42 results on '"Lerdrup M"'
Search Results
2. CDK12/CDK13 inhibition disrupts a transcriptional program critical for glioblastoma survival
- Author
-
Lier, S., Rein, Kjell Arne, Lund, S., Lång, A., Lång, E., Meyer, N., Dutta, A., Anand, S., Nesse, G., Johansen, R., Klungland, A., Rinholm, J., Boe, S., Anand, A., Pollard, S., Lerdrup, M., Pandey, D., Lier, S., Rein, Kjell Arne, Lund, S., Lång, A., Lång, E., Meyer, N., Dutta, A., Anand, S., Nesse, G., Johansen, R., Klungland, A., Rinholm, J., Boe, S., Anand, A., Pollard, S., Lerdrup, M., and Pandey, D.
- Abstract
Background Glioblastoma multiforme (GBM) is the most prevalent and aggressive malignant tumor of the central nervous system. With a median survival of only one year, GBM patients have a particularly poor prognosis, highlighting a clear need for novel therapeutic strategies to target this disease. Transcriptional cyclin-dependent kinases (CDK), which phosphorylate key residues of RNA polymerase II (RNAPII) C-terminal domain (CTD), play a major role in sustaining aberrant transcriptional programs that are key to development and maintenance of cancer cells. Material and Methods We used pharmacological inhibition and genetic ablation to study effects of CDK12/CDK13 depletion on the proliferatory and migratory capacity of GBM cells and mouse xenografts. SLAM-seq, CUT&RUN and cell cycle assays were used to study the mechanistic effects of CDK12/CDK13 depletion in GBM cells. Results CDK12/CDK13 depletion markedly reduced the proliferatory and migratory capacity of GBM cells, as well as in vivo growth. CDK12/CDK13 inhibition potentiated existing chemotherapeutic treatments. Mechanistically, inhibition of CDK12/CDK13 leads to a genome-wide abrogation of RNAPII CTD phosphorylation, which in turn disrupts transcription and cell cycle progression in GBM cells. Conclusion These results provide proof-of-concept for the potential of CDK12 and CDK13 as therapeutic targets for GBM.
- Published
- 2022
3. PLZF targets developmental enhancers for activation during osteogenic differentiation of human mesenchymal stem cells
- Author
-
Singh, S.A., Lerdrup, M., Gomes, A.L.R., van de Werken, H.J.G., Johansen, J.V., Andersson, R., Sandelin, A., Helin, K. (Kristian), Hansen, K. (Klaus), Singh, S.A., Lerdrup, M., Gomes, A.L.R., van de Werken, H.J.G., Johansen, J.V., Andersson, R., Sandelin, A., Helin, K. (Kristian), and Hansen, K. (Klaus)
- Abstract
The PLZF transcription factor is essential for osteogenic differentiation of hMSCs; however, its regulation and molecular function during this process is not fully understood. Here, we revealed that the ZBTB16 locus encoding PLZF, is repressed by Polycomb (PcG) and H3K27me3 in naive hMSCs. At the pre-osteoblast stage of differentiation, the locus lost PcG binding and H3K27me3, gained JMJD3 recruitment, and H3K27ac resulting in high expression of PLZF. Subsequently, PLZF was recruited to osteogenic enhancers, influencing H3K27 acetylation and expression of nearby genes important for osteogenic function. Furthermore, we identified a latent enhancer within the ZBTB16/PLZF locus itself that became active, gained PLZF, p300 and Mediator binding and looped to the promoter of the nicotinamide N-methyltransferase (NNMT) gene. The increased expression of NNMT correlated with a decline in SAM levels, which is dependent on PLZF and is required for osteogenic differentiation
- Published
- 2019
- Full Text
- View/download PDF
4. PLZF targets developmental enhancers for activation during osteogenic differentiation of human mesenchymal stem cells
- Author
-
Singh, SA, Lerdrup, M, Gomes, ALR, van de Werken, Harmen, Johansen, JV, Andersson, R, Sandelin, A, Helin, K, Hansen, K, Singh, SA, Lerdrup, M, Gomes, ALR, van de Werken, Harmen, Johansen, JV, Andersson, R, Sandelin, A, Helin, K, and Hansen, K
- Published
- 2019
5. A histone H4K20 methylation-mediated chromatin compaction threshold ensures genome integrity by limiting DNA replication licensing
- Author
-
Shoaib, M, Walter, D, Gillespie, PJ, Izard, F, Fahrenkrog, B, Lleres, D, Lerdrup, M, Johansen, JV, Hansen, K, Julien, E, Blow, JJ, Sørensen, CS, Shoaib, M, Walter, D, Gillespie, PJ, Izard, F, Fahrenkrog, B, Lleres, D, Lerdrup, M, Johansen, JV, Hansen, K, Julien, E, Blow, JJ, and Sørensen, CS
- Published
- 2018
6. A Novel Specific Role for IB Kinase Complex-associated Protein in Cytosolic Stress Signaling
- Author
-
Holmberg, Christian Henrik, Katz, S., Lerdrup, M., Herdegen, T., Jaattela, M., Aronheim, A., Kallunki, T., Holmberg, Christian Henrik, Katz, S., Lerdrup, M., Herdegen, T., Jaattela, M., Aronheim, A., and Kallunki, T.
- Published
- 2002
7. Depletion of the AP-1 repressor JDP2 induces cell death similar to apoptosis
- Author
-
Lerdrup, M., primary, Holmberg, C., additional, Dietrich, N., additional, Shaulian, E., additional, Herdegen, T., additional, Jäättelä, M., additional, and Kallunki, T., additional
- Published
- 2005
- Full Text
- View/download PDF
8. JNK2 mediates TNF-induced cell death in mouse embryonic fibroblasts via regulation of both caspase and cathepsin protease pathways
- Author
-
Dietrich, N, primary, Thastrup, J, additional, Holmberg, C, additional, Gyrd-Hansen, M, additional, Fehrenbacher, N, additional, Lademann, U, additional, Lerdrup, M, additional, Herdegen, T, additional, Jäättelä, M, additional, and Kallunki, T, additional
- Published
- 2003
- Full Text
- View/download PDF
9. JNK2 mediates TNF-induced cell death in mouse embryonic fibroblasts via regulation of both caspase and cathepsin protease pathways.
- Author
-
Dietrich, N, Thastrup, J, Holmberg, C, Gyrd-Hansen, M, Fehrenbacher, N, Lademann, U, Lerdrup, M, Herdegen, T, Jäättelä, M, and Kallunki, T
- Subjects
TUMOR necrosis factors ,APOPTOSIS ,CYTOCHROME c ,CELL death ,FIBROBLASTS ,CYTOKINES - Abstract
Recent studies strongly suggest an active involvement of the c-Jun N-terminal kinase (JNK) signaling pathway in tumor necrosis factor (TNF)-induced apoptosis. The direct evidence for the role of JNK and its isoforms has been missing and the mechanism of how JNK actually could facilitate this process has remained unclear. In this study, we show that Jnk2-/- primary mouse embryonic fibroblasts (pMEFs) exhibit resistance towards TNF-induced apoptosis as compared to corresponding wild-type and Jnk1-/- pMEFs. JNK2-deficient pMEFs could be resensitized to TNF via retroviral transduction of any of the four different JNK2 splicing variants. Jnk2-/- pMEFs displayed deficient and delayed effector caspase activation as well as impaired cytosolic cystein cathepsin activity: processes that both were needed for efficient TNF-induced apoptosis in pMEFs. Our work demonstrates that JNK has a central role in the promotion of TNF-induced apoptosis in pMEFs, and that the JNK2 isoform can regulate both mitochondrial and lysosomal death pathways in these cells.Cell Death and Differentiation (2004) 11, 301-313. doi:10.1038/sj.cdd.4401353 Published online 12 December 2003 [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
10. The Histone Methyltransferase EZH2 Controls Drug Resistance in Acute Myeloid Leukemia (AML)
- Author
-
Goellner, S., Agrawal-Singh, S., Schenk, T., Klein, H. -U, Rohde, C., Sauer, T., Lerdrup, M., Tavor, S., Stoelzel, F., Ehninger, G., Koehler, G., Martin Dugas, Zelent, A., Thiede, C., Berdel, W. E., Hansen, K., and Mueller-Tidow, C.
11. A NOVEL EPIGENETIC RESISTANCE MECHANISM TO THE FLT3 INHIBITOR PKC412 INDUCES CROSS-RESISTANCE TO STANDARD CHEMOTHERAPY IN ACUTE MYELOID LEUKEMIA (AML)
- Author
-
Goellner, S., Agrawal-Singh, S., Oellerich, T., Schenk, T., Rohde, C., Klein, H. U., Sauer, T., Lerdrup, M., Tavor, S., Stoelzel, F., Ehninger, G., Koehler, G., Pan, K. T., Urlaub, H., Serve, H., Martin Dugas, Berdel, W. E., Hansen, K., Zelent, A., Thiede, C., and Mueller-Tidow, C.
