121 results on '"Schotta G"'
Search Results
2. Resistance to mesenchymal reprogramming sustains clonal propagation in metastatic breast cancer
- Author
-
Saini, M., Schmidleitner, L., Moreno, H.D., Donato, E., Falcone, M., Bartsch, J.M., Vogel, V., Würth, R., Pfarr, N., Espinet, E., Lehmann, M., Königshoff, M., Reitberger, M., Haas, S., Graf, E., Schwarzmayr, T., Strom, T.M., Spaich, S., Sütterlin, M., Schneeweiss, A., Weichert, W., Schotta, G., Reichert, M., Aceto, N., Sprick, M.R., Trumpp, A., Scheel, C.H., and Klein, C.
- Subjects
Cancer Research - Abstract
The acquisition of mesenchymal traits is considered a hallmark of breast cancer progression. However, the functional relevance of epithelial-to-mesenchymal transition (EMT) remains controversial and context dependent. Here, we isolate epithelial and mesenchymal populations from human breast cancer metastatic biopsies and assess their functional potential in vivo. Strikingly, progressively decreasing epithelial cell adhesion molecule (EPCAM) levels correlate with declining disease propagation. Mechanistically, we find that persistent EPCAM expression marks epithelial clones that resist EMT induction and propagate competitively. In contrast, loss of EPCAM defines clones arrested in a mesenchymal state, with concomitant suppression of tumorigenicity and metastatic potential. This dichotomy results from distinct clonal trajectories impacting global epigenetic programs that are determined by the interplay between human ZEB1 and its target GRHL2. Collectively, our results indicate that susceptibility to irreversible EMT restrains clonal propagation, whereas resistance to mesenchymal reprogramming sustains disease spread in multiple models of human metastatic breast cancer, including patient-derived cells in vivo.
- Published
- 2023
3. The Role of Histone Modifications in Epigenetic Transitions During Normal and Perturbed Development
- Author
-
Kubicek, S., Schotta, G., Lachner, M., Sengupta, R., Kohlmaier, A., Perez-Burgos, L., Linderson, Y., Martens, J. H. A., O’Sullivan, R. J., Fodor, B. D., Yonezawa, M., Peters, A. H. F. M., Jenuwein, T., Stock, G., editor, Lessl, M., editor, Berger, S. L., editor, Nakanishi, O., editor, and Haendler, B., editor
- Published
- 2006
- Full Text
- View/download PDF
4. Environmental signals rather than layered ontogeny imprint the function of type 2 conventional dendritic cells in young and adult mice
- Author
-
Papaioannou, N.E., Salei, N., Rambichler, S., Ravi, K., Popovic, J., Küntzel, V., Lehmann, C.H.K., Fiancette, R., Salvermoser, J., Gajdasik, D.W., Mettler, R., Messerer, D., Carrelha, J., Ohnmacht, C., Haller, D., Stumm, R., Straub, T., Jacobsen, S.E.W., Schulz, C., Withers, D.R., Schotta, G., Dudziak, D., and Schraml, B.U.
- Subjects
Male ,T-Lymphocytes ,Science ,Primary Cell Culture ,Antigen-presenting cells ,Mice, Transgenic ,Cell Separation ,Adaptive Immunity ,Article ,Mice ,Animals ,RNA-Seq ,ddc:610 ,Myelopoiesis ,Age Factors ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Dendritic Cells ,Flow Cytometry ,Hematopoietic Stem Cells ,Gene regulation in immune cells ,Models, Animal ,Cytokines ,Female ,Single-Cell Analysis ,Transcriptome - Abstract
Conventional dendritic cells (cDC) are key activators of naive T cells, and can be targeted in adults to induce adaptive immunity, but in early life are considered under-developed or functionally immature. Here we show that, in early life, when the immune system develops, cDC2 exhibit a dual hematopoietic origin and, like other myeloid and lymphoid cells, develop in waves. Developmentally distinct cDC2 in early life, despite being distinguishable by fate mapping, are transcriptionally and functionally similar. cDC2 in early and adult life, however, are exposed to distinct cytokine environments that shape their transcriptional profile and alter their ability to sense pathogens, secrete cytokines and polarize T cells. We further show that cDC2 in early life, despite being distinct from cDC2 in adult life, are functionally competent and can induce T cell responses. Our results thus highlight the potential of harnessing cDC2 for boosting immunity in early life., Type 2 conventional dendritic cells (cDC2) are important immune activators in adults, but their development and functions at the neonatal stage remain unclear. Here the authors show, using fate-mapping and single-cell RNA sequencing, that neonatal cDC2 come from multiple origins, but converge functionally as potent immune activators upon proper stimuli.
- Published
- 2021
5. Epigenetic regulation of development by histone lysine methylation
- Author
-
Dambacher, S, Hahn, M, and Schotta, G
- Published
- 2010
- Full Text
- View/download PDF
6. Controlled expression of tagged proteins in Drosophila using a new modular P-element vector system
- Author
-
Schotta, G. and Reuter, G.
- Published
- 2000
- Full Text
- View/download PDF
7. Der Verlust von intestinal epithelialem SETDB1 führt zu fehlender Repression endogener Retroviren, Genotoxizität und intestinaler Entzündung
- Author
-
Južnić, L, additional, Peuker, K, additional, Strigli, A, additional, Brosch, M, additional, Herrmann, A, additional, Häsler, R, additional, Koch, M, additional, Matthiesen, L, additional, Zeissig, Y, additional, Löscher, B.-S, additional, Nuber, A, additional, Schotta, G, additional, Neumeister, V, additional, Chavakis, T, additional, Kurth, T, additional, Lesche, M, additional, Dahl, A, additional, von Mässenhausen, A, additional, Linkermann, A, additional, Schreiber, S, additional, Aden, K, additional, Rosenstiel, P, additional, Franke, A, additional, Hampe, J, additional, and Zeissig, S, additional
- Published
- 2020
- Full Text
- View/download PDF
8. Epigenetics of eu- and heterochromatin in inverted and conventional nuclei from mouse retina
- Author
-
Eberhart, A., Feodorova, Y., Song, C., Wanner, G., Kiseleva, E., Furukawa, T., Kimura, Hiroshi, Schotta, G., Leonhardt, H., Joffe, B., and Solovei, I.
