, implying that deregulation of the 3D genome topology could play an important role in tumor evolution and the acquisition of drug resistance., Competing Interests: Declaration of interests E.d.W. is a co-founder of Cergentis B.V., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. MND1 enables homologous recombination in somatic cells primarily outside the context of replication.
- Author
-
Koob L, Friskes A, van Bergen L, Feringa FM, van den Broek B, Koeleman ES, van Beek E, Schubert M, Blomen VA, Brummelkamp TR, Krenning L, and Medema RH
- Subjects
- Humans, DNA Breaks, Double-Stranded, Recombinational DNA Repair, S Phase, Cell Cycle Proteins metabolism, DNA Repair genetics, Homologous Recombination genetics
- Abstract
Faithful and timely repair of DNA double-strand breaks (DSBs) is fundamental for the maintenance of genomic integrity. Here, we demonstrate that the meiotic recombination co-factor MND1 facilitates the repair of DSBs in somatic cells. We show that MND1 localizes to DSBs, where it stimulates DNA repair through homologous recombination (HR). Importantly, MND1 is not involved in the response to replication-associated DSBs, implying that it is dispensable for HR-mediated repair of one-ended DSBs. Instead, we find that MND1 specifically plays a role in the response to two-ended DSBs that are induced by irradiation (IR) or various chemotherapeutic drugs. Surprisingly, we find that MND1 is specifically active in G2 phase, whereas it only marginally affects repair during S phase. MND1 localization to DSBs is dependent on resection of the DNA ends and seemingly occurs through direct binding of MND1 to RAD51-coated ssDNA. Importantly, the lack of MND1-driven HR repair directly potentiates the toxicity of IR-induced damage, which could open new possibilities for therapeutic intervention, specifically in HR-proficient tumors., (© 2023 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2023
- Full Text
- View/download PDF
25. UNCAN.eu, a European Initiative to UNderstand CANcer.
- Author
-
Solary E, Blanc P, Boutros M, Girvalaki C, Locatelli F, Medema RH, Nagy P, and Tabernero J
- Subjects
- Humans, Europe, Neoplasms
- Abstract
"UNCAN.eu" refers to a collective European effort seeking to enable a leap forward in our understanding of cancer. This initiative, which includes the creation of a European cancer research data hub, will pave the way to new advances in cancer care. Starting on September 1, 2022, a 15-month coordination and support action will generate a blueprint for UNCAN.eu. Here, we summarize the cancer research issues that the blueprint will propose to tackle at the European level., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
26. From fluorescent foci to sequence: Illuminating DNA double strand break repair by high-throughput sequencing technologies.
- Author
-
Vergara X, Schep R, Medema RH, and van Steensel B
- Subjects
- DNA metabolism, High-Throughput Nucleotide Sequencing, Technology, DNA Breaks, Double-Stranded, DNA Repair
- Abstract
Technologies to study DNA double-strand break (DSB) repair have traditionally mostly relied on fluorescence read-outs, either by microscopy or flow cytometry. The advent of high throughput sequencing (HTS) has created fundamentally new opportunities to study the mechanisms underlying DSB repair. Here, we review the suite of HTS-based assays that are used to study three different aspects of DNA repair: detection of broken ends, protein recruitment and pathway usage. We highlight new opportunities that HTS technology offers towards a better understanding of the DSB repair process., Competing Interests: Conflict of interest None., (Copyright © 2022 Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
27. Double-strand break toxicity is chromatin context independent.
- Author
-
Friskes A, Koob L, Krenning L, Severson TM, Koeleman ES, Vergara X, Schubert M, van den Berg J, Evers B, Manjón AG, Joosten S, Kim Y, Zwart W, and Medema RH
- Subjects
- CRISPR-Cas Systems, Chromatin genetics, DNA Repair, Humans, Lamins, RNA, DNA Breaks, Double-Stranded
- Abstract
Cells respond to double-strand breaks (DSBs) by activating DNA damage response pathways, including cell cycle arrest. We have previously shown that a single double-strand break generated via CRISPR/Cas9 is sufficient to delay cell cycle progression and compromise cell viability. However, we also found that the cellular response to DSBs can vary, independent of the number of lesions. This implies that not all DSBs are equally toxic, and raises the question if the location of a single double-strand break could influence its toxicity. To systematically investigate if DSB-location is a determinant of toxicity we performed a CRISPR/Cas9 screen targeting 6237 single sites in the human genome. Next, we developed a data-driven framework to design CRISPR/Cas9 sgRNA (crRNA) pools targeting specific chromatin features. The chromatin context was defined using ChromHMM states, Lamin-B1 DAM-iD, DNAseI hypersensitivity, and RNA-sequencing data. We computationally designed 6 distinct crRNA pools, each containing 10 crRNAs targeting the same chromatin state. We show that the toxicity of a DSB is highly similar across the different ChromHMM states. Rather, we find that the major determinants of toxicity of a sgRNA are cutting efficiency and off-target effects. Thus, chromatin features have little to no effect on the toxicity of a single CRISPR/Cas9-induced DSB., (© The Author(s) 2022. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