12. Sex-specific DNA-replication in the early mammalian embryo.
- Author
-
Halliwell JA, Martin-Gonzalez J, Hashim A, Dahl JA, Hoffmann ER, and Lerdrup M
- Subjects
- Animals, Female, Mice, Male, Chromatin metabolism, Chromatin genetics, Oocytes metabolism, DNA Replication Timing, Genome, Embryonic Development genetics, Mice, Inbred C57BL, Zygote metabolism, Embryo, Mammalian metabolism, DNA Replication
- Abstract
The timing of DNA replication in mammals is crucial for minimizing errors and influenced by genome usage and chromatin states. Replication timing in the newly formed mammalian embryo remains poorly understood. Here, we have investigated replication timing in mouse zygotes and 2-cell embryos, revealing that zygotes lack a conventional replication timing program, which then emerges in 2-cell embryos. This program differs from embryonic stem cells and generally correlates with transcription and genome compartmentalization of both parental genomes. However, consistent and systematic differences existed between the replication timing of the two parental genomes, including considerably later replication of maternal pericentromeric regions compared to paternal counterparts. Moreover, maternal chromatin modified by Polycomb Repressive Complexes in the oocyte, undergoes early replication, despite belonging to the typically late-replicating B-compartment of the genome. This atypical and asynchronous replication of the two parental genomes may advance our understanding of replication stress in early human embryos and trigger strategies to reduce errors and aneuploidies., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Single-cell m 6 A mapping in vivo using picoMeRIP-seq.
- Author
-
Li Y, Wang Y, Vera-Rodriguez M, Lindeman LC, Skuggen LE, Rasmussen EMK, Jermstad I, Khan S, Fosslie M, Skuland T, Indahl M, Khodeer S, Klemsdal EK, Jin KX, Dalen KT, Fedorcsak P, Greggains GD, Lerdrup M, Klungland A, Au KF, and Dahl JA
- Subjects
- Animals, Mice, RNA, Messenger genetics, Embryonic Stem Cells, Cells, Cultured, Zebrafish genetics, Zebrafish metabolism, RNA genetics
- Abstract
Current N
6 -methyladenosine (m6 A) mapping methods need large amounts of RNA or are limited to cultured cells. Through optimized sample recovery and signal-to-noise ratio, we developed picogram-scale m6 A RNA immunoprecipitation and sequencing (picoMeRIP-seq) for studying m6 A in vivo in single cells and scarce cell types using standard laboratory equipment. We benchmark m6 A mapping on titrations of poly(A) RNA and embryonic stem cells and in single zebrafish zygotes, mouse oocytes and embryos., (© 2023. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
14. An organoid-based CRISPR-Cas9 screen for regulators of intestinal epithelial maturation and cell fate.
- Author
-
Hansen SL, Larsen HL, Pikkupeura LM, Maciag G, Guiu J, Müller I, Clement DL, Mueller C, Johansen JV, Helin K, Lerdrup M, and Jensen KB
- Subjects
- Animals, Mice, Cell Differentiation genetics, Fetus, Organoids, CRISPR-Cas Systems, Adult Stem Cells
- Abstract
Generation of functionally mature organs requires exquisite control of transcriptional programs governing cell state transitions during development. Despite advances in understanding the behavior of adult intestinal stem cells and their progeny, the transcriptional regulators that control the emergence of the mature intestinal phenotype remain largely unknown. Using mouse fetal and adult small intestinal organoids, we uncover transcriptional differences between the fetal and adult state and identify rare adult-like cells present in fetal organoids. This suggests that fetal organoids have an inherent potential to mature, which is locked by a regulatory program. By implementing a CRISPR-Cas9 screen targeting transcriptional regulators expressed in fetal organoids, we establish Smarca4 and Smarcc1 as important factors safeguarding the immature progenitor state. Our approach demonstrates the utility of organoid models in the identification of factors regulating cell fate and state transitions during tissue maturation and reveals that SMARCA4 and SMARCC1 prevent precocious differentiation during intestinal development.
- Published
- 2023
- Full Text
- View/download PDF
15. Integrator facilitates RNAPII removal to prevent transcription-replication collisions and genome instability.
- Author
-
Bhowmick R, Mehta KPM, Lerdrup M, and Cortez D
- Subjects
- Humans, Transcription, Genetic, DNA Helicases genetics, DNA Helicases metabolism, Genomic Instability, RNA Polymerase II genetics, RNA Polymerase II metabolism, DNA Replication
- Abstract
DNA replication preferentially initiates close to active transcription start sites (TSSs) in the human genome. Transcription proceeds discontinuously with an accumulation of RNA polymerase II (RNAPII) in a paused state near the TSS. Consequently, replication forks inevitably encounter paused RNAPII soon after replication initiates. Hence, dedicated machinery may be needed to remove RNAPII and facilitate unperturbed fork progression. In this study, we discovered that Integrator, a transcription termination machinery involved in the processing of RNAPII transcripts, interacts with the replicative helicase at active forks and promotes the removal of RNAPII from the path of the replication fork. Integrator-deficient cells have impaired replication fork progression and accumulate hallmarks of genome instability including chromosome breaks and micronuclei. The Integrator complex resolves co-directional transcription-replication conflicts to facilitate faithful DNA replication., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. Lamin A/C impairments cause mitochondrial dysfunction by attenuating PGC1α and the NAMPT-NAD+ pathway.
- Author
-
Maynard S, Hall A, Galanos P, Rizza S, Yamamoto T, Gram HH, Munk SHN, Shoaib M, Sørensen CS, Bohr VA, Lerdrup M, Maya-Mendoza A, and Bartek J
- Subjects
- Animals, Chromatin metabolism, DNA, Mitochondrial metabolism, Fibroblasts metabolism, Humans, Lamin Type A genetics, Mice, Mitochondria metabolism, NAD metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha genetics, Sirtuin 1 genetics, Lamin Type A metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Progeria metabolism
- Abstract
Mutations in the lamin A/C gene (LMNA) cause laminopathies such as the premature aging Hutchinson Gilford progeria syndrome (HGPS) and altered lamin A/C levels are found in diverse malignancies. The underlying lamin-associated mechanisms remain poorly understood. Here we report that lamin A/C-null mouse embryo fibroblasts (Lmna-/- MEFs) and human progerin-expressing HGPS fibroblasts both display reduced NAD+ levels, unstable mitochondrial DNA and attenuated bioenergetics. This mitochondrial dysfunction is associated with reduced chromatin recruitment (Lmna-/- MEFs) or low levels (HGPS) of PGC1α, the key transcription factor for mitochondrial homeostasis. Lmna-/- MEFs showed reduced expression of the NAD+-biosynthesis enzyme NAMPT and attenuated activity of the NAD+-dependent deacetylase SIRT1. We find high PARylation in lamin A/C-aberrant cells, further decreasing the NAD+ pool and consistent with impaired DNA base excision repair in both cell models, a condition that fuels DNA damage-induced PARylation under oxidative stress. Further, ATAC-sequencing revealed a substantially altered chromatin landscape in Lmna-/- MEFs, including aberrantly reduced accessibility at the Nampt gene promoter. Thus, we identified a new role of lamin A/C as a key modulator of mitochondrial function through impairments of PGC1α and the NAMPT-NAD+ pathway, with broader implications for the aging process., (© The Author(s) 2022. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
17. RAD51 protects human cells from transcription-replication conflicts.
- Author
-
Bhowmick R, Lerdrup M, Gadi SA, Rossetti GG, Singh MI, Liu Y, Halazonetis TD, and Hickson ID
- Subjects
- Chromosomes metabolism, Humans, S Phase genetics, Transcription, Genetic, DNA Replication, Rad51 Recombinase genetics, Rad51 Recombinase metabolism
- Abstract
Oncogene activation during tumorigenesis promotes DNA replication stress (RS), which subsequently drives the formation of cancer-associated chromosomal rearrangements. Many episodes of physiological RS likely arise due to conflicts between the DNA replication and transcription machineries operating simultaneously at the same loci. One role of the RAD51 recombinase in human cells is to protect replication forks undergoing RS. Here, we have identified a key role for RAD51 in preventing transcription-replication conflicts (TRCs) from triggering replication fork breakage. The genomic regions most affected by RAD51 deficiency are characterized by being replicated and transcribed in early S-phase and show significant overlap with loci prone to cancer-associated amplification. Consistent with a role for RAD51 in protecting against transcription-replication conflicts, many of the adverse effects of RAD51 depletion are ameliorated by inhibiting early S-phase transcription. We propose a model whereby RAD51 suppresses fork breakage and subsequent inadvertent amplification of genomic loci prone to experiencing TRCs., Competing Interests: Declaration of interests The authors confirm that there are no relevant financial or nonfinancial competing interests to report., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