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Epigenomics ,X Chromosome ,Euchromatin ,Chromosomal Proteins, Non-Histone ,Heterochromatin ,Epigenetic code ,Retinal Rod Photoreceptor Cells/metabolism/ultrastructure ,Histones/metabolism ,Chromosomal Proteins, Non-Histone/metabolism ,Biology ,Retina ,X-inactivation ,Epigenesis, Genetic ,Histones ,Mice ,Retinal Rod Photoreceptor Cells ,X Chromosome Inactivation ,Cell Nucleus/*genetics/metabolism/ultrastructure ,Genetics ,Animals ,Histone code ,DNA (Cytosine-5-)-Methyltransferases ,Euchromatin/*genetics/metabolism/ultrastructure ,Cell Nucleus ,Mice, Knockout ,Heterochromatin/*genetics/metabolism/ultrastructure ,DNA Methylation ,Molecular biology ,Cell biology ,Chromatin ,Retina/*metabolism ,Histone ,Sex Chromatin ,DNA (Cytosine-5-)-Methyltransferase/metabolism ,biology.protein ,Heterochromatin protein 1 - Abstract
To improve light propagation through the retina, the rod nuclei of nocturnal mammals are uniquely changed compared to the nuclei of other cells. In particular, the main classes of chromatin are segregated in them and form regular concentric shells in order; inverted in comparison to conventional nuclei. A broad study of the epigenetic landscape of the inverted and conventional mouse retinal nuclei indicated several differences between them and several features of general interest for the organization of the mammalian nuclei. In difference to nuclei with conventional architecture, the packing density of pericentromeric satellites and LINE-rich chromatin is similar in inverted rod nuclei; euchromatin has a lower packing density in both cases. A high global chromatin condensation in rod nuclei minimizes the structural difference between active and inactive X chromosome homologues. DNA methylation is observed primarily in the chromocenter, Dnmt1 is primarily associated with the euchromatic shell. Heterochromatin proteins HP1-alpha and HP1-beta localize in heterochromatic shells, whereas HP1-gamma is associated with euchromatin. For most of the 25 studied histone modifications, we observed predominant colocalization with a certain main chromatin class. Both inversions in rod nuclei and maintenance of peripheral heterochromatin in conventional nuclei are not affected by a loss or depletion of the major silencing core histone modifications in respective knock-out mice, but for different reasons. Maintenance of peripheral heterochromatin appears to be ensured by redundancy both at the level of enzymes setting the epigenetic code (writers) and the code itself, whereas inversion in rods rely on the absence of the peripheral heterochromatin tethers (absence of code readers).
- Published
- 2013
- Full Text
- View/download PDF
9. Regulatory RNA triggers formation of non-canonical heterochromatin in growth-arrested cells
- Author
-
Bierhoff, H, Dammert, M, Brocks, D, Dambacher, S, Schotta, G, and Grummt, I.
- Published
- 2014
10. -globin expression is regulated by SUV4-20h1
- Author
-
Wang, Y., primary, Rank, G., additional, Li, Z., additional, Wang, Y., additional, Ju, J., additional, Nuber, A., additional, Wu, Y., additional, Liu, M., additional, Nie, M., additional, Huang, F., additional, Cerruti, L., additional, Ma, C., additional, Tan, R., additional, Schotta, G., additional, Jane, S. M., additional, Zeng, C. K., additional, and Zhao, Q., additional
- Published
- 2016
- Full Text
- View/download PDF
11. A chromatin-wide transition to H4K20 monomethylation impairs genome integrity and programmed DNA rearrangements in the mouse
- Author
-
Schotta, G., Sengupta, R., Kubicek, S., Malin, S., Kauer, M., Callén, E., Celeste, A., Pagani, M., Opravil, S., De La Rosa-Velazquez, I., Espejo, A., Bedford, M., Nussenzweig, A., Busslinger, M., and Jenuwein, T.
- Abstract
H4K20 methylation is a broad chromatin modification that has been linked with diverse epigenetic functions. Several enzymes target H4K20 methylation, consistent with distinct mono-, di-, and trimethylation states controlling different biological outputs. To analyze the roles of H4K20 methylation states, we generated conditional null alleles for the two Suv4-20h histone methyltransferase (HMTase) genes in the mouse. Suv4-20h-double-null (dn) mice are perinatally lethal and have lost nearly all H4K20me3 and H4K20me2 states. The genome-wide transition to an H4K20me1 state results in increased sensitivity to damaging stress, since Suv4-20h-dn chromatin is less efficient for DNA double-strand break (DSB) repair and prone to chromosomal aberrations. Notably, Suv4-20h-dn B cells are defective in immunoglobulin class-switch recombination, and Suv4-20h-dn deficiency impairs the stem cell pool of lymphoid progenitors. Thus, conversion to an H4K20me1 state results in compromised chromatin that is insufficient to protect genome integrity and to process a DNA-rearranging differentiation program in the mouse.
- Published
- 2008
12. The role of histone modifications in epigenetic transitions during normal and perturbed development
- Author
-
Kubicek S, Schotta G, Lachner M, Sengupta R, Kohlmaier A, Perez-Burgos L, Linderson Y, Jh, Martens, Sullivan Rj, O., Barna Fodor, Yonezawa M, Ah, Peters, and Jenuwein T
- Subjects
Histones ,Cell Transformation, Neoplastic ,Animals ,Embryonic Development ,Humans ,Cell Differentiation ,Cell Lineage ,Methylation ,Chromatin ,Epigenesis, Genetic ,Protein Structure, Tertiary - Abstract
Epigenetic mechanisms control eukaryotic development beyond DNA-stored information. DNA methylation, histone modifications and variants, nucleosome remodeling and noncoding RNAs all contribute to the dynamic make-up of chromatin under distinct developmental options. In particular, the great diversity of covalent histone tail modifications has been proposed to be ideally suited for imparting epigenetic information. While most of the histone tail modifications represent transient marks at transcriptionally permissive chromatin, some modifications appear more robust at silent chromatin regions, where they index repressive epigenetic states with functions also outside transcriptional regulation. Under-representation of repressive histone marks could be indicative of epigenetic plasticity in stem, young and tumor cells, while committed and senescent (old) cells often display increased levels of these more stable modifications. Here, we discuss profiles of normal and aberrant histone lysine methylation patterns, as they occur during the transition of an embryonic to a differentiated cell or in controlled self-renewal vs pro-neoplastic or metastatic conditions. Elucidating these histone modification patterns promises to have important implications for novel advances in stem cell research, nuclear reprogramming and cancer, and may offer novel targets for the combat of tumor cells, potentially leading to new diagnostic and therapeutic avenues in human biology and disease.