28. Indisulam synergizes with palbociclib to induce senescence through inhibition of CDK2 kinase activity.
- Author
-
Pogacar Z, Johnson JL, Krenning L, De Conti G, Jochems F, Lieftink C, Velds A, Wardak L, Groot K, Schepers A, Wang L, Song JY, van de Ven M, van Tellingen O, Medema RH, Beijersbergen RL, Bernards R, and Leite de Oliveira R
- Subjects
- Cell Line, Tumor, Cyclin-Dependent Kinase 2, Cyclin-Dependent Kinase 4 metabolism, Humans, Piperazines, Pyridines, Sulfonamides, Cyclin-Dependent Kinase 6 metabolism, Protein Kinase Inhibitors pharmacology
- Abstract
Inducing senescence in cancer cells is emerging as a new therapeutic strategy. In order to find ways to enhance senescence induction by palbociclib, a CDK4/6 inhibitor approved for treatment of metastatic breast cancer, we performed functional genetic screens in palbociclib-resistant cells. Using this approach, we found that loss of CDK2 results in strong senescence induction in palbociclib-treated cells. Treatment with the CDK2 inhibitor indisulam, which phenocopies genetic CDK2 inactivation, led to sustained senescence induction when combined with palbociclib in various cell lines and lung cancer xenografts. Treating cells with indisulam led to downregulation of cyclin H, which prevented CDK2 activation. Combined treatment with palbociclib and indisulam induced a senescence program and sensitized cells to senolytic therapy. Our data indicate that inhibition of CDK2 through indisulam treatment can enhance senescence induction by CDK4/6 inhibition., Competing Interests: R.B is the founder of the company Oncosence (https://www.oncosence.com), which aims to develop senescence-inducing and senolytic compounds to treat cancer. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2022
- Full Text
- View/download PDF
29. Drug-Induced Epigenomic Plasticity Reprograms Circadian Rhythm Regulation to Drive Prostate Cancer toward Androgen Independence.
- Author
-
Linder S, Hoogstraat M, Stelloo S, Eickhoff N, Schuurman K, de Barros H, Alkemade M, Bekers EM, Severson TM, Sanders J, Huang CF, Morova T, Altintas UB, Hoekman L, Kim Y, Baca SC, Sjöström M, Zaalberg A, Hintzen DC, de Jong J, Kluin RJC, de Rink I, Giambartolomei C, Seo JH, Pasaniuc B, Altelaar M, Medema RH, Feng FY, Zoubeidi A, Freedman ML, Wessels LFA, Butler LM, Lack NA, van der Poel H, Bergman AM, and Zwart W
- Subjects
- ARNTL Transcription Factors genetics, Cell Line, Tumor, Circadian Rhythm, Drug Resistance, Neoplasm genetics, Epigenomics, Humans, Male, Nitriles therapeutic use, Receptors, Androgen genetics, Androgens pharmacology, Androgens therapeutic use, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology
- Abstract
In prostate cancer, androgen receptor (AR)-targeting agents are very effective in various disease stages. However, therapy resistance inevitably occurs, and little is known about how tumor cells adapt to bypass AR suppression. Here, we performed integrative multiomics analyses on tissues isolated before and after 3 months of AR-targeting enzalutamide monotherapy from patients with high-risk prostate cancer enrolled in a neoadjuvant clinical trial. Transcriptomic analyses demonstrated that AR inhibition drove tumors toward a neuroendocrine-like disease state. Additionally, epigenomic profiling revealed massive enzalutamide-induced reprogramming of pioneer factor FOXA1 from inactive chromatin sites toward active cis-regulatory elements that dictate prosurvival signals. Notably, treatment-induced FOXA1 sites were enriched for the circadian clock component ARNTL. Posttreatment ARNTL levels were associated with patients' clinical outcomes, and ARNTL knockout strongly decreased prostate cancer cell growth. Our data highlight a remarkable cistromic plasticity of FOXA1 following AR-targeted therapy and revealed an acquired dependency on the circadian regulator ARNTL, a novel candidate therapeutic target., Significance: Understanding how prostate cancers adapt to AR-targeted interventions is critical for identifying novel drug targets to improve the clinical management of treatment-resistant disease. Our study revealed an enzalutamide-induced epigenomic plasticity toward prosurvival signaling and uncovered the circadian regulator ARNTL as an acquired vulnerability after AR inhibition, presenting a novel lead for therapeutic development. See related commentary by Zhang et al., p. 2017. This article is highlighted in the In This Issue feature, p. 2007., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
30. Life of double minutes: generation, maintenance, and elimination.
- Author
-
Ilić M, Zaalberg IC, Raaijmakers JA, and Medema RH
- Subjects
- Chromosome Aberrations, DNA, Humans, Oncogenes, Gene Amplification, Neoplasms genetics
- Abstract
Advances in genome sequencing have revealed a type of extrachromosomal DNA, historically named double minutes (also referred to as ecDNA), to be common in a wide range of cancer types, but not in healthy tissues. These cancer-associated circular DNA molecules contain one or a few genes that are amplified when double minutes accumulate. Double minutes harbor oncogenes or drug resistance genes that contribute to tumor aggressiveness through copy number amplification in combination with favorable epigenetic properties. Unequal distribution of double minutes over daughter cells contributes to intratumoral heterogeneity, thereby increasing tumor adaptability. In this review, we discuss various models delineating the mechanism of generation of double minutes. Furthermore, we highlight how double minutes are maintained, how they evolve, and discuss possible mechanisms driving their elimination., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