18. The TRESLIN-MTBP complex couples completion of DNA replication with S/G2 transition.
- Author
-
Zonderland G, Vanzo R, Gadi SA, Martín-Doncel E, Coscia F, Mund A, Lerdrup M, Benada J, Boos D, and Toledo L
- Subjects
- Carrier Proteins metabolism, Checkpoint Kinase 1 genetics, DNA-Binding Proteins genetics, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, DNA Replication
- Abstract
It has been proposed that ATR kinase senses the completion of DNA replication to initiate the S/G2 transition. In contrast to this model, we show here that the TRESLIN-MTBP complex prevents a premature entry into G2 from early S-phase independently of ATR/CHK1 kinases. TRESLIN-MTBP acts transiently at pre-replication complexes (preRCs) to initiate origin firing and is released after the subsequent recruitment of CDC45. This dynamic behavior of TRESLIN-MTBP implements a monitoring system that checks the activation of replication forks and senses the rate of origin firing to prevent the entry into G2. This system detects the decline in the number of origins of replication that naturally occurs in very late S, which is the signature that cells use to determine the completion of DNA replication and permit the S/G2 transition. Our work introduces TRESLIN-MTBP as a key player in cell-cycle control independent of canonical checkpoints., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. Histone editing elucidates the functional roles of H3K27 methylation and acetylation in mammals.
- Author
-
Sankar A, Mohammad F, Sundaramurthy AK, Wang H, Lerdrup M, Tatar T, and Helin K
- Subjects
- Acetylation, Animals, Chromatin genetics, Chromatin metabolism, Mammals genetics, Methylation, Mice, Polycomb Repressive Complex 2 genetics, Protein Processing, Post-Translational genetics, Drosophila melanogaster genetics, Histones genetics, Histones metabolism
- Abstract
Posttranslational modifications of histones (PTMs) are associated with specific chromatin and gene expression states
1,2 . Although studies in Drosophila melanogaster have revealed phenotypic associations between chromatin-modifying enzymes and their histone substrates, comparable studies in mammalian models do not exist3-5 . Here, we use CRISPR base editing in mouse embryonic stem cells (mESCs) to address the regulatory role of lysine 27 of histone H3 (H3K27), a substrate for Polycomb repressive complex 2 (PRC2)-mediated methylation and CBP/EP300-mediated acetylation6,7 . By generating pan-H3K27R (pK27R) mutant mESCs, where all 28 alleles of H3.1, H3.2 and H3.3 have been mutated, we demonstrate similarity in transcription patterns of genes and differentiation to PRC2-null mutants. Moreover, H3K27 acetylation is not essential for gene derepression linked to loss of H3K27 methylation, or de novo activation of genes during cell-fate transition to epiblast-like cells (EpiLCs). In conclusion, our results show that H3K27 is an essential substrate for PRC2 in mESCs, whereas other PTMs in addition to H3K27 acetylation are likely involved in mediating CBP/EP300 function. Our work demonstrates the feasibility of large-scale multicopy gene editing to interrogate histone PTM function in mammalian cells., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
20. Histone H4 lysine 20 mono-methylation directly facilitates chromatin openness and promotes transcription of housekeeping genes.
- Author
-
Shoaib M, Chen Q, Shi X, Nair N, Prasanna C, Yang R, Walter D, Frederiksen KS, Einarsson H, Svensson JP, Liu CF, Ekwall K, Lerdrup M, Nordenskiöld L, and Sørensen CS
- Subjects
- Amino Acid Sequence, Animals, Cell Cycle genetics, Cell Line, Histone-Lysine N-Methyltransferase metabolism, Histones chemistry, Humans, Magnetic Resonance Spectroscopy, Methylation, Mice, Models, Biological, Nucleosomes metabolism, Protein Conformation, Chromatin metabolism, Genes, Essential, Histones metabolism, Lysine metabolism, Transcription, Genetic
- Abstract
Histone lysine methylations have primarily been linked to selective recruitment of reader or effector proteins that subsequently modify chromatin regions and mediate genome functions. Here, we describe a divergent role for histone H4 lysine 20 mono-methylation (H4K20me1) and demonstrate that it directly facilitates chromatin openness and accessibility by disrupting chromatin folding. Thus, accumulation of H4K20me1 demarcates highly accessible chromatin at genes, and this is maintained throughout the cell cycle. In vitro, H4K20me1-containing nucleosomal arrays with nucleosome repeat lengths (NRL) of 187 and 197 are less compact than unmethylated (H4K20me0) or trimethylated (H4K20me3) arrays. Concordantly, and in contrast to trimethylated and unmethylated tails, solid-state NMR data shows that H4K20 mono-methylation changes the H4 conformational state and leads to more dynamic histone H4-tails. Notably, the increased chromatin accessibility mediated by H4K20me1 facilitates gene expression, particularly of housekeeping genes. Altogether, we show how the methylation state of a single histone H4 residue operates as a focal point in chromatin structure control. While H4K20me1 directly promotes chromatin openness at highly transcribed genes, it also serves as a stepping-stone for H4K20me3-dependent chromatin compaction., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
21. Folate Deficiency Triggers the Abnormal Segregation of a Region With Large Cluster of CG-Rich Trinucleotide Repeats on Human Chromosome 2.
- Author
-
Garribba L, Vogel I, Lerdrup M, Gonçalves Dinis MM, Ren L, and Liu Y
- Abstract
Folate deficiency is associated with a broad range of human disorders, including anemia, fetal neural tube defects, age-associated dementia and several types of cancer. It is well established that a subgroup of rare fragile sites (RFSs) containing expanded CGG trinucleotide repeat (TNR) sequences display instability when cells are deprived of folate. However, given that folate sensitive RFSs exist in a very small percentage of the population, they are unlikely to be the cause of the widespread health problems associated with folate deficiency. We hypothesized that folate deficiency could specifically affect DNA replication at regions containing CG-rich repeat sequences. For this, we identified a region on human chromosome 2 (Chr2) comprising more than 300 CG-rich TNRs (termed "FOLD1") by examining the human genome database. Via the analysis of chromosome shape and segregation in mitosis, we demonstrate that, when human cells are cultured under folate stress conditions, Chr2 is prone to display a "kink" or "bending" at FOLD1 in metaphase and nondisjunction in anaphase. Furthermore, long-term folate deprivation causes Chr2 aneuploidy. Our results provide new evidence on the abnormalities folate deficiency could cause in human cells. This could facilitate future studies on the deleterious health conditions associated with folate deficiency., 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 Garribba, Vogel, Lerdrup, Gonçalves Dinis, Ren and Liu.)
- Published
- 2021
- Full Text
- View/download PDF
22. Mutant FOXL2 C134W Hijacks SMAD4 and SMAD2/3 to Drive Adult Granulosa Cell Tumors.
- Author
-
Weis-Banke SE, Lerdrup M, Kleine-Kohlbrecher D, Mohammad F, Sidoli S, Jensen ON, Yanase T, Nakamura T, Iwase A, Stylianou A, Abu-Rustum NR, Aghajanian C, Soslow R, Da Cruz Paula A, Koche RP, Weigelt B, Christensen J, Helin K, and Cloos PAC
- Subjects
- Cell Line, Tumor, Cells, Cultured, Female, Forkhead Box Protein L2 metabolism, Granulosa Cell Tumor genetics, Granulosa Cell Tumor metabolism, Humans, Mutation, Smad2 Protein metabolism, Smad3 Protein metabolism, Smad4 Protein metabolism, Epithelial-Mesenchymal Transition genetics, Forkhead Box Protein L2 genetics, Gene Expression Regulation, Neoplastic genetics, Granulosa Cell Tumor pathology, Smad Proteins metabolism
- Abstract
The mutant protein FOXL2
C134W is expressed in at least 95% of adult-type ovarian granulosa cell tumors (AGCT) and is considered to be a driver of oncogenesis in this disease. However, the molecular mechanism by which FOXL2C134W contributes to tumorigenesis is not known. Here, we show that mutant FOXL2C134W acquires the ability to bind SMAD4, forming a FOXL2C134W /SMAD4/SMAD2/3 complex that binds a novel hybrid DNA motif AGHCAHAA, unique to the FOXL2C134W mutant. This binding induced an enhancer-like chromatin state, leading to transcription of nearby genes, many of which are characteristic of epithelial-to-mesenchymal transition. FOXL2C134W also bound hybrid loci in primary AGCT. Ablation of SMAD4 or SMAD2/3 resulted in strong reduction of FOXL2C134W binding at hybrid sites and decreased expression of associated genes. Accordingly, inhibition of TGFβ mitigated the transcriptional effect of FOXL2C134W . Our results provide mechanistic insight into AGCT pathogenesis, identifying FOXL2C134W and its interaction with SMAD4 as potential therapeutic targets to this condition. SIGNIFICANCE: FOXL2C134W hijacks SMAD4 and leads to the expression of genes involved in EMT, stemness, and oncogenesis in AGCT, making FOXL2C134W and the TGFβ pathway therapeutic targets in this condition. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/17/3466/F1.large.jpg., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
23. KDM4A regulates the maternal-to-zygotic transition by protecting broad H3K4me3 domains from H3K9me3 invasion in oocytes.