- Published
- 2006
13. The Role of Histone Modifications in Epigenetic Transitions During Normal and Perturbed Development
- Author
-
Kubicek, S., primary, Schotta, G., additional, Lachner, M., additional, Sengupta, R., additional, Kohlmaier, A., additional, Perez-Burgos, L., additional, Linderson, Y., additional, Martens, J. H. A., additional, O’Sullivan, R. J., additional, Fodor, B. D., additional, Yonezawa, M., additional, Peters, A. H. F. M., additional, and Jenuwein, T., additional
- Full Text
- View/download PDF
14. Histone H4 Lysine 20 methylation: key player in epigenetic regulation of genomic integrity
- Author
-
Jorgensen, S., primary, Schotta, G., additional, and Sorensen, C. S., additional
- Published
- 2013
- Full Text
- View/download PDF
15. Active and repressive chromatin are interspersed without spreading in an imprinted gene cluster in the mammalian genome
- Author
-
Regha, K., Sloane, M. A., Huang, R., Pauler, F. M., Warczok, K. E., Melikant, B., Radolf, M., Martens, J.H.A., Schotta, G., Jenuwein, T., Barlow, D. P., Regha, K., Sloane, M. A., Huang, R., Pauler, F. M., Warczok, K. E., Melikant, B., Radolf, M., Martens, J.H.A., Schotta, G., Jenuwein, T., and Barlow, D. P.
- Abstract
Item does not contain fulltext
- Published
- 2007
16. H4K20 monomethylation faces the WNT
- Author
-
Schotta, G., primary
- Published
- 2011
- Full Text
- View/download PDF
17. Trilogies of Histone Lysine Methylation as Epigenetic Landmarks of the Eukaryotic Genome
- Author
-
LACHNER, M., primary, SENGUPTA, R., additional, SCHOTTA, G., additional, and JENUWEIN, T., additional
- Published
- 2004
- Full Text
- View/download PDF
18. The Role of Histone Modifications in Epigenetic Transitions During Normal and Perturbed Development.
- Author
-
Stock, G., Lessl, M., Berger, S. L., Nakanishi, O., Haendler, B., Kubicek, S., Schotta, G., Lachner, M., Sengupta, R., Kohlmaier, A., Perez-Burgos, L., Linderson, Y., Martens, J. H. A., O'Sullivan, R. J., Fodor, B. D., Yonezawa, M., Peters, A. H. F. M., and Jenuwein, T.
- Abstract
Epigenetic mechanisms control eukaryotic development beyond DNA-stored information. DNA methylation, histone modifications and variants, nucleosome remodeling and noncoding RNAs all contribute to the dynamic make-up of chromatin under distinct developmental options. In particular, the great diversity of covalent histone tail modifications has been proposed to be ideally suited for imparting epigenetic information. While most of the histone tail modifications represent transient marks at transcriptionally permissive chromatin, some modifications appear more robust at silent chromatin regions, where they index repressive epigenetic states with functions also outside transcriptional regulation. Under-representation of repressive histone marks could be indicative of epigenetic plasticity in stem, young and tumor cells, while committed and senescent (old) cells often display increased levels of these more stable modifications. Here, we discuss profiles of normal and aberrant histone lysine methylation patterns, as they occur during the transition of an embryonic to a differentiated cell or in controlled self-renewal vs pro-neoplastic or metastatic conditions. Elucidating these histone modification patterns promises to have important implications for novel advances in stem cell research, nuclear reprogramming and cancer, and may offer novel targets for the combat of tumor cells, potentially leading to new diagnostic and therapeutic avenues in human biology and disease. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
19. Central role of Drosophila SU(VAR)3-9 in histone H3-K9 methylation and heterochromatic gene silencing
- Author
-
Schotta, G., primary
- Published
- 2002
- Full Text
- View/download PDF
20. FSHD muscular dystrophy region gene 1 binds Suv4-20h1 histone methyltransferase and impairs myogenesis
- Author
-
Cristina Godio, Alexandros Xynos, Gunnar Schotta, Maria Victoria Neguembor, Mariaelena Pistoni, Davide Gabellini, Roberta Caccia, Davide Corona, Sergia Bortolanza, Maria Cristina Onorati, Neguembor MV, Xynos A, Onorati MC, Caccia R, Bortolanza S, Godio C, Pistoni M, Corona D, Schotta G, and Gabellini D
- Subjects
Muscle Development ,Evolution, Molecular ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genetics ,medicine ,Facioscapulohumeral muscular dystrophy ,Myocyte ,Animals ,Humans ,Epigenetics ,Muscular dystrophy ,Myopathy ,Molecular Biology ,030304 developmental biology ,Cell Nucleus ,Mice, Knockout ,0303 health sciences ,Muscle Cells ,biology ,Myogenesis ,Microfilament Proteins ,Nuclear Proteins ,Proteins ,RNA-Binding Proteins ,Cell Differentiation ,Cell Biology ,General Medicine ,Histone-Lysine N-Methyltransferase ,Muscular Dystrophy, Animal ,medicine.disease ,Molecular biology ,Histone ,Drosophila melanogaster ,HEK293 Cells ,Phenotype ,Organ Specificity ,Histone methyltransferase ,Epigenetic deregulation by FRG1 ,Gene Knockdown Techniques ,biology.protein ,medicine.symptom ,Carrier Proteins ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant myopathy with a strong epigenetic component. It is associated with deletion of a macrosatellite repeat leading to over-expression of the nearby genes. Among them, we focused on FSHD region gene 1 (FRG1) since its over-expression in mice, Xenopus laevis and Caenorhabditis elegans, leads to muscular dystrophy-like defects, suggesting that FRG1 plays a relevant role in muscle biology. Here we show that, when over-expressed, FRG1 binds and interferes with the activity of the histone methyltransferase Suv4-20h1 both in mammals and Drosophila. Accordingly, FRG1 over-expression or Suv4-20h1 knockdown inhibits myogenesis. Moreover, Suv4-20h KO mice develop muscular dystrophy signs. Finally, we identify the FRG1/Suv4-20h1 target Eid3 as a novel myogenic inhibitor that contributes to the muscle differentiation defects. Our study suggests a novel role of FRG1 as epigenetic regulator of muscle differentiation and indicates that Suv4-20h1 has a gene-specific function in myogenesis.