31. The impact of monosomies, trisomies and segmental aneuploidies on chromosomal stability.
- Author
-
Hintzen DC, Soto M, Schubert M, Bakker B, Spierings DCJ, Szuhai K, Lansdorp PM, Kluin RJC, Foijer F, Medema RH, and Raaijmakers JA
- Subjects
- Chromosomal Instability, Genetic Testing, Humans, Monosomy, Aneuploidy, Trisomy genetics
- Abstract
Aneuploidy and chromosomal instability are both commonly found in cancer. Chromosomal instability leads to karyotype heterogeneity in tumors and is associated with therapy resistance, metastasis and poor prognosis. It has been hypothesized that aneuploidy per se is sufficient to drive CIN, however due to limited models and heterogenous results, it has remained controversial which aspects of aneuploidy can drive CIN. In this study we systematically tested the impact of different types of aneuploidies on the induction of CIN. We generated a plethora of isogenic aneuploid clones harboring whole chromosome or segmental aneuploidies in human p53-deficient RPE-1 cells. We observed increased segregation errors in cells harboring trisomies that strongly correlated to the number of gained genes. Strikingly, we found that clones harboring only monosomies do not induce a CIN phenotype. Finally, we found that an initial chromosome breakage event and subsequent fusion can instigate breakage-fusion-bridge cycles. By investigating the impact of monosomies, trisomies and segmental aneuploidies on chromosomal instability we further deciphered the complex relationship between aneuploidy and CIN., Competing Interests: No, there is no conflict of interest. My manuscript contains the following statement: "The authors declare that they have no conflict of interest."
- Published
- 2022
- Full Text
- View/download PDF
32. Unexpected gene activation following CRISPR-Cas9-mediated genome editing.
- Author
-
Manjón AG, Linder S, Teunissen H, Friskes A, Zwart W, de Wit E, and Medema RH
- Subjects
- Transcriptional Activation, CRISPR-Cas Systems, Gene Editing
- Abstract
The discovery of the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and its development as a genome editing tool has revolutionized the field of molecular biology. In the DNA damage field, CRISPR has brought an alternative to induce endogenous double-strand breaks (DSBs) at desired genomic locations and study the DNA damage response and its consequences. Many systems for sgRNA delivery have been reported in order to efficiently generate this DSB, including lentiviral vectors. However, some of the consequences of these systems are not yet well understood. Here, we report that lentiviral-based sgRNA vectors can integrate into the endogenous genomic target location, leading to undesired activation of the target gene. By generating a DSB in the regulatory region of the ABCB1 gene using a lentiviral sgRNA vector, we can induce the formation of Taxol-resistant colonies. We show that these colonies upregulate ABCB1 via integration of the EEF1A1 and the U6 promoters from the sgRNA vector. We believe that this is an unreported CRISPR/Cas9 on-target effect that researchers need to be aware of when using lentiviral vectors for genome editing., (© 2021 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
33. A novel assay for screening WIP1 phosphatase substrates in nuclear extracts.
- Author
-
Storchova R, Burdova K, Palek M, Medema RH, and Macurek L
- Subjects
- Acetylation, Adaptor Proteins, Signal Transducing genetics, Bone Neoplasms genetics, Bone Neoplasms metabolism, Cell Nucleus genetics, DNA Damage, DNA Repair, Humans, Osteosarcoma genetics, Osteosarcoma metabolism, Phosphorylation, Protein Interaction Domains and Motifs, Protein Phosphatase 2C genetics, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Adaptor Proteins, Signal Transducing metabolism, Biological Assay methods, Bone Neoplasms pathology, Cell Nucleus metabolism, Osteosarcoma pathology, Protein Phosphatase 2C metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Upon exposure to genotoxic stress, cells activate DNA damage response (DDR) that coordinates DNA repair with a temporal arrest in the cell cycle progression. DDR is triggered by activation of ataxia telangiectasia mutated/ataxia telangiectasia and Rad3-related protein kinases that phosphorylate multiple targets including tumor suppressor protein tumor suppressor p53 (p53). In addition, DNA damage can activate parallel stress response pathways [such as mitogen-activated protein kinase p38 alpha (p38)/MAPK-activated protein kinase 2 (MK2) kinases] contributing to establishing the cell cycle arrest. Wild-type p53-induced phosphatase 1 (WIP1) controls timely inactivation of DDR and is needed for recovery from the G2 checkpoint by counteracting the function of p53. Here, we developed a simple in vitro assay for testing WIP1 substrates in nuclear extracts. Whereas we did not detect any activity of WIP1 toward p38/MK2, we confirmed p53 as a substrate of WIP1. Inhibition or inactivation of WIP1 in U2OS cells increased phosphorylation of p53 at S15 and potentiated its acetylation at K382. Further, we identified Deleted in breast cancer gene 1 (DBC1) as a new substrate of WIP1 but surprisingly, depletion of DBC1 did not interfere with the ability of WIP1 to regulate p53 acetylation. Instead, we have found that WIP1 activity suppresses p53-K382 acetylation by inhibiting the interaction between p53 and the acetyltransferase p300. Newly established phosphatase assay allows an easy comparison of WIP1 ability to dephosphorylate various proteins and thus contributes to identification of its physiological substrates., (© 2021 Federation of European Biochemical Societies.)