- Author
-
Sankar A, Lerdrup M, Manaf A, Johansen JV, Gonzalez JM, Borup R, Blanshard R, Klungland A, Hansen K, Andersen CY, Dahl JA, Helin K, and Hoffmann ER
- Subjects
- Animals, Embryo Implantation, Embryo, Mammalian, Female, Fertilization genetics, Heterochromatin chemistry, Heterochromatin metabolism, Histone Demethylases genetics, Histones genetics, Male, Metaphase, Methylation, Mice, Mice, Knockout, Oocytes cytology, Oocytes growth & development, Promoter Regions, Genetic, Transcription, Genetic, Zygote cytology, Zygote growth & development, Histone Demethylases metabolism, Histones metabolism, Oocytes metabolism, Protein Processing, Post-Translational, Zygote metabolism
- Abstract
The importance of germline-inherited post-translational histone modifications on priming early mammalian development is just emerging
1-4 . Histone H3 lysine 9 (H3K9) trimethylation is associated with heterochromatin and gene repression during cell-fate change5 , whereas histone H3 lysine 4 (H3K4) trimethylation marks active gene promoters6 . Mature oocytes are transcriptionally quiescent and possess remarkably broad domains of H3K4me3 (bdH3K4me3)1,2 . It is unknown which factors contribute to the maintenance of the bdH3K4me3 landscape. Lysine-specific demethylase 4A (KDM4A) demethylates H3K9me3 at promoters marked by H3K4me3 in actively transcribing somatic cells7 . Here, we report that KDM4A-mediated H3K9me3 demethylation at bdH3K4me3 in oocytes is crucial for normal pre-implantation development and zygotic genome activation after fertilization. The loss of KDM4A in oocytes causes aberrant H3K9me3 spreading over bdH3K4me3, resulting in insufficient transcriptional activation of genes, endogenous retroviral elements and chimeric transcripts initiated from long terminal repeats during zygotic genome activation. The catalytic activity of KDM4A is essential for normal epigenetic reprogramming and pre-implantation development. Hence, KDM4A plays a crucial role in preserving the maternal epigenome integrity required for proper zygotic genome activation and transfer of developmental control to the embryo.- Published
- 2020
- Full Text
- View/download PDF
24. User-Friendly and Interactive Analysis of ChIP-Seq Data Using EaSeq.
- Author
-
Lerdrup M and Hansen K
- Subjects
- Animals, Binding Sites, Gene Regulatory Networks, High-Throughput Nucleotide Sequencing, Histones metabolism, Humans, Mice, Sequence Analysis, DNA, Software, Transcription Factors metabolism, User-Computer Interface, Workflow, Chromatin Immunoprecipitation Sequencing methods, Computational Biology methods, DNA chemistry, DNA metabolism
- Abstract
ChIP-seq is a central method to gain understanding of the regulatory networks in the genome of stem cells and during differentiation. Exploration and analysis of such genome-wide data often leads to unexpected discoveries and new hypotheses. It therefore accelerates and improves the discovery phase, when scientists with biological understanding are enabled to analyze and visualize data. EaSeq ( http://easeq.net ) offers integrated exploration of genome-wide data in a visual, versatile, user-friendly, and interactive manner that connects abstract interpretations to the signal distribution at the underlying loci. Here we introduce the interface, data types, and acquisition, and guide the reader through two example workflows. These workflows will enable the reader to perform genome-wide analysis and visualization of transcription factor binding sites and histone marks. This includes making basic plots; finding, annotating, sorting, and filtering of peaks; using EaSeq as a genome browser; measuring ChIP-seq signal and calculating ratios; as well as data import and export.
- Published
- 2020
- Full Text
- View/download PDF
25. Going low to reach high: Small-scale ChIP-seq maps new terrain.
- Author
-
Fosslie M, Manaf A, Lerdrup M, Hansen K, Gilfillan GD, and Dahl JA
- Subjects
- Animals, Cell Line, Tumor, Cells, Cultured, Genome genetics, Genomics, Histones genetics, Histones metabolism, Mice, Microfluidic Analytical Techniques, Oocytes metabolism, Chromatin Immunoprecipitation Sequencing
- Abstract
Chromatin immunoprecipitation (ChIP) enables mapping of specific histone modifications or chromatin-associated factors in the genome and represents a powerful tool in the study of chromatin and genome regulation. Importantly, recent technological advances that couple ChIP with whole-genome high-throughput sequencing (ChIP-seq) now allow the mapping of chromatin factors throughout the genome. However, the requirement for large amounts of ChIP-seq input material has long made it challenging to assess chromatin profiles of cell types only available in limited numbers. For many cell types, it is not feasible to reach high numbers when collecting them as homogeneous cell populations in vivo. Nonetheless, it is an advantage to work with pure cell populations to reach robust biological conclusions. Here, we review (a) how ChIP protocols have been scaled down for use with as little as a few hundred cells; (b) which considerations to be aware of when preparing small-scale ChIP-seq and analyzing data; and (c) the potential of small-scale ChIP-seq datasets for elucidating chromatin dynamics in various biological systems, including some examples such as oocyte maturation and preimplantation embryo development. This article is categorized under: Laboratory Methods and Technologies > Genetic/Genomic Methods Developmental Biology > Developmental Processes in Health and Disease Biological Mechanisms > Cell Fates., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
26. PLZF targets developmental enhancers for activation during osteogenic differentiation of human mesenchymal stem cells.
- Author
-
Agrawal Singh S, Lerdrup M, Gomes AR, van de Werken HJ, Vilstrup Johansen J, Andersson R, Sandelin A, Helin K, and Hansen K
- Subjects
- Acetylation, Cell Lineage genetics, Chromatin metabolism, Epigenesis, Genetic, Genetic Loci, Histones metabolism, Humans, Lysine metabolism, Nicotinamide N-Methyltransferase genetics, Nicotinamide N-Methyltransferase metabolism, Promoter Regions, Genetic, Promyelocytic Leukemia Zinc Finger Protein genetics, Protein Binding, RNA genetics, Transcriptome genetics, Cell Differentiation genetics, Embryonic Development genetics, Enhancer Elements, Genetic genetics, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Osteogenesis genetics, Promyelocytic Leukemia Zinc Finger Protein metabolism
- Abstract
The PLZF transcription factor is essential for osteogenic differentiation of hMSCs; however, its regulation and molecular function during this process is not fully understood. Here, we revealed that the ZBTB16 locus encoding PLZF, is repressed by Polycomb (PcG) and H3K27me3 in naive hMSCs. At the pre-osteoblast stage of differentiation, the locus lost PcG binding and H3K27me3, gained JMJD3 recruitment, and H3K27ac resulting in high expression of PLZF. Subsequently, PLZF was recruited to osteogenic enhancers, influencing H3K27 acetylation and expression of nearby genes important for osteogenic function. Furthermore, we identified a latent enhancer within the ZBTB16/PLZF locus itself that became active, gained PLZF, p300 and Mediator binding and looped to the promoter of the nicotinamide N-methyltransferase ( NNMT ) gene. The increased expression of NNMT correlated with a decline in SAM levels, which is dependent on PLZF and is required for osteogenic differentiation., Competing Interests: SA, ML, AG, Hv, RA, AS, KH, KH No competing interests declared, (© 2019, Agrawal Singh et al.)
- Published
- 2019
- Full Text
- View/download PDF
27. Proteogenomic Characterization of Patient-Derived Xenografts Highlights the Role of REST in Neuroendocrine Differentiation of Castration-Resistant Prostate Cancer.