- Published
- 2013
21. G-quadruplexes in an SVA retrotransposon cause aberrant TAF1 gene expression in X-linked dystonia parkinsonism.
- Author
-
Nicoletto G, Terreri M, Maurizio I, Ruggiero E, Cernilogar FM, Vaine CA, Cottini MV, Shcherbakova I, Penney EB, Gallina I, Monchaud D, Bragg DC, Schotta G, and Richter SN
- Subjects
- Humans, Histone Acetyltransferases genetics, Histone Acetyltransferases metabolism, Retroelements genetics, Fibroblasts metabolism, Short Interspersed Nucleotide Elements genetics, Neural Stem Cells metabolism, Gene Expression Regulation, Minisatellite Repeats genetics, TATA-Binding Protein Associated Factors genetics, TATA-Binding Protein Associated Factors metabolism, G-Quadruplexes, Transcription Factor TFIID genetics, Transcription Factor TFIID metabolism, Dystonic Disorders genetics, Genetic Diseases, X-Linked genetics, Genetic Diseases, X-Linked metabolism
- Abstract
G-quadruplexes (G4s) are non-canonical nucleic acid structures that form in guanine (G)-rich genomic regions. X-linked dystonia parkinsonism (XDP) is an inherited neurodegenerative disease in which a SINE-VNTR-Alu (SVA) retrotransposon, characterised by amplification of a G-rich repeat, is inserted into the coding sequence of TAF1, a key partner of RNA polymerase II. XDP SVA alters TAF1 expression, but the cause of this outcome in XDP remains unknown. To assess whether G4s form in XDP SVA and affect TAF1 expression, we first characterised bioinformatically predicted XDP SVA G4s in vitro. We next showed that highly stable G4s can form and stop polymerase amplification at the SVA region from patient-derived fibroblasts and neural progenitor cells. Using chromatin immunoprecipitazion (ChIP) with an anti-G4 antibody coupled to sequencing or quantitative PCR, we showed that XDP SVA G4s are folded even when embedded in a chromatin context in patient-derived cells. Using the G4 ligands BRACO-19 and quarfloxin and total RNA-sequencing analysis, we showed that stabilisation of the XDP SVA G4s reduces TAF1 transcripts downstream and around the SVA, and increases upstream transcripts, while destabilisation using the G4 unfolder PhpC increases TAF1 transcripts. Our data indicate that G4 formation in the XDP SVA is a major cause of aberrant TAF1 expression, opening the way for the development of strategies to unfold G4s and potentially target the disease., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
22. Evolution of T-cell fitness through AML progression: enhanced bispecific T-cell engager-mediated function of bone marrow T cells at remission compared to initial diagnosis and relapse.
- Author
-
Kazerani M, Marcinek A, Philipp N, Brauchle B, Taylor JJ, Moreno HD, Terrasi A, Tast B, Rohrbacher L, Wang Y, Warm M, Emhardt AJ, Magno G, Spiekermann K, Herold T, Straub T, Theurich S, Schotta G, Kischel R, Bücklein VL, and Subklewe M
- Subjects
- Humans, Remission Induction, Antibodies, Bispecific therapeutic use, Neoplasm Recurrence, Local pathology, Male, Female, Recurrence, Bone Marrow Cells pathology, Bone Marrow Cells immunology, Middle Aged, T-Lymphocytes immunology, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute therapy, Disease Progression
- Published
- 2024
- Full Text
- View/download PDF
23. β1 integrin signaling governs necroptosis via the chromatin-remodeling factor CHD4.
- Author
-
Sun Z, Cernilogar FM, Horvatic H, Yeroslaviz A, Abdullah Z, Schotta G, and Hornung V
- Subjects
- Humans, Mi-2 Nucleosome Remodeling and Deacetylase Complex genetics, Transcription Factors genetics, Nucleosomes, Fibrosis, Inflammation, Integrin beta1 genetics, Necroptosis
- Abstract
Fibrosis, characterized by sustained activation of myofibroblasts and excessive extracellular matrix (ECM) deposition, is known to be associated with chronic inflammation. Receptor-interacting protein kinase 3 (RIPK3), the central kinase of necroptosis signaling, is upregulated in fibrosis and contributes to tumor necrosis factor (TNF)-mediated inflammation. In bile-duct-ligation-induced liver fibrosis, we found that myofibroblasts are the major cell type expressing RIPK3. Genetic ablation of β1 integrin, the major profibrotic ECM receptor in fibroblasts, not only abolished ECM fibrillogenesis but also blunted RIPK3 expression via a mechanism mediated by the chromatin-remodeling factor chromodomain helicase DNA-binding protein 4 (CHD4). While the function of CHD4 has been conventionally linked to the nucleosome-remodeling deacetylase (NuRD) and CHD4-ADNP-HP1(ChAHP) complexes, we found that CHD4 potently repressed a set of genes, including Ripk3, with high locus specificity but independent of either the NuRD or the ChAHP complex. Thus, our data uncover that β1 integrin intrinsically links fibrotic signaling to RIPK3-driven inflammation via a novel mode of action of CHD4., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. PD-1 instructs a tumor-suppressive metabolic program that restricts glycolysis and restrains AP-1 activity in T cell lymphoma.