- Published
- 2021
- Full Text
- View/download PDF
34. CIP2A Interacts with TopBP1 and Drives Basal-Like Breast Cancer Tumorigenesis.
- Author
-
Laine A, Nagelli SG, Farrington C, Butt U, Cvrljevic AN, Vainonen JP, Feringa FM, Grönroos TJ, Gautam P, Khan S, Sihto H, Qiao X, Pavic K, Connolly DC, Kronqvist P, Elo LL, Maurer J, Wennerberg K, Medema RH, Joensuu H, Peuhu E, de Visser K, Narla G, and Westermarck J
- Subjects
- 9,10-Dimethyl-1,2-benzanthracene, Animals, Cell Cycle, Cell Line, Tumor, Cell Proliferation, DNA Damage, Female, Humans, Immunohistochemistry, Mice, Mice, Knockout, Mice, Transgenic, Mitosis, Mutation, Proteome, Recombination, Genetic, Signal Transduction, Autoantigens metabolism, Breast Neoplasms metabolism, Carcinogenesis, Carrier Proteins metabolism, DNA-Binding Proteins metabolism, Intracellular Signaling Peptides and Proteins metabolism, Membrane Proteins metabolism, Nuclear Proteins metabolism
- Abstract
Basal-like breast cancers (BLBC) are characterized by defects in homologous recombination (HR), deficient mitotic checkpoint, and high-proliferation activity. Here, we discover CIP2A as a candidate driver of BLBC. CIP2A was essential for DNA damage-induced initiation of mouse BLBC-like mammary tumors and for survival of HR-defective BLBC cells. CIP2A was dispensable for normal mammary gland development and for unperturbed mitosis, but selectively essential for mitotic progression of DNA damaged cells. A direct interaction between CIP2A and a DNA repair scaffold protein TopBP1 was identified, and CIP2A inhibition resulted in enhanced DNA damage-induced TopBP1 and RAD51 recruitment to chromatin in mammary epithelial cells. In addition to its role in tumor initiation, and survival of BRCA-deficient cells, CIP2A also drove proliferative MYC and E2F1 signaling in basal-like triple-negative breast cancer (BL-TNBC) cells. Clinically, high CIP2A expression was associated with poor patient prognosis in BL-TNBCs but not in other breast cancer subtypes. Small-molecule reactivators of PP2A (SMAP) inhibited CIP2A transcription, phenocopied the CIP2A-deficient DNA damage response (DDR), and inhibited growth of patient-derived BLBC xenograft. In summary, these results demonstrate that CIP2A directly interacts with TopBP1 and coordinates DNA damage-induced mitotic checkpoint and proliferation, thereby driving BLBC initiation and progression. SMAPs could serve as a surrogate therapeutic strategy to inhibit the oncogenic activity of CIP2A in BLBCs. SIGNIFICANCE: These results identify CIP2A as a nongenetic driver and therapeutic target in basal-like breast cancer that regulates DNA damage-induced G
2 -M checkpoint and proliferative signaling., (©2021 The Authors; Published by the American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
35. 3D genomics across the tree of life reveals condensin II as a determinant of architecture type.
- Author
-
Hoencamp C, Dudchenko O, Elbatsh AMO, Brahmachari S, Raaijmakers JA, van Schaik T, Sedeño Cacciatore Á, Contessoto VG, van Heesbeen RGHP, van den Broek B, Mhaskar AN, Teunissen H, St Hilaire BG, Weisz D, Omer AD, Pham M, Colaric Z, Yang Z, Rao SSP, Mitra N, Lui C, Yao W, Khan R, Moroz LL, Kohn A, St Leger J, Mena A, Holcroft K, Gambetta MC, Lim F, Farley E, Stein N, Haddad A, Chauss D, Mutlu AS, Wang MC, Young ND, Hildebrandt E, Cheng HH, Knight CJ, Burnham TLU, Hovel KA, Beel AJ, Mattei PJ, Kornberg RD, Warren WC, Cary G, Gómez-Skarmeta JL, Hinman V, Lindblad-Toh K, Di Palma F, Maeshima K, Multani AS, Pathak S, Nel-Themaat L, Behringer RR, Kaur P, Medema RH, van Steensel B, de Wit E, Onuchic JN, Di Pierro M, Lieberman Aiden E, and Rowland BD
- Subjects
- Adenosine Triphosphatases chemistry, Algorithms, Animals, Cell Nucleolus ultrastructure, Cell Nucleus ultrastructure, Centromere ultrastructure, Chromosomes chemistry, Chromosomes, Human chemistry, Chromosomes, Human ultrastructure, DNA-Binding Proteins chemistry, Genome, Human, Genomics, Heterochromatin ultrastructure, Humans, Interphase, Mitosis, Models, Biological, Multiprotein Complexes chemistry, Telomere ultrastructure, Adenosine Triphosphatases genetics, Adenosine Triphosphatases physiology, Biological Evolution, Chromosomes ultrastructure, DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Eukaryota genetics, Genome, Multiprotein Complexes genetics, Multiprotein Complexes physiology
- Abstract
We investigated genome folding across the eukaryotic tree of life. We find two types of three-dimensional (3D) genome architectures at the chromosome scale. Each type appears and disappears repeatedly during eukaryotic evolution. The type of genome architecture that an organism exhibits correlates with the absence of condensin II subunits. Moreover, condensin II depletion converts the architecture of the human genome to a state resembling that seen in organisms such as fungi or mosquitoes. In this state, centromeres cluster together at nucleoli, and heterochromatin domains merge. We propose a physical model in which lengthwise compaction of chromosomes by condensin II during mitosis determines chromosome-scale genome architecture, with effects that are retained during the subsequent interphase. This mechanism likely has been conserved since the last common ancestor of all eukaryotes., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