- Author
-
Flores-Morales A, Bergmann TB, Lavallee C, Batth TS, Lin D, Lerdrup M, Friis S, Bartels A, Kristensen G, Krzyzanowska A, Xue H, Fazli L, Hansen KH, Røder MA, Brasso K, Moreira JM, Bjartell A, Wang Y, Olsen JV, Collins CC, and Iglesias-Gato D
- Subjects
- Animals, Carcinoma, Neuroendocrine pathology, Cell Cycle genetics, Cell Line, Tumor, Computational Biology methods, Disease Models, Animal, Disease Susceptibility, Gene Expression Profiling, Heterografts, Humans, Male, Mice, Proportional Hazards Models, Prostatectomy, Prostatic Neoplasms, Castration-Resistant pathology, Prostatic Neoplasms, Castration-Resistant surgery, Carcinoma, Neuroendocrine genetics, Carcinoma, Neuroendocrine metabolism, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant metabolism, Proteogenomics methods, Repressor Proteins genetics, Repressor Proteins metabolism
- Abstract
Purpose: An increasing number of castration-resistant prostate cancer (CRPC) tumors exhibit neuroendocrine (NE) features. NE prostate cancer (NEPC) has poor prognosis, and its development is poorly understood. Experimental Design: We applied mass spectrometry-based proteomics to a unique set of 17 prostate cancer patient-derived xenografts (PDX) to characterize the effects of castration in vivo , and the proteome differences between NEPC and prostate adenocarcinomas. Genome-wide profiling of REST-occupied regions in prostate cancer cells was correlated to the expression changes in vivo to investigate the role of the transcriptional repressor REST in castration-induced NEPC differentiation., Results: An average of 4,881 proteins were identified and quantified from each PDX. Proteins related to neurogenesis, cell-cycle regulation, and DNA repair were found upregulated and elevated in NEPC, while the reduced levels of proteins involved in mitochondrial functions suggested a prevalent glycolytic metabolism of NEPC tumors. Integration of the REST chromatin bound regions with expression changes indicated a direct role of REST in regulating neuronal gene expression in prostate cancer cells. Mechanistically, depletion of REST led to cell-cycle arrest in G
1 , which could be rescued by p53 knockdown. Finally, the expression of the REST-regulated gene secretagogin (SCGN) correlated with an increased risk of suffering disease relapse after radical prostatectomy., Conclusions: This study presents the first deep characterization of the proteome of NEPC and suggests that concomitant inhibition of REST and the p53 pathway would promote NEPC. We also identify SCGN as a novel prognostic marker in prostate cancer., (©2018 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
28. Histone H4K20 methylation mediated chromatin compaction threshold ensures genome integrity by limiting DNA replication licensing.
- Author
-
Shoaib M, Walter D, Gillespie PJ, Izard F, Fahrenkrog B, Lleres D, Lerdrup M, Johansen JV, Hansen K, Julien E, Blow JJ, and Sørensen CS
- Subjects
- Cell Line, Tumor, Chromatin genetics, DNA Damage genetics, DNA Damage physiology, DNA Replication genetics, Flow Cytometry, Histones genetics, Humans, Microscopy, Fluorescence, RNA, Small Interfering genetics, Chromatin metabolism, DNA Replication physiology, Histones metabolism
- Abstract
The decompaction and re-establishment of chromatin organization immediately after mitosis is essential for genome regulation. Mechanisms underlying chromatin structure control in daughter cells are not fully understood. Here we show that a chromatin compaction threshold in cells exiting mitosis ensures genome integrity by limiting replication licensing in G1 phase. Upon mitotic exit, chromatin relaxation is controlled by SET8-dependent methylation of histone H4 on lysine 20. In the absence of either SET8 or H4K20 residue, substantial genome-wide chromatin decompaction occurs allowing excessive loading of the origin recognition complex (ORC) in the daughter cells. ORC overloading stimulates aberrant recruitment of the MCM2-7 complex that promotes single-stranded DNA formation and DNA damage. Restoring chromatin compaction restrains excess replication licensing and loss of genome integrity. Our findings identify a cell cycle-specific mechanism whereby fine-tuned chromatin relaxation suppresses excessive detrimental replication licensing and maintains genome integrity at the cellular transition from mitosis to G1 phase.
- Published
- 2018
- Full Text
- View/download PDF
29. Loss of the histone methyltransferase EZH2 induces resistance to multiple drugs in acute myeloid leukemia.
- Author
-
Göllner S, Oellerich T, Agrawal-Singh S, Schenk T, Klein HU, Rohde C, Pabst C, Sauer T, Lerdrup M, Tavor S, Stölzel F, Herold S, Ehninger G, Köhler G, Pan KT, Urlaub H, Serve H, Dugas M, Spiekermann K, Vick B, Jeremias I, Berdel WE, Hansen K, Zelent A, Wickenhauser C, Müller LP, Thiede C, and Müller-Tidow C
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Antineoplastic Agents pharmacology, Blotting, Western, Bortezomib pharmacology, CDC2 Protein Kinase, Cell Line, Tumor, Cyclin-Dependent Kinases metabolism, Cytarabine pharmacology, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enhancer of Zeste Homolog 2 Protein metabolism, Female, Flow Cytometry, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, HSP90 Heat-Shock Proteins metabolism, Homeodomain Proteins genetics, Humans, Immunohistochemistry, Immunoprecipitation, Indoles pharmacology, Leukemia, Myeloid, Acute genetics, Male, Mass Spectrometry, Mice, Middle Aged, Neoplasm Transplantation, Proteasome Endopeptidase Complex metabolism, Protein Processing, Post-Translational, Proteomics, Pyridones pharmacology, Young Adult, Drug Resistance, Neoplasm genetics, Enhancer of Zeste Homolog 2 Protein genetics, Histones metabolism, Leukemia, Myeloid, Acute drug therapy, Protein Kinase Inhibitors
- Abstract
In acute myeloid leukemia (AML), therapy resistance frequently occurs, leading to high mortality among patients. However, the mechanisms that render leukemic cells drug resistant remain largely undefined. Here, we identified loss of the histone methyltransferase EZH2 and subsequent reduction of histone H3K27 trimethylation as a novel pathway of acquired resistance to tyrosine kinase inhibitors (TKIs) and cytotoxic drugs in AML. Low EZH2 protein levels correlated with poor prognosis in AML patients. Suppression of EZH2 protein expression induced chemoresistance of AML cell lines and primary cells in vitro and in vivo. Low EZH2 levels resulted in derepression of HOX genes, and knockdown of HOXB7 and HOXA9 in the resistant cells was sufficient to improve sensitivity to TKIs and cytotoxic drugs. The endogenous loss of EZH2 expression in resistant cells and primary blasts from a subset of relapsed AML patients resulted from enhanced CDK1-dependent phosphorylation of EZH2 at Thr487. This interaction was stabilized by heat shock protein 90 (HSP90) and followed by proteasomal degradation of EZH2 in drug-resistant cells. Accordingly, inhibitors of HSP90, CDK1 and the proteasome prevented EZH2 degradation, decreased HOX gene expression and restored drug sensitivity. Finally, patients with reduced EZH2 levels at progression to standard therapy responded to the combination of bortezomib and cytarabine, concomitant with the re-establishment of EZH2 expression and blast clearance. These data suggest restoration of EZH2 protein as a viable approach to overcome treatment resistance in this AML patient population.
- Published
- 2017
- Full Text
- View/download PDF
30. Broad histone H3K4me3 domains in mouse oocytes modulate maternal-to-zygotic transition.
- Author
-
Dahl JA, Jung I, Aanes H, Greggains GD, Manaf A, Lerdrup M, Li G, Kuan S, Li B, Lee AY, Preissl S, Jermstad I, Haugen MH, Suganthan R, Bjørås M, Hansen K, Dalen KT, Fedorcsak P, Ren B, and Klungland A
- Subjects
- Acetylation, Animals, Cell Line, Tumor, Chromatin genetics, Chromatin Immunoprecipitation, Embryonic Development genetics, Female, Genome genetics, Histones chemistry, Humans, Male, Methylation, Mice, Sequence Analysis, DNA, Transcription Initiation Site, Zygote cytology, Chromatin metabolism, DNA Methylation, Gene Expression Regulation, Developmental, Histones metabolism, Lysine metabolism, Oocytes metabolism, Zygote metabolism
- Abstract
Maternal-to-zygotic transition (MZT) is essential for the formation of a new individual, but is still poorly understood despite recent progress in analysis of gene expression and DNA methylation in early embryogenesis. Dynamic histone modifications may have important roles in MZT, but direct measurements of chromatin states have been hindered by technical difficulties in profiling histone modifications from small quantities of cells. Recent improvements allow for 500 cell-equivalents of chromatin per reaction, but require 10,000 cells for initial steps or require a highly specialized microfluidics device that is not readily available. We developed a micro-scale chromatin immunoprecipitation and sequencing (μChIP-seq) method, which we used to profile genome-wide histone H3 lysine methylation (H3K4me3) and acetylation (H3K27ac) in mouse immature and metaphase II oocytes and in 2-cell and 8-cell embryos. Notably, we show that ~22% of the oocyte genome is associated with broad H3K4me3 domains that are anti-correlated with DNA methylation. The H3K4me3 signal becomes confined to transcriptional-start-site regions in 2-cell embryos, concomitant with the onset of major zygotic genome activation. Active removal of broad H3K4me3 domains by the lysine demethylases KDM5A and KDM5B is required for normal zygotic genome activation and is essential for early embryo development. Our results provide insight into the onset of the developmental program in mouse embryos and demonstrate a role for broad H3K4me3 domains in MZT.