- Author
-
Wartewig T, Daniels J, Schulz M, Hameister E, Joshi A, Park J, Morrish E, Venkatasubramani AV, Cernilogar FM, van Heijster FHA, Hundshammer C, Schneider H, Konstantinidis F, Gabler JV, Klement C, Kurniawan H, Law C, Lee Y, Choi S, Guitart J, Forne I, Giustinani J, Müschen M, Jain S, Weinstock DM, Rad R, Ortonne N, Schilling F, Schotta G, Imhof A, Brenner D, Choi J, and Ruland J
- Subjects
- Mice, Animals, Humans, Transcription Factor AP-1 genetics, Transcription Factor AP-1 metabolism, Programmed Cell Death 1 Receptor genetics, Programmed Cell Death 1 Receptor metabolism, Genes, Tumor Suppressor, Acetyl Coenzyme A metabolism, Glycolysis genetics, Lymphoma, T-Cell genetics, Lymphoma, T-Cell, Peripheral
- Abstract
The PDCD1-encoded immune checkpoint receptor PD-1 is a key tumor suppressor in T cells that is recurrently inactivated in T cell non-Hodgkin lymphomas (T-NHLs). The highest frequencies of PDCD1 deletions are detected in advanced disease, predicting inferior prognosis. However, the tumor-suppressive mechanisms of PD-1 signaling remain unknown. Here, using tractable mouse models for T-NHL and primary patient samples, we demonstrate that PD-1 signaling suppresses T cell malignancy by restricting glycolytic energy and acetyl coenzyme A (CoA) production. In addition, PD-1 inactivation enforces ATP citrate lyase (ACLY) activity, which generates extramitochondrial acetyl-CoA for histone acetylation to enable hyperactivity of activating protein 1 (AP-1) transcription factors. Conversely, pharmacological ACLY inhibition impedes aberrant AP-1 signaling in PD-1-deficient T-NHLs and is toxic to these cancers. Our data uncover genotype-specific vulnerabilities in PDCD1-mutated T-NHL and identify PD-1 as regulator of AP-1 activity., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
25. Resistance to mesenchymal reprogramming sustains clonal propagation in metastatic breast cancer.
- Author
-
Saini M, Schmidleitner L, Moreno HD, Donato E, Falcone M, Bartsch JM, Klein C, Vogel V, Würth R, Pfarr N, Espinet E, Lehmann M, Königshoff M, Reitberger M, Haas S, Graf E, Schwarzmayr T, Strom TM, Spaich S, Sütterlin M, Schneeweiss A, Weichert W, Schotta G, Reichert M, Aceto N, Sprick MR, Trumpp A, and Scheel CH
- Subjects
- Humans, Female, Epithelial Cell Adhesion Molecule, Cell Line, Tumor, Breast metabolism, Clone Cells metabolism, Epithelial-Mesenchymal Transition, Breast Neoplasms pathology
- Abstract
The acquisition of mesenchymal traits is considered a hallmark of breast cancer progression. However, the functional relevance of epithelial-to-mesenchymal transition (EMT) remains controversial and context dependent. Here, we isolate epithelial and mesenchymal populations from human breast cancer metastatic biopsies and assess their functional potential in vivo. Strikingly, progressively decreasing epithelial cell adhesion molecule (EPCAM) levels correlate with declining disease propagation. Mechanistically, we find that persistent EPCAM expression marks epithelial clones that resist EMT induction and propagate competitively. In contrast, loss of EPCAM defines clones arrested in a mesenchymal state, with concomitant suppression of tumorigenicity and metastatic potential. This dichotomy results from distinct clonal trajectories impacting global epigenetic programs that are determined by the interplay between human ZEB1 and its target GRHL2. Collectively, our results indicate that susceptibility to irreversible EMT restrains clonal propagation, whereas resistance to mesenchymal reprogramming sustains disease spread in multiple models of human metastatic breast cancer, including patient-derived cells in vivo., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. WDR5 is required for DUX4 expression and its pathological effects in FSHD muscular dystrophy.
- Author
-
Mocciaro E, Giambruno R, Micheloni S, Cernilogar FM, Andolfo A, Consonni C, Pannese M, Ferri G, Runfola V, Schotta G, and Gabellini D
- Subjects
- Humans, Gene Expression Regulation, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Intracellular Signaling Peptides and Proteins genetics, Muscle Cells metabolism, Muscle, Skeletal metabolism, Transcription Factors metabolism, Muscular Dystrophy, Facioscapulohumeral metabolism
- Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is one of the most prevalent neuromuscular disorders. The disease is linked to copy number reduction and/or epigenetic alterations of the D4Z4 macrosatellite on chromosome 4q35 and associated with aberrant gain of expression of the transcription factor DUX4, which triggers a pro-apoptotic transcriptional program leading to muscle wasting. As today, no cure or therapeutic option is available to FSHD patients. Given its centrality in FSHD, blocking DUX4 expression with small molecule drugs is an attractive option. We previously showed that the long non protein-coding RNA DBE-T is required for aberrant DUX4 expression in FSHD. Using affinity purification followed by proteomics, here we identified the chromatin remodeling protein WDR5 as a novel DBE-T interactor and a key player required for the biological activity of the lncRNA. We found that WDR5 is required for the expression of DUX4 and its targets in primary FSHD muscle cells. Moreover, targeting WDR5 rescues both cell viability and myogenic differentiation of FSHD patient cells. Notably, comparable results were obtained by pharmacological inhibition of WDR5. Importantly, WDR5 targeting was safe to healthy donor muscle cells. Our results support a pivotal role of WDR5 in the activation of DUX4 expression identifying a druggable target for an innovative therapeutic approach for FSHD., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2023
- Full Text
- View/download PDF
27. Dominant role of DNA methylation over H3K9me3 for IAP silencing in endoderm.
- Author
-
Wang Z, Fan R, Russo A, Cernilogar FM, Nuber A, Schirge S, Shcherbakova I, Dzhilyanova I, Ugur E, Anton T, Richter L, Leonhardt H, Lickert H, and Schotta G
- Subjects
- Animals, Chromatin metabolism, DNA metabolism, Endoderm metabolism, Histone Methyltransferases metabolism, Histones genetics, Histones metabolism, Mice, DNA Methylation, Endogenous Retroviruses metabolism
- Abstract
Silencing of endogenous retroviruses (ERVs) is largely mediated by repressive chromatin modifications H3K9me3 and DNA methylation. On ERVs, these modifications are mainly deposited by the histone methyltransferase Setdb1 and by the maintenance DNA methyltransferase Dnmt1. Knock-out of either Setdb1 or Dnmt1 leads to ERV de-repression in various cell types. However, it is currently not known if H3K9me3 and DNA methylation depend on each other for ERV silencing. Here we show that conditional knock-out of Setdb1 in mouse embryonic endoderm results in ERV de-repression in visceral endoderm (VE) descendants and does not occur in definitive endoderm (DE). Deletion of Setdb1 in VE progenitors results in loss of H3K9me3 and reduced DNA methylation of Intracisternal A-particle (IAP) elements, consistent with up-regulation of this ERV family. In DE, loss of Setdb1 does not affect H3K9me3 nor DNA methylation, suggesting Setdb1-independent pathways for maintaining these modifications. Importantly, Dnmt1 knock-out results in IAP de-repression in both visceral and definitive endoderm cells, while H3K9me3 is unaltered. Thus, our data suggest a dominant role of DNA methylation over H3K9me3 for IAP silencing in endoderm cells. Our findings suggest that Setdb1-meditated H3K9me3 is not sufficient for IAP silencing, but rather critical for maintaining high DNA methylation., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