36. Impact of chromatin context on Cas9-induced DNA double-strand break repair pathway balance.
- Author
-
Schep R, Brinkman EK, Leemans C, Vergara X, van der Weide RH, Morris B, van Schaik T, Manzo SG, Peric-Hupkes D, van den Berg J, Beijersbergen RL, Medema RH, and van Steensel B
- Subjects
- Base Sequence, DNA End-Joining Repair, Euchromatin metabolism, Gene Rearrangement, Genome, Human, Heterochromatin metabolism, Humans, INDEL Mutation genetics, K562 Cells, Kinetics, Protein Binding, Reproducibility of Results, CRISPR-Associated Protein 9 metabolism, Chromatin metabolism, DNA Breaks, Double-Stranded, DNA Repair
- Abstract
DNA double-strand break (DSB) repair is mediated by multiple pathways. It is thought that the local chromatin context affects the pathway choice, but the underlying principles are poorly understood. Using a multiplexed reporter assay in combination with Cas9 cutting, we systematically measure the relative activities of three DSB repair pathways as a function of chromatin context in >1,000 genomic locations. This reveals that non-homologous end-joining (NHEJ) is broadly biased toward euchromatin, while the contribution of microhomology-mediated end-joining (MMEJ) is higher in specific heterochromatin contexts. In H3K27me3-marked heterochromatin, inhibition of the H3K27 methyltransferase EZH2 reverts the balance toward NHEJ. Single-stranded template repair (SSTR), often used for precise CRISPR editing, competes with MMEJ and is moderately linked to chromatin context. These results provide insight into the impact of chromatin on DSB repair pathway balance and guidance for the design of Cas9-mediated genome editing experiments., Competing Interests: Declaration of interests B.v.S. is a member of the Advisory Board of Molecular Cell., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
37. Resistance of Hypoxic Cells to Ionizing Radiation Is Mediated in Part via Hypoxia-Induced Quiescence.
- Author
-
Menegakis A, Klompmaker R, Vennin C, Arbusà A, Damen M, van den Broek B, Zips D, van Rheenen J, Krenning L, and Medema RH
- Subjects
- Cell Line, Tumor, Humans, Cell Hypoxia physiology, Radiation, Ionizing
- Abstract
Double strand breaks (DSBs) are highly toxic to a cell, a property that is exploited in radiation therapy. A critical component for the damage induction is cellular oxygen, making hypoxic tumor areas refractory to the efficacy of radiation treatment. During a fractionated radiation regimen, these hypoxic areas can be re-oxygenated. Nonetheless, hypoxia still constitutes a negative prognostic factor for the patient's outcome. We hypothesized that this might be attributed to specific hypoxia-induced cellular traits that are maintained upon reoxygenation. Here, we show that reoxygenation of hypoxic non-transformed RPE-1 cells fully restored induction of DSBs but the cells remain radioresistant as a consequence of hypoxia-induced quiescence. With the use of the cell cycle indicators (FUCCI), cell cycle-specific radiation sensitivity, the cell cycle phase duration with live cell imaging, and single cell tracing were assessed. We observed that RPE-1 cells experience a longer G1 phase under hypoxia and retain a large fraction of cells that are non-cycling. Expression of HPV oncoprotein E7 prevents hypoxia-induced quiescence and abolishes the radioprotective effect. In line with this, HPV-negative cancer cell lines retain radioresistance, while HPV-positive cancer cell lines are radiosensitized upon reoxygenation. Quiescence induction in hypoxia and its HPV-driven prevention was observed in 3D multicellular spheroids. Collectively, we identify a new hypoxia-dependent radioprotective phenotype due to hypoxia-induced quiescence that accounts for a global decrease in radiosensitivity that can be retained upon reoxygenation and is absent in cells expressing oncoprotein E7.
- Published
- 2021
- Full Text
- View/download PDF
38. Combined Inactivation of Pocket Proteins and APC/C Cdh1 by Cdk4/6 Controls Recovery from DNA Damage in G1 Phase.
- Author
-
Shaltiel IA, Llopis A, Aprelia M, Klompmaker R, Menegakis A, Krenning L, and Medema RH
- Subjects
- Humans, Transfection, Antigens, CD metabolism, Cadherins metabolism, Cell Cycle Proteins metabolism, Cyclin-Dependent Kinase 4 metabolism, Cyclin-Dependent Kinase 6 metabolism, DNA Damage genetics, G1 Phase physiology
- Abstract
Most Cyclin-dependent kinases (Cdks) are redundant for normal cell division. Here we tested whether these redundancies are maintained during cell cycle recovery after a DNA damage-induced arrest in G1. Using non-transformed RPE-1 cells, we find that while Cdk4 and Cdk6 act redundantly during normal S-phase entry, they both become essential for S-phase entry after DNA damage in G1. We show that this is due to a greater overall dependency for Cdk4/6 activity, rather than to independent functions of either kinase. In addition, we show that inactivation of pocket proteins is sufficient to overcome the inhibitory effects of complete Cdk4/6 inhibition in otherwise unperturbed cells, but that this cannot revert the effects of Cdk4/6 inhibition in DNA damaged cultures. Indeed, we could confirm that, in addition to inactivation of pocket proteins, Cdh1-dependent anaphase-promoting complex/cyclosome (APC/C
Cdh1 ) activity needs to be inhibited to promote S-phase entry in damaged cultures. Collectively, our data indicate that DNA damage in G1 creates a unique situation where high levels of Cdk4/6 activity are required to inactivate pocket proteins and APC/CCdh1 to promote the transition from G1 to S phase.- Published
- 2021
- Full Text
- View/download PDF
39. Sustained CHK2 activity, but not ATM activity, is critical to maintain a G1 arrest after DNA damage in untransformed cells.