- Published
- 2016
- Full Text
- View/download PDF
31. An interactive environment for agile analysis and visualization of ChIP-sequencing data.
- Author
-
Lerdrup M, Johansen JV, Agrawal-Singh S, and Hansen K
- Subjects
- Animals, Genome, Mice, Polycomb-Group Proteins analysis, Chromatin Immunoprecipitation methods, Software
- Abstract
To empower experimentalists with a means for fast and comprehensive chromatin immunoprecipitation sequencing (ChIP-seq) data analyses, we introduce an integrated computational environment, EaSeq. The software combines the exploratory power of genome browsers with an extensive set of interactive and user-friendly tools for genome-wide abstraction and visualization. It enables experimentalists to easily extract information and generate hypotheses from their own data and public genome-wide datasets. For demonstration purposes, we performed meta-analyses of public Polycomb ChIP-seq data and established a new screening approach to analyze more than 900 datasets from mouse embryonic stem cells for factors potentially associated with Polycomb recruitment. EaSeq, which is freely available and works on a standard personal computer, can substantially increase the throughput of many analysis workflows, facilitate transparency and reproducibility by automatically documenting and organizing analyses, and enable a broader group of scientists to gain insights from ChIP-seq data.
- Published
- 2016
- Full Text
- View/download PDF
32. β-Catenin Regulates Primitive Streak Induction through Collaborative Interactions with SMAD2/SMAD3 and OCT4.
- Author
-
Funa NS, Schachter KA, Lerdrup M, Ekberg J, Hess K, Dietrich N, Honoré C, Hansen K, and Semb H
- Subjects
- Base Sequence, Cell Line, Cell Lineage, Gene Expression Regulation, Developmental, Humans, Models, Biological, Molecular Sequence Data, Neural Crest cytology, Nodal Protein metabolism, Promoter Regions, Genetic genetics, Protein Binding, Stem Cells cytology, Stem Cells metabolism, Wnt Signaling Pathway genetics, Octamer Transcription Factor-3 metabolism, Primitive Streak metabolism, Smad2 Protein metabolism, Smad3 Protein metabolism, beta Catenin metabolism
- Abstract
Canonical Wnt and Nodal signaling are both required for induction of the primitive streak (PS), which guides organization of the early embryo. The Wnt effector β-catenin is thought to function in these early lineage specification decisions via transcriptional activation of Nodal signaling. Here, we demonstrate a broader role for β-catenin in PS formation by analyzing its genome-wide binding in a human embryonic stem cell model of PS induction. β-catenin occupies regulatory regions in numerous PS and neural crest genes, and direct interactions between β-catenin and the Nodal effectors SMAD2/SMAD3 are required at these regions for PS gene activation. Furthermore, OCT4 binding in proximity to these sites is likewise required for PS induction, suggesting a collaborative interaction between β-catenin and OCT4. Induction of neural crest genes by β-catenin is repressed by SMAD2/SMAD3, ensuring proper lineage specification. This study provides mechanistic insight into how Wnt signaling controls early cell lineage decisions., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
33. Dopamine signaling leads to loss of Polycomb repression and aberrant gene activation in experimental parkinsonism.
- Author
-
Södersten E, Feyder M, Lerdrup M, Gomes AL, Kryh H, Spigolon G, Caboche J, Fisone G, and Hansen K
- Subjects
- Animals, Disease Models, Animal, Dopamine and cAMP-Regulated Phosphoprotein 32 metabolism, Female, Genetic Loci, Histones metabolism, Levodopa pharmacology, Mice, Mice, Knockout, Phosphorylation drug effects, Protein Binding, RNA, Messenger genetics, Ribosomal Protein S6 Kinases, 90-kDa metabolism, Dopamine metabolism, Gene Expression Regulation drug effects, Parkinsonian Disorders genetics, Parkinsonian Disorders metabolism, Polycomb-Group Proteins genetics, Signal Transduction, Transcriptional Activation
- Abstract
Polycomb group (PcG) proteins bind to and repress genes in embryonic stem cells through lineage commitment to the terminal differentiated state. PcG repressed genes are commonly characterized by the presence of the epigenetic histone mark H3K27me3, catalyzed by the Polycomb repressive complex 2. Here, we present in vivo evidence for a previously unrecognized plasticity of PcG-repressed genes in terminally differentiated brain neurons of parkisonian mice. We show that acute administration of the dopamine precursor, L-DOPA, induces a remarkable increase in H3K27me3S28 phosphorylation. The induction of the H3K27me3S28p histone mark specifically occurs in medium spiny neurons expressing dopamine D1 receptors and is dependent on Msk1 kinase activity and DARPP-32-mediated inhibition of protein phosphatase-1. Chromatin immunoprecipitation (ChIP) experiments showed that increased H3K27me3S28p was accompanied by reduced PcG binding to regulatory regions of genes. An analysis of the genome wide distribution of L-DOPA-induced H3K27me3S28 phosphorylation by ChIP sequencing (ChIP-seq) in combination with expression analysis by RNA-sequencing (RNA-seq) showed that the induction of H3K27me3S28p correlated with increased expression of a subset of PcG repressed genes. We found that induction of H3K27me3S28p persisted during chronic L-DOPA administration to parkisonian mice and correlated with aberrant gene expression. We propose that dopaminergic transmission can activate PcG repressed genes in the adult brain and thereby contribute to long-term maladaptive responses including the motor complications, or dyskinesia, caused by prolonged administration of L-DOPA in Parkinson's disease.
- Published
- 2014
- Full Text
- View/download PDF
34. DNA methylation changes are a late event in acute promyelocytic leukemia and coincide with loss of transcription factor binding.
- Author
-
Schoofs T, Rohde C, Hebestreit K, Klein HU, Göllner S, Schulze I, Lerdrup M, Dietrich N, Agrawal-Singh S, Witten A, Stoll M, Lengfelder E, Hofmann WK, Schlenke P, Büchner T, Hansen K, Berdel WE, Rosenbauer F, Dugas M, and Müller-Tidow C
- Subjects
- Animals, Cell Transformation, Neoplastic genetics, Chromosomes, Human ultrastructure, CpG Islands, DNA, Neoplasm metabolism, Disease Progression, Gene Knock-In Techniques, Hematopoietic Stem Cells metabolism, Humans, Leukemia, Promyelocytic, Acute drug therapy, Mice, Mice, Inbred C57BL, Neoplasm Proteins physiology, Neoplastic Stem Cells metabolism, Oncogene Proteins, Fusion physiology, Phenotype, Polycomb Repressive Complex 2 metabolism, Preleukemia genetics, Recombinant Fusion Proteins physiology, Repressor Proteins metabolism, Tretinoin therapeutic use, DNA Methylation, DNA, Neoplasm genetics, Gene Expression Regulation, Leukemic, Leukemia, Promyelocytic, Acute genetics, Transcription Factors metabolism
- Abstract
The origin of aberrant DNA methylation in cancer remains largely unknown. In the present study, we elucidated the DNA methylome in primary acute promyelocytic leukemia (APL) and the role of promyelocytic leukemia-retinoic acid receptor α (PML-RARα) in establishing these patterns. Cells from APL patients showed increased genome-wide DNA methylation with higher variability than healthy CD34(+) cells, promyelocytes, and remission BM cells. A core set of differentially methylated regions in APL was identified. Age at diagnosis, Sanz score, and Flt3-mutation status characterized methylation subtypes. Transcription factor-binding sites (eg, the c-myc-binding sites) were associated with low methylation. However, SUZ12- and REST-binding sites identified in embryonic stem cells were preferentially DNA hypermethylated in APL cells. Unexpectedly, PML-RARα-binding sites were also protected from aberrant DNA methylation in APL cells. Consistent with this, myeloid cells from preleukemic PML-RARα knock-in mice did not show altered DNA methylation and the expression of PML-RARα in hematopoietic progenitor cells prevented differentiation without affecting DNA methylation. Treatment of APL blasts with all-trans retinoic acid also did not result in immediate DNA methylation changes. The results of the present study suggest that aberrant DNA methylation is associated with leukemia phenotype but is not required for PML-RARα-mediated initiation of leukemogenesis.