28. DNA sequence-dependent formation of heterochromatin nanodomains.
- Author
-
Thorn GJ, Clarkson CT, Rademacher A, Mamayusupova H, Schotta G, Rippe K, and Teif VB
- Subjects
- Animals, Base Sequence, Chromatin, Chromobox Protein Homolog 5, Chromosomal Proteins, Non-Histone metabolism, Histones genetics, Histones metabolism, Homeodomain Proteins genetics, Mice, Nerve Tissue Proteins metabolism, Heterochromatin genetics, Nucleosomes genetics
- Abstract
The mammalian epigenome contains thousands of heterochromatin nanodomains (HNDs) marked by di- and trimethylation of histone H3 at lysine 9 (H3K9me2/3), which have a typical size of 3-10 nucleosomes. However, what governs HND location and extension is only partly understood. Here, we address this issue by introducing the chromatin hierarchical lattice framework (ChromHL) that predicts chromatin state patterns with single-nucleotide resolution. ChromHL is applied to analyse four HND types in mouse embryonic stem cells that are defined by histone methylases SUV39H1/2 or GLP, transcription factor ADNP or chromatin remodeller ATRX. We find that HND patterns can be computed from PAX3/9, ADNP and LINE1 sequence motifs as nucleation sites and boundaries that are determined by DNA sequence (e.g. CTCF binding sites), cooperative interactions between nucleosomes as well as nucleosome-HP1 interactions. Thus, ChromHL rationalizes how patterns of H3K9me2/3 are established and changed via the activity of protein factors in processes like cell differentiation., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
29. Specific effects of somatic GATA2 zinc finger mutations on erythroid differentiation.
- Author
-
Redondo Monte E, Leubolt G, Windisch R, Kerbs P, Dutta S, Landspersky T, Istvánffy R, Oostendorp RAJ, Chen-Wichmann L, Herold T, Cusan M, Schotta G, Wichmann C, and Greif PA
- Subjects
- Animals, Cell Differentiation genetics, Humans, K562 Cells, Mice, Mutation, Zinc Fingers, GATA2 Transcription Factor genetics, Leukemia, Erythroblastic, Acute genetics, Leukemia, Myeloid
- Abstract
GATA2 zinc-finger (ZF) mutations are associated with distinct entities of myeloid malignancies. The specific distribution of these mutations points toward different mechanisms of leukemogenesis depending on the ZF domain affected. In this study, we compared recurring somatic mutations in ZF1 and ZF2. All tested ZF mutants disrupted DNA binding in vitro. In transcription assays, co-expression of FOG1 counteracted GATA2-dependent transcriptional activation, while a variable response to FOG1-mediated repression was observed for individual GATA2 mutants. In primary murine bone marrow cells, GATA2 wild-type (WT) expression inhibited colony formation, while this effect was reduced for both mutants A318T (ZF1) and L359V (ZF2) with a shift toward granulopoiesis. In primary human CD34
+ bone marrow cells and in the myeloid cell line K562, ectopic expression of GATA2 L359V, but not A318T or G320D, caused a block of erythroid differentiation accompanied by downregulation of GATA1, STAT5B, and PLCG1. Our findings may explain the role of GATA2 L359V during the progression of chronic myeloid leukemia and the collaboration of GATA2 ZF1 alterations with CEBPA double mutations in erythroleukemia., (Copyright © 2022 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
30. HDAC2 Facilitates Pancreatic Cancer Metastasis.
- Author
-
Krauß L, Urban BC, Hastreiter S, Schneider C, Wenzel P, Hassan Z, Wirth M, Lankes K, Terrasi A, Klement C, Cernilogar FM, Öllinger R, de Andrade Krätzig N, Engleitner T, Schmid RM, Steiger K, Rad R, Krämer OH, Reichert M, Schotta G, Saur D, and Schneider G
- Subjects
- Animals, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Cycle genetics, Cell Line, Tumor, Cell Proliferation genetics, Epithelial-Mesenchymal Transition genetics, Histone Deacetylase 2 metabolism, Humans, Kaplan-Meier Estimate, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Neoplasm Metastasis, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Signal Transduction genetics, Mice, Carcinoma, Pancreatic Ductal genetics, Gene Expression Profiling methods, Gene Expression Regulation, Neoplastic, Histone Deacetylase 2 genetics, Pancreatic Neoplasms genetics
- Abstract
The mortality of patients with pancreatic ductal adenocarcinoma (PDAC) is strongly associated with metastasis, a multistep process that is incompletely understood in this disease. Although genetic drivers of PDAC metastasis have not been defined, transcriptional and epigenetic rewiring can contribute to the metastatic process. The epigenetic eraser histone deacetylase 2 (HDAC2) has been connected to less differentiated PDAC, but the function of HDAC2 in PDAC has not been comprehensively evaluated. Using genetically defined models, we show that HDAC2 is a cellular fitness factor that controls cell cycle in vitro and metastasis in vivo, particularly in undifferentiated, mesenchymal PDAC cells. Unbiased expression profiling detected a core set of HDAC2-regulated genes. HDAC2 controlled expression of several prosurvival receptor tyrosine kinases connected to mesenchymal PDAC, including PDGFRα, PDGFRβ, and EGFR. The HDAC2-maintained program disabled the tumor-suppressive arm of the TGFβ pathway, explaining impaired metastasis formation of HDAC2-deficient PDAC. These data identify HDAC2 as a tractable player in the PDAC metastatic cascade. The complexity of the function of epigenetic regulators like HDAC2 implicates that an increased understanding of these proteins is needed for implementation of effective epigenetic therapies., Significance: HDAC2 has a context-specific role in undifferentiated PDAC and the capacity to disseminate systemically, implicating HDAC2 as targetable protein to prevent metastasis., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