- Author
-
García-Santisteban I, Llopis A, Krenning L, Vallejo-Rodríguez J, van den Broek B, Zubiaga AM, and Medema RH
- Subjects
- Ataxia Telangiectasia Mutated Proteins metabolism, Cell Line, Checkpoint Kinase 2 metabolism, Humans, Ataxia Telangiectasia Mutated Proteins genetics, Checkpoint Kinase 2 genetics, DNA Damage, G1 Phase Cell Cycle Checkpoints genetics
- Abstract
Background: The G1 checkpoint is a critical regulator of genomic stability in untransformed cells, preventing cell cycle progression after DNA damage. DNA double-strand breaks (DSBs) recruit and activate ATM, a kinase which in turn activates the CHK2 kinase to establish G1 arrest. While the onset of G1 arrest is well understood, the specific role that ATM and CHK2 play in regulating G1 checkpoint maintenance remains poorly characterized., Results: Here we examine the impact of ATM and CHK2 activities on G1 checkpoint maintenance in untransformed cells after DNA damage caused by DSBs. We show that ATM becomes dispensable for G1 checkpoint maintenance as early as 1 h after DSB induction. In contrast, CHK2 kinase activity is necessary to maintain the G1 arrest, independently of ATM, ATR, and DNA-PKcs, implying that the G1 arrest is maintained in a lesion-independent manner. Sustained CHK2 activity is achieved through auto-activation and its acute inhibition enables cells to abrogate the G1-checkpoint and enter into S-phase. Accordingly, we show that CHK2 activity is lost in cells that recover from the G1 arrest, pointing to the involvement of a phosphatase with fast turnover., Conclusion: Our data indicate that G1 checkpoint maintenance relies on CHK2 and that its negative regulation is crucial for G1 checkpoint recovery after DSB induction.
- Published
- 2021
- Full Text
- View/download PDF
40. The Widely Used Antihelmintic Drug Albendazole is a Potent Inducer of Loss of Heterozygosity.
- Author
-
Will Castro LSEP, Pieters W, Alemdehy MF, Aslam MA, Buoninfante OA, Raaijmakers JA, Pilzecker B, van den Berk PCM, Te Riele H, Medema RH, Pedrosa RC, and Jacobs H
- Abstract
The antihelmintic drug ABZ and its metabolites belong to the chemical family of benzimidazoles (BZM) that act as potent tubulin polymerization inhibitors, suggesting a potential re-direction of BZMs for cancer therapy. Applying UV-Vis spectrometry we here demonstrate ABZ as a DNA intercalator. This insight led us to determine the primary mode of ABZ action in mammalian cells. As revealed by RNA sequencing, ABZ did neither grossly affect replication as analyzed by survival and replication stress signaling, nor the transcriptome. Actually, unbiased transcriptome analysis revealed a marked cell cycle signature in ABZ exposed cells. Indeed, short-term exposure to ABZ arrested mammalian cells in G2/M cell cycle stages associated with frequent gains and losses of chromatin. Cellular analyses revealed ABZ as a potent mammalian spindle poison for normal and malignant cells, explaining the serious chromosome segregation defects. Since chromosomal aberrations promote both cancer development and cell death, we determined if besides its general cytotoxicity, ABZ could predispose to tumor development. As measured by loss of heterozygosity (LOH) in vitro and in vivo ABZ was found as a potent inducer of LOH and accelerator of chromosomal missegregation., 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 © 2021 Will Castro, Pieters, Alemdehy, Aslam, Buoninfante, Raaijmakers, Pilzecker, van den Berk, te Riele, Medema, Pedrosa and Jacobs.)
- Published
- 2021
- Full Text
- View/download PDF
41. Centrosomes: Please keep your social distance!
- Author
-
Krenning L, Raaijmakers JA, and Medema RH
- Subjects
- Cell Cycle Checkpoints, Centrosome, Chromosome Segregation
- Abstract
Accurate control of centrosome number is essential for proper chromosome segregation, and it is well established that centrosome abnormalities can trigger a p53-dependent cell cycle arrest. Two new studies published in The EMBO Journal demonstrate how PIDD1 is recruited to centrosomes and that the localization of PIDD1 to distal appendages of centrosomes is required for PIDDosome activation at clustered supernumerary centrosomes., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
42. A FOXO-dependent replication checkpoint restricts proliferation of damaged cells.
- Author
-
Hornsveld M, Feringa FM, Krenning L, van den Berg J, Smits LMM, Nguyen NBT, Rodríguez-Colman MJ, Dansen TB, Medema RH, and Burgering BMT
- Subjects
- Cell Proliferation, Humans, Cell Cycle physiology, DNA Damage genetics, DNA Replication genetics, Genomic Instability genetics
- Abstract
DNA replication is challenged by numerous exogenous and endogenous factors that can interfere with the progression of replication forks. Substantial accumulation of single-stranded DNA during DNA replication activates the DNA replication stress checkpoint response that slows progression from S/G2 to M phase to protect genomic integrity. Whether and how mild replication stress restricts proliferation remains controversial. Here, we identify a cell cycle exit mechanism that prevents S/G2 phase arrested cells from undergoing mitosis after exposure to mild replication stress through premature activation of the anaphase promoting complex/cyclosome (APC/C
CDH1 ). We find that replication stress causes a gradual decrease of the levels of the APC/CCDH1 inhibitor EMI1/FBXO5 through Forkhead box O (FOXO)-mediated inhibition of its transcription factor E2F1. By doing so, FOXOs limit the time during which the replication stress checkpoint is reversible and thereby play an important role in maintaining genomic stability., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