- Published
- 2013
- Full Text
- View/download PDF
35. REST-mediated recruitment of polycomb repressor complexes in mammalian cells.
- Author
-
Dietrich N, Lerdrup M, Landt E, Agrawal-Singh S, Bak M, Tommerup N, Rappsilber J, Södersten E, and Hansen K
- Subjects
- Animals, Binding Sites, Embryonic Stem Cells cytology, Embryonic Stem Cells metabolism, Gene Expression Regulation, Developmental, HEK293 Cells, Humans, Jumonji Domain-Containing Histone Demethylases metabolism, Mice, Neurons metabolism, Polycomb-Group Proteins, Protein Binding, Teratoma genetics, Tretinoin pharmacology, Cell Differentiation drug effects, CpG Islands genetics, Repressor Proteins genetics, Repressor Proteins metabolism
- Abstract
Polycomb Repressive Complex (PRC) 1 and PRC2 regulate genes involved in differentiation and development. However, the mechanism for how PRC1 and PRC2 are recruited to genes in mammalian cells is unclear. Here we present evidence for an interaction between the transcription factor REST, PRC1, and PRC2 and show that RNF2 and REST co-regulate a number of neuronal genes in human teratocarcinoma cells (NT2-D1). Using NT2-D1 cells as a model of neuronal differentiation, we furthermore showed that retinoic-acid stimulation led to displacement of PRC1 at REST binding sites, reduced H3K27Me3, and increased gene expression. Genome-wide analysis of Polycomb binding in Rest⁻/⁻ and Eed⁻/⁻ mouse embryonic stem (mES) cells showed that Rest was required for PRC1 recruitment to a subset of Polycomb regulated neuronal genes. Furthermore, we found that PRC1 can be recruited to Rest binding sites independently of CpG islands and the H3K27Me3 mark. Surprisingly, PRC2 was frequently increased around Rest binding sites located in CpG-rich regions in the Rest⁻/⁻ mES cells, indicating a more complex interplay where Rest also can limit PRC2 recruitment. Therefore, we propose that Rest has context-dependent functions for PRC1- and PRC2- recruitment, which allows this transcription factor to act both as a recruiter of Polycomb as well as a limiting factor for PRC2 recruitment at CpG islands., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2012
- Full Text
- View/download PDF
36. Differential effects of EGFR ligands on endocytic sorting of the receptor.
- Author
-
Roepstorff K, Grandal MV, Henriksen L, Knudsen SL, Lerdrup M, Grøvdal L, Willumsen BM, and van Deurs B
- Subjects
- Amphiregulin, Animals, Betacellulin, Cell Line, EGF Family of Proteins, Epidermal Growth Factor metabolism, Epiregulin, ErbB Receptors genetics, Glycoproteins metabolism, Heparin-binding EGF-like Growth Factor, Humans, Hydrogen-Ion Concentration, Intercellular Signaling Peptides and Proteins metabolism, Lysosomal-Associated Membrane Protein 1 metabolism, Phosphorylation, Proto-Oncogene Proteins c-cbl metabolism, Signal Transduction physiology, Transforming Growth Factor alpha metabolism, Ubiquitination, Vesicular Transport Proteins metabolism, Endocytosis physiology, ErbB Receptors metabolism, Ligands, Protein Transport physiology
- Abstract
Endocytic downregulation is a pivotal mechanism turning off signalling from the EGF receptor (EGFR). It is well established that whereas EGF binding leads to lysosomal degradation of EGFR, transforming growth factor (TGF)-alpha causes receptor recycling. TGF-alpha therefore leads to continuous signalling and is a more potent mitogen than EGF. In addition to EGF and TGF-alpha, five EGFR ligands have been identified. Although many of these ligands are upregulated in cancers, very little is known about their effect on EGFR trafficking. We have compared the effect of six different ligands on endocytic trafficking of EGFR. We find that, whereas they all stimulate receptor internalization, they have very diverse effects on endocytic sorting. Heparin-binding EGF-like growth factor and Betacellulin target all EGFRs for lysosomal degradation. In contrast, TGF-alpha and epiregulin lead to complete receptor recycling. EGF leads to lysosomal degradation of the majority but not all EGFRs. Amphiregulin does not target EGFR for lysosomal degradation but causes fast as well as slow EGFR recycling. The Cbl ubiquitin ligases, especially c-Cbl, are responsible for EGFR ubiquitination after stimulation with all ligands, and persistent EGFR phosphorylation and ubiquitination largely correlate with receptor degradation.
- Published
- 2009
- Full Text
- View/download PDF
37. A model for transmission of the H3K27me3 epigenetic mark.
- Author
-
Hansen KH, Bracken AP, Pasini D, Dietrich N, Gehani SS, Monrad A, Rappsilber J, Lerdrup M, and Helin K
- Subjects
- Carrier Proteins genetics, Carrier Proteins metabolism, Catalysis, Cell Line, Cells, Cultured, Chromatin genetics, Chromatin metabolism, DNA-Binding Proteins genetics, Enhancer of Zeste Homolog 2 Protein, Fibroblasts metabolism, G1 Phase physiology, Genes, Reporter, Histones genetics, Humans, Kidney cytology, Luciferases metabolism, Lysine genetics, Lysine metabolism, Methylation, Mutation, Neoplasm Proteins, Nuclear Proteins genetics, Nuclear Proteins metabolism, Polycomb Repressive Complex 2, Polycomb-Group Proteins, Promoter Regions, Genetic, RNA, Small Interfering metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, S Phase physiology, Transcription Factors genetics, Transfection, DNA-Binding Proteins metabolism, Epigenesis, Genetic, Histones metabolism, Models, Biological, Transcription Factors metabolism
- Abstract
Organization of chromatin by epigenetic mechanisms is essential for establishing and maintaining cellular identity in developing and adult organisms. A key question that remains unresolved about this process is how epigenetic marks are transmitted to the next cell generation during cell division. Here we provide a model to explain how trimethylated Lys 27 of histone 3 (H3K27me3), which is catalysed by the EZH2-containing Polycomb Repressive Complex 2 (PRC2), is maintained in proliferating cells. We show that the PRC2 complex binds to the H3K27me3 mark and colocalizes with this mark in G1 phase and with sites of ongoing DNA replication. Efficient binding requires an intact trimeric PRC2 complex containing EZH2, EED and SUZ12, but is independent of the catalytic SET domain of EZH2. Using a heterologous reporter system, we show that transient recruitment of the PRC2 complex to chromatin, upstream of the transcriptional start site, is sufficient to maintain repression through endogenous PRC2 during subsequent cell divisions. Thus, we suggest that once the H3K27me3 is established, it recruits the PRC2 complex to maintain the mark at sites of DNA replication, leading to methylation of H3K27 on the daughter strands during incorporation of newly synthesized histones. This mechanism ensures maintenance of the H3K27me3 epigenetic mark in proliferating cells, not only during DNA replication when histones synthesized de novo are incorporated, but also outside S phase, thereby preserving chromatin structure and transcriptional programs.
- Published
- 2008
- Full Text
- View/download PDF
38. Endocytic downregulation of ErbB receptors: mechanisms and relevance in cancer.
- Author
-
Roepstorff K, Grøvdal L, Grandal M, Lerdrup M, and van Deurs B
- Subjects
- Animals, Down-Regulation, Humans, Models, Biological, Neoplasms metabolism, Neoplasms pathology, Signal Transduction, Ubiquitin metabolism, Endocytosis physiology, ErbB Receptors metabolism
- Abstract
ErbB receptors (EGFR (ErbB1), ErbB2, ErbB3, and ErbB4) are important regulators of normal growth and differentiation, and they are involved in the pathogenesis of cancer. Following ligand binding and receptor activation, EGFR is endocytosed and transported to lysosomes where the receptor is degraded. This downregulation of EGFR is a complex and tightly regulated process. The functions of ErbB2, ErbB3, and ErbB4 are also regulated by endocytosis to some extent, although the current knowledge of these processes is sparse. Impaired endocytic downregulation of signaling receptors is frequently associated with cancer, since it can lead to increased and uncontrolled receptor signaling. In this review we describe the current knowledge of ErbB receptor endocytic downregulation. In addition, we outline how ErbB receptors can escape endocytic downregulation in cancer, and we discuss how targeted anti-cancer therapy may induce endocytic downregulation of ErbB receptors.