31. Morc3 silences endogenous retroviruses by enabling Daxx-mediated histone H3.3 incorporation.
- Author
-
Groh S, Milton AV, Marinelli LK, Sickinger CV, Russo A, Bollig H, de Almeida GP, Schmidt A, Forné I, Imhof A, and Schotta G
- Subjects
- Adenosine Triphosphatases metabolism, Animals, Cell Line, Chromatin, Co-Repressor Proteins metabolism, DNA Methylation, DNA-Binding Proteins metabolism, Endogenous Retroviruses metabolism, Gene Knockout Techniques, Histone-Lysine N-Methyltransferase, Histones genetics, Histones metabolism, Humans, Molecular Chaperones metabolism, Protein Binding, Proteomics, Sumoylation, Adenosine Triphosphatases genetics, Co-Repressor Proteins genetics, DNA-Binding Proteins genetics, Endogenous Retroviruses genetics, Gene Silencing, Molecular Chaperones genetics
- Abstract
Endogenous retroviruses (ERVs) comprise a significant portion of mammalian genomes. Although specific ERV loci feature regulatory roles for host gene expression, most ERV integrations are transcriptionally repressed by Setdb1-mediated H3K9me3 and DNA methylation. However, the protein network which regulates the deposition of these chromatin modifications is still incompletely understood. Here, we perform a genome-wide single guide RNA (sgRNA) screen for genes involved in ERV silencing and identify the GHKL ATPase protein Morc3 as a top-scoring hit. Morc3 knock-out (ko) cells display de-repression, reduced H3K9me3, and increased chromatin accessibility of distinct ERV families. We find that the Morc3 ATPase cycle and Morc3 SUMOylation are important for ERV chromatin regulation. Proteomic analyses reveal that Morc3 mutant proteins fail to interact with the histone H3.3 chaperone Daxx. This interaction depends on Morc3 SUMOylation and Daxx SUMO binding. Notably, in Morc3 ko cells, we observe strongly reduced histone H3.3 on Morc3 binding sites. Thus, our data demonstrate Morc3 as a critical regulator of Daxx-mediated histone H3.3 incorporation to ERV regions., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
32. Publisher Correction: Epithelial cell plasticity drives endoderm formation during gastrulation.
- Author
-
Scheibner K, Schirge S, Burtscher I, Büttner M, Sterr M, Yang D, Böttcher A, Ansarullah, Irmler M, Beckers J, Cernilogar FM, Schotta G, Theis FJ, and Lickert H
- Published
- 2021
- Full Text
- View/download PDF
33. Epithelial cell plasticity drives endoderm formation during gastrulation.
- Author
-
Scheibner K, Schirge S, Burtscher I, Büttner M, Sterr M, Yang D, Böttcher A, Ansarullah, Irmler M, Beckers J, Cernilogar FM, Schotta G, Theis FJ, and Lickert H
- Subjects
- Animals, Blastocyst cytology, Blastocyst metabolism, Cell Differentiation, Cell Line, Endoderm cytology, Endoderm metabolism, Epithelial Cells metabolism, Gene Expression Regulation, Developmental, Gestational Age, Hepatocyte Nuclear Factor 3-beta genetics, Hepatocyte Nuclear Factor 3-beta metabolism, Mice, Mice, Transgenic, Mouse Embryonic Stem Cells metabolism, Phenotype, Snail Family Transcription Factors genetics, Snail Family Transcription Factors metabolism, Time Factors, Blastocyst physiology, Cell Plasticity, Endoderm physiology, Epithelial Cells physiology, Epithelial-Mesenchymal Transition, Gastrulation, Mouse Embryonic Stem Cells physiology
- Abstract
It is generally accepted that epiblast cells ingress into the primitive streak by epithelial-to-mesenchymal transition (EMT) to give rise to the mesoderm; however, it is less clear how the endoderm acquires an epithelial fate. Here, we used embryonic stem cell and mouse embryo knock-in reporter systems to combine time-resolved lineage labelling with high-resolution single-cell transcriptomics. This allowed us to resolve the morphogenetic programs that segregate the mesoderm from the endoderm germ layer. Strikingly, while the mesoderm is formed by classical EMT, the endoderm is formed independent of the key EMT transcription factor Snail1 by mechanisms of epithelial cell plasticity. Importantly, forkhead box transcription factor A2 (Foxa2) acts as an epithelial gatekeeper and EMT suppressor to shield the endoderm from undergoing a mesenchymal transition. Altogether, these results not only establish the morphogenetic details of germ layer formation, but also have broader implications for stem cell differentiation and cancer metastasis., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
34. Promoter G-quadruplexes and transcription factors cooperate to shape the cell type-specific transcriptome.
- Author
-
Lago S, Nadai M, Cernilogar FM, Kazerani M, Domíniguez Moreno H, Schotta G, and Richter SN
- Subjects
- Base Sequence, Binding Sites genetics, Cell Line, Cell Line, Tumor, DNA chemistry, DNA genetics, DNA metabolism, Humans, Nucleic Acid Conformation, Protein Binding, Sp1 Transcription Factor metabolism, Transcription Factor AP-1 metabolism, G-Quadruplexes, Gene Expression Profiling methods, Promoter Regions, Genetic genetics, Transcription Factors metabolism, Transcriptome genetics
- Abstract
Cell identity is maintained by activation of cell-specific gene programs, regulated by epigenetic marks, transcription factors and chromatin organization. DNA G-quadruplex (G4)-folded regions in cells were reported to be associated with either increased or decreased transcriptional activity. By G4-ChIP-seq/RNA-seq analysis on liposarcoma cells we confirmed that G4s in promoters are invariably associated with high transcription levels in open chromatin. Comparing G4 presence, location and transcript levels in liposarcoma cells to available data on keratinocytes, we showed that the same promoter sequences of the same genes in the two cell lines had different G4-folding state: high transcript levels consistently associated with G4-folding. Transcription factors AP-1 and SP1, whose binding sites were the most significantly represented in G4-folded sequences, coimmunoprecipitated with their G4-folded promoters. Thus, G4s and their associated transcription factors cooperate to determine cell-specific transcriptional programs, making G4s to strongly emerge as new epigenetic regulators of the transcription machinery.