43. Combining Supervised and Unsupervised Machine Learning Methods for Phenotypic Functional Genomics Screening.
- Author
-
Omta WA, van Heesbeen RG, Shen I, de Nobel J, Robers D, van der Velden LM, Medema RH, Siebes APJM, Feelders AJ, Brinkkemper S, Klumperman JS, Spruit MR, Brinkhuis MJS, and Egan DA
- Subjects
- Genome, Human genetics, Humans, Phenotype, RNA, Small Interfering genetics, Genomics, High-Throughput Screening Assays methods, Supervised Machine Learning, Unsupervised Machine Learning
- Abstract
There has been an increase in the use of machine learning and artificial intelligence (AI) for the analysis of image-based cellular screens. The accuracy of these analyses, however, is greatly dependent on the quality of the training sets used for building the machine learning models. We propose that unsupervised exploratory methods should first be applied to the data set to gain a better insight into the quality of the data. This improves the selection and labeling of data for creating training sets before the application of machine learning. We demonstrate this using a high-content genome-wide small interfering RNA screen. We perform an unsupervised exploratory data analysis to facilitate the identification of four robust phenotypes, which we subsequently use as a training set for building a high-quality random forest machine learning model to differentiate four phenotypes with an accuracy of 91.1% and a kappa of 0.85. Our approach enhanced our ability to extract new knowledge from the screen when compared with the use of unsupervised methods alone.
- Published
- 2020
- Full Text
- View/download PDF
44. Life in the (Pluripotent) Fast Lane: Cdk1 Preserves the Epigenome to Maintain Pluripotency.
- Author
-
Raaijmakers JA and Medema RH
- Subjects
- Animals, CDC2 Protein Kinase, Cell Differentiation, Epigenesis, Genetic, Mice, Embryonic Stem Cells, Epigenome
- Abstract
Michowski and colleagues (2020) engineered analog-sensitive Cdk1 knockin mice to identify Cdk1 targets in embryonic stem cells, which led them to discover a novel function for Cdk1 in shaping the epigenetic landscape by direct regulation of epigenetic modulators., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
45. Targeting the cyclin-dependent kinase 5 in metastatic melanoma.
- Author
-
Sharma S, Zhang T, Michowski W, Rebecca VW, Xiao M, Ferretti R, Suski JM, Bronson RT, Paulo JA, Frederick D, Fassl A, Boland GM, Geng Y, Lees JA, Medema RH, Herlyn M, Gygi SP, and Sicinski P
- Subjects
- Animals, Cell Line, Tumor, Cell Movement drug effects, Cell Movement genetics, Cyclin-Dependent Kinase 5 antagonists & inhibitors, Cyclin-Dependent Kinase 5 genetics, Female, Gene Dosage, Humans, Male, Melanoma drug therapy, Melanoma genetics, Melanoma mortality, Melanoma, Experimental drug therapy, Melanoma, Experimental genetics, Mice, Mice, Knockout, Phosphorylation drug effects, Phosphorylation genetics, Prognosis, Skin pathology, Skin Neoplasms drug therapy, Skin Neoplasms genetics, Skin Neoplasms mortality, Vimentin metabolism, Xenograft Model Antitumor Assays, Cyclin-Dependent Kinase 5 metabolism, Melanoma pathology, Melanoma, Experimental pathology, Skin Neoplasms pathology
- Abstract
The cyclin-dependent kinase 5 (CDK5), originally described as a neuronal-specific kinase, is also frequently activated in human cancers. Using conditional CDK5 knockout mice and a mouse model of highly metastatic melanoma, we found that CDK5 is dispensable for the growth of primary tumors. However, we observed that ablation of CDK5 completely abrogated the metastasis, revealing that CDK5 is essential for the metastatic spread. In mouse and human melanoma cells CDK5 promotes cell invasiveness by directly phosphorylating an intermediate filament protein, vimentin, thereby inhibiting assembly of vimentin filaments. Chemical inhibition of CDK5 blocks the metastatic spread of patient-derived melanomas in patient-derived xenograft (PDX) mouse models. Hence, inhibition of CDK5 might represent a very potent therapeutic strategy to impede the metastatic dissemination of malignant cells., Competing Interests: Competing interest statement: P.S. has been a consultant at Novartis, Genovis, Guidepoint, The Planning Shop, ORIC Pharmaceuticals, and Exo Therapeutics; his laboratory receives research funding from Novartis. W.M. is currently an employee of Cedilla Therapeutics.