- Published
- 2008
- Full Text
- View/download PDF
39. Endocytic down-regulation of ErbB2 is stimulated by cleavage of its C-terminus.
- Author
-
Lerdrup M, Bruun S, Grandal MV, Roepstorff K, Kristensen MM, Hommelgaard AM, and van Deurs B
- Subjects
- Benzoquinones pharmacology, Cell Membrane drug effects, Cell Membrane metabolism, Down-Regulation drug effects, Humans, Lactams, Macrocyclic pharmacology, Lysosomes drug effects, Lysosomes metabolism, Mutant Proteins metabolism, Proteasome Endopeptidase Complex metabolism, Receptor, ErbB-2 metabolism, Recombinant Fusion Proteins metabolism, Reproducibility of Results, Structure-Activity Relationship, Down-Regulation genetics, Endocytosis drug effects, Receptor, ErbB-2 chemistry, Receptor, ErbB-2 genetics
- Abstract
High ErbB2 levels are associated with cancer, and impaired endocytosis of ErbB2 could contribute to its overexpression. Therefore, knowledge about the mechanisms underlying endocytic down-regulation of ErbB2 is warranted. The C-terminus of ErbB2 can be cleaved after various stimuli, and after inhibition of HSP90 with geldanamycin this cleavage is accompanied by proteasome-dependent endocytosis of ErbB2. However, it is unknown whether C-terminal cleavage is linked to endocytosis. To study ErbB2 cleavage and endocytic trafficking, we fused yellow fluorescent protein (YFP) and cyan fluorescent protein (CFP) to the N- and C-terminus of ErbB2, respectively (YFP-ErbB2-CFP). After geldanamycin stimulation YFP-ErbB2-CFP became cleaved in nonapoptotic cells in a proteasome-dependent manner, and a markedly larger relative amount of cleaved YFP-ErbB2-CFP was observed in early endosomes than in the plasma membrane. Furthermore, cleavage took place at the plasma membrane, and cleaved ErbB2 was internalized and degraded far more efficiently than full-length ErbB2. Concordantly, a C-terminally truncated ErbB2 was also readily endocytosed and degraded in lysosomes compared with full-length ErbB2. Altogether, we suggest that geldanamycin leads to C-terminal cleavage of ErbB2, which releases the receptor from a retention mechanism and causes endocytosis and lysosomal degradation of ErbB2.
- Published
- 2007
- Full Text
- View/download PDF
40. Geldanamycin stimulates internalization of ErbB2 in a proteasome-dependent way.
- Author
-
Lerdrup M, Hommelgaard AM, Grandal M, and van Deurs B
- Subjects
- Acetylcysteine analogs & derivatives, Acetylcysteine metabolism, Animals, Biological Assay methods, Cell Line, Tumor, Cell Membrane metabolism, Cell Membrane ultrastructure, Fluorescence Recovery After Photobleaching, Humans, Lysosomes metabolism, Microscopy, Confocal methods, Protein Structure, Tertiary, Benzoquinones metabolism, Cysteine Proteinase Inhibitors metabolism, Endocytosis physiology, HSP90 Heat-Shock Proteins antagonists & inhibitors, Lactams, Macrocyclic metabolism, Proteasome Endopeptidase Complex metabolism, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 metabolism
- Abstract
The potent oncoprotein and receptor tyrosine kinase ErbB2 is remarkable because it resists efficient downregulation. However, ErbB2 can be downregulated by the HSP-90 inhibitor geldanamycin, but the underlying cellular mechanisms are uncertain. Apparently, delivery of ErbB2 to lysosomes, cleavage of the ErbB2 kinase domain and proteasomal activity are all processes that are involved. Using a non-invasive confocal microscopical assay allowing quantitative analysis of ErbB2 internalization in cell populations, we show that whereas ErbB2 is resistant to internalization in untreated SK-BR-3 cells, geldanamycin stimulates internalization and subsequent degradation in lysosomes. This process depends on proteasomal activity, which is a regulatory upstream event in ErbB2 internalization rather than the actual mechanism of degradation. ErbB2 can be internalized as a full-length protein, thus cleavage of the ErbB2 kinase domain is not a requirement for geldanamycin-stimulated internalization. Moreover, as shown by FRAP (fluorescence recovery after photobleaching) and electron microscopy, geldanamycin induces an increase in the amount of mobile ErbB2 and a redistribution of ErbB2 in the plasma membrane making the receptor accessible to endocytosis. Cells with most ErbB2 endocytosis also have the highest fraction of mobile ErbB2. It is concluded that geldanamycin stimulates internalization of full-length ErbB2 in a proteasome-dependent manner leading to lysosomal degradation.
- Published
- 2006
- Full Text
- View/download PDF
41. Association with membrane protrusions makes ErbB2 an internalization-resistant receptor.
- Author
-
Hommelgaard AM, Lerdrup M, and van Deurs B
- Subjects
- Actins metabolism, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal, Humanized, Biotin pharmacology, Blotting, Western, Bridged Bicyclo Compounds, Heterocyclic metabolism, Cell Line, Tumor, Centrifugation, Density Gradient, Cholesterol metabolism, Clathrin chemistry, Clathrin metabolism, Cross-Linking Reagents pharmacology, Cytoskeleton metabolism, Detergents pharmacology, Down-Regulation, Endosomes metabolism, Epidermal Growth Factor metabolism, Humans, Immunohistochemistry, Ligands, Membrane Microdomains, Microscopy, Confocal, Microscopy, Electron, Microscopy, Fluorescence, Models, Biological, Neuregulin-1 metabolism, Protein Binding, Protein Structure, Tertiary, Receptors, Transferrin metabolism, Signal Transduction, Sucrose pharmacology, Thiazoles metabolism, Thiazolidines, Trastuzumab, Cell Membrane metabolism, Receptor, ErbB-2 physiology
- Abstract
In contrast to the epidermal growth factor (EGF) receptor, ErbB2 is known to remain at the plasma membrane after ligand binding and dimerization. However, why ErbB2 is not efficiently down-regulated has remained elusive. Basically, two possibilities exist: ErbB2 is internalization resistant or it is efficiently recycled after internalization. By a combination of confocal microscopy, immunogold labeling electron microscopy, and biochemical techniques we show that ErbB2 is preferentially associated with membrane protrusions. Moreover, it is efficiently excluded from clathrin-coated pits and is not seen in transferrin receptor-containing endosomes. This pattern is not changed after binding of EGF, heregulin, or herceptin. The exclusion from coated pits is so pronounced that it cannot just be explained by lack of an internalization signal. Although ErbB2 is a raft-associated protein, the localization of ErbB2 to protrusions is not a result of raft binding. Also, an intact actin cytoskeleton is not required for keeping ErbB2 away from coated pits. However, after efficient cross-linking, ErbB2 is removed from protrusions to occur on the bulk membrane, in coated pits, and in endosomes. These data show that ErbB2 is a remarkably internalization-resistant receptor and suggest that the mechanism underlying the firm association of ErbB2 with protrusions also is the reason for this resistance.
- Published
- 2004
- Full Text
- View/download PDF
42. A novel specific role for I kappa B kinase complex-associated protein in cytosolic stress signaling.
- Author
-
Holmberg C, Katz S, Lerdrup M, Herdegen T, Jäättelä M, Aronheim A, and Kallunki T
- Subjects
- Base Sequence, Cloning, Molecular, DNA Primers, DNA, Complementary, Humans, I-kappa B Kinase, JNK Mitogen-Activated Protein Kinases, Mitogen-Activated Protein Kinases metabolism, Mutagenesis, Polymerase Chain Reaction methods, Protein Serine-Threonine Kinases genetics, Recombinant Proteins metabolism, Restriction Mapping, Saccharomyces cerevisiae genetics, Sequence Deletion, Stress, Physiological, Cytosol physiology, Protein Serine-Threonine Kinases metabolism, Signal Transduction physiology
- Abstract
We demonstrate here a novel role for the I kappa B kinase complex-associated protein (IKAP) in the regulation of activation of the mammalian stress response via the c-Jun N-terminal kinase (JNK)-signaling pathway. We cloned IKAP as a JNK-associating protein using the Ras recruitment yeast two-hybrid system. IKAP efficiently and specifically enhanced JNK activation induced by ectopic expression of MEKK1 and ASK1, upstream activators of JNK. Importantly, IKAP also enhanced JNK activation induced by ultraviolet light irradiation as well as treatments with tumor necrosis factor or epidermal growth factor. The JNK association site in IKAP was mapped to the C-terminal part of IKAP. Interestingly, this region is deleted from IKAP expressed in the autonomous nervous system of the patients affected by familial dysautonomia. Ectopic expression of this C-terminal fragment of IKAP was sufficient to support JNK activation. Taken together, our data demonstrate a novel role for IKAP in the regulation of the JNK-mediated stress signaling. Additionally, our results point to a role of JNK signaling in familial dysautonomia and, thus, further support the involvement of JNK signaling in the development, survival, and degeneration of the sensory and autonomic nervous system.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.