- Published
- 2020
- Full Text
- View/download PDF
46. Degree and site of chromosomal instability define its oncogenic potential.
- Author
-
Hoevenaar WHM, Janssen A, Quirindongo AI, Ma H, Klaasen SJ, Teixeira A, van Gerwen B, Lansu N, Morsink FHM, Offerhaus GJA, Medema RH, Kops GJPL, and Jelluma N
- Subjects
- Adenoma genetics, Aneuploidy, Animals, Cell Proliferation, Chromosome Segregation, Colon pathology, Disease Models, Animal, Female, Gastrointestinal Neoplasms genetics, Intestinal Neoplasms genetics, Intestines pathology, Karyotype, Mice, Mice, Inbred C57BL, Mice, Transgenic, Organoids, Carcinogenesis genetics, Chromosomal Instability, Oncogenes genetics
- Abstract
Most human cancers are aneuploid, due to a chromosomal instability (CIN) phenotype. Despite being hallmarks of cancer, however, the roles of CIN and aneuploidy in tumor formation have not unequivocally emerged from animal studies and are thus still unclear. Using a conditional mouse model for diverse degrees of CIN, we find that a particular range is sufficient to drive very early onset spontaneous adenoma formation in the intestine. In mice predisposed to intestinal cancer (Apc
Min/+ ), moderate CIN causes a remarkable increase in adenoma burden in the entire intestinal tract and especially in the distal colon, which resembles human disease. Strikingly, a higher level of CIN promotes adenoma formation in the distal colon even more than moderate CIN does, but has no effect in the small intestine. Our results thus show that CIN can be potently oncogenic, but that certain levels of CIN can have contrasting effects in distinct tissues.- Published
- 2020
- Full Text
- View/download PDF
47. PHF6 promotes non-homologous end joining and G2 checkpoint recovery.
- Author
-
Warmerdam DO, Alonso-de Vega I, Wiegant WW, van den Broek B, Rother MB, Wolthuis RM, Freire R, van Attikum H, Medema RH, and Smits VA
- Subjects
- Cell Line, Tumor, DNA End-Joining Repair, G2 Phase Cell Cycle Checkpoints, Growth Disorders, Humans, Hypogonadism, Mental Retardation, X-Linked, Repressor Proteins genetics
- Abstract
The cellular response to DNA breaks is influenced by chromatin compaction. To identify chromatin regulators involved in the DNA damage response, we screened for genes that affect recovery following DNA damage using an RNAi library of chromatin regulators. We identified genes involved in chromatin remodeling, sister chromatid cohesion, and histone acetylation not previously associated with checkpoint recovery. Among these is the PHD finger protein 6 (PHF6), a gene mutated in Börjeson-Forssman-Lehmann syndrome and leukemic cancers. We find that loss of PHF6 dramatically compromises checkpoint recovery in G2 phase cells. Moreover, PHF6 is rapidly recruited to sites of DNA lesions in a PARP-dependent manner and required for efficient DNA repair through classical non-homologous end joining. These results indicate that PHF6 is a novel DNA damage response regulator that promotes end joining-mediated repair, thereby stimulating timely recovery from the G2 checkpoint., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
48. [Environmental regulations impede cancer research and treatment].
- Author
-
Kenter MJH, Clevers JC, Cornelissen J, and Medema RH
- Subjects
- Humans, Netherlands, Biomedical Research, Neoplasms therapy, Organisms, Genetically Modified, Risk Assessment methods
- Abstract
In order to legally conduct clinical research into new cancer treatments with medicinal products based on genetically modified organisms (GMOs) and treat cancer patients with these products, Dutch hospitals must obtain an environmental permit from the Minister of Infrastructure & Water Management. In the Netherlands, permit applications are assessed more strictly than in other EU member states, even if the products do not pose any real risk to the population and the environment. As a result, Dutch patients have to wait longer before the clinical trial or therapy with these GMO products can commence. This makes current Dutch environmental policy a serious obstacle to public health. Recently physicians and researchers have therefore pleaded with Dutch MPs for an adjustment of the lower national environmental regulations. Unfortunately, these pleas were not supported by said elected members of parliament, and the Ministry of Infrastructure & Water Management seems to be holding on to a pointless environmental permit for clinical research and treatment of cancer patients with these GMO medicinal products.
- Published
- 2019
49. Distinct Roles for Condensin's Two ATPase Sites in Chromosome Condensation.
- Author
-
Elbatsh AMO, Kim E, Eeftens JM, Raaijmakers JA, van der Weide RH, García-Nieto A, Bravo S, Ganji M, Uit de Bos J, Teunissen H, Medema RH, de Wit E, Haering CH, Dekker C, and Rowland BD
- Subjects
- Adenosine Triphosphatases genetics, Adenosine Triphosphatases physiology, Adenosine Triphosphate chemistry, Binding Sites genetics, Binding Sites physiology, Cell Cycle Proteins metabolism, Cell Line, Tumor, Chromatin physiology, Chromosomal Proteins, Non-Histone metabolism, Chromosomes metabolism, Chromosomes physiology, DNA metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Humans, Multiprotein Complexes physiology, Protein Binding physiology, Protein Subunits metabolism, Cohesins, Adenosine Triphosphatases metabolism, Chromatin Assembly and Disassembly physiology, DNA-Binding Proteins metabolism, Multiprotein Complexes metabolism
- Abstract
Condensin is a conserved SMC complex that uses its ATPase machinery to structure genomes, but how it does so is largely unknown. We show that condensin's ATPase has a dual role in chromosome condensation. Mutation of one ATPase site impairs condensation, while mutating the second site results in hyperactive condensin that compacts DNA faster than wild-type, both in vivo and in vitro. Whereas one site drives loop formation, the second site is involved in the formation of more stable higher-order Z loop structures. Using hyperactive condensin I, we reveal that condensin II is not intrinsically needed for the shortening of mitotic chromosomes. Condensin II rather is required for a straight chromosomal axis and enables faithful chromosome segregation by counteracting the formation of ultrafine DNA bridges. SMC complexes with distinct roles for each ATPase site likely reflect a universal principle that enables these molecular machines to intricately control chromosome architecture., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
50. Killing a zombie: a full deletion of the BUB1 gene in HAP1 cells.
- Author
-
Raaijmakers JA and Medema RH
- Subjects
- Sequence Deletion, M Phase Cell Cycle Checkpoints
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.