- Published
- 2021
- Full Text
- View/download PDF
35. Suv4-20h2 protects against influenza virus infection by suppression of chromatin loop formation.
- Author
-
Shiimori M, Ichida Y, Nukiwa R, Sakuma T, Abe H, Kajitani R, Fujino Y, Kikuchi A, Kawamura T, Kodama T, Toyooka S, Shirahige K, Schotta G, Kuba K, Itoh T, and Imai Y
- Abstract
The spatial organization of chromatin is known to be highly dynamic in response to environmental stress. However, it remains unknown how chromatin dynamics contributes to or modulates disease pathogenesis. Here, we show that upon influenza virus infection, the H4K20me3 methyltransferase Suv4-20h2 binds the viral protein NP, which results in the inactivation of Suv4-20h2 and the dissociation of cohesin from Suv4-20h2. Inactivation of Suv4-20h2 by viral infection or genetic deletion allows the formation of an active chromatin loop at the HoxC8-HoxC6 loci coincident with cohesin loading. HoxC8 and HoxC6 proteins in turn enhance viral replication by inhibiting the Wnt-β-catenin mediated interferon response. Importantly, loss of Suv4-20h2 augments the pathology of influenza infection in vivo . Thus, Suv4-20h2 acts as a safeguard against influenza virus infection by suppressing cohesin-mediated loop formation., Competing Interests: The authors declare no competing interests., (© 2021.)
- Published
- 2021
- Full Text
- View/download PDF
36. Epstein-Barr virus inactivates the transcriptome and disrupts the chromatin architecture of its host cell in the first phase of lytic reactivation.
- Author
-
Buschle A, Mrozek-Gorska P, Cernilogar FM, Ettinger A, Pich D, Krebs S, Mocanu B, Blum H, Schotta G, Straub T, and Hammerschmidt W
- Subjects
- Binding Sites, Cell Line, Chromatin chemistry, Chromatin metabolism, DNA metabolism, Herpesvirus 4, Human genetics, Herpesvirus 4, Human metabolism, Humans, Chromatin virology, Gene Expression Regulation, Herpesvirus 4, Human physiology, Trans-Activators metabolism, Transcriptome
- Abstract
Epstein-Barr virus (EBV), a herpes virus also termed HHV 4 and the first identified human tumor virus, establishes a stable, long-term latent infection in human B cells, its preferred host. Upon induction of EBV's lytic phase, the latently infected cells turn into a virus factory, a process that is governed by EBV. In the lytic, productive phase, all herpes viruses ensure the efficient induction of all lytic viral genes to produce progeny, but certain of these genes also repress the ensuing antiviral responses of the virally infected host cells, regulate their apoptotic death or control the cellular transcriptome. We now find that EBV causes previously unknown massive and global alterations in the chromatin of its host cell upon induction of the viral lytic phase and prior to the onset of viral DNA replication. The viral initiator protein of the lytic cycle, BZLF1, binds to >105 binding sites with different sequence motifs in cellular chromatin in a concentration dependent manner implementing a binary molar switch probably to prevent noise-induced erroneous induction of EBV's lytic phase. Concomitant with DNA binding of BZLF1, silent chromatin opens locally as shown by ATAC-seq experiments, while previously wide-open cellular chromatin becomes inaccessible on a global scale within hours. While viral transcripts increase drastically, the induction of the lytic phase results in a massive reduction of cellular transcripts and a loss of chromatin-chromatin interactions of cellular promoters with their distal regulatory elements as shown in Capture-C experiments. Our data document that EBV's lytic cycle induces discrete early processes that disrupt the architecture of host cellular chromatin and repress the cellular epigenome and transcriptome likely supporting the efficient de novo synthesis of this herpes virus., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
37. Loss-of-function mutations in the histone methyltransferase EZH2 promote chemotherapy resistance in AML.
- Author
-
Kempf JM, Weser S, Bartoschek MD, Metzeler KH, Vick B, Herold T, Völse K, Mattes R, Scholz M, Wange LE, Festini M, Ugur E, Roas M, Weigert O, Bultmann S, Leonhardt H, Schotta G, Hiddemann W, Jeremias I, and Spiekermann K
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation, Neoplastic drug effects, Humans, Leukemia, Myeloid, Acute diagnosis, Neoplasm Recurrence, Local pathology, Up-Regulation drug effects, Up-Regulation genetics, Xenograft Model Antitumor Assays, Drug Resistance, Neoplasm genetics, Enhancer of Zeste Homolog 2 Protein genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Loss of Function Mutation genetics
- Abstract
Chemotherapy resistance is the main impediment in the treatment of acute myeloid leukaemia (AML). Despite rapid advances, the various mechanisms inducing resistance development remain to be defined in detail. Here we report that loss-of-function mutations (LOF) in the histone methyltransferase EZH2 have the potential to confer resistance against the chemotherapeutic agent cytarabine. We identify seven distinct EZH2 mutations leading to loss of H3K27 trimethylation via multiple mechanisms. Analysis of matched diagnosis and relapse samples reveal a heterogenous regulation of EZH2 and a loss of EZH2 in 50% of patients. We confirm that loss of EZH2 induces resistance against cytarabine in the cell lines HEK293T and K562 as well as in a patient-derived xenograft model. Proteomics and transcriptomics analysis reveal that resistance is conferred by upregulation of multiple direct and indirect EZH2 target genes that are involved in apoptosis evasion, augmentation of proliferation and alteration of transmembrane transporter function. Our data indicate that loss of EZH2 results in upregulation of its target genes, providing the cell with a selective growth advantage, which mediates chemotherapy resistance.
- Published
- 2021
- Full Text
- View/download PDF
38. SETDB1 is required for intestinal epithelial differentiation and the prevention of intestinal inflammation.
- Author
-
Južnić L, Peuker K, Strigli A, Brosch M, Herrmann A, Häsler R, Koch M, Matthiesen L, Zeissig Y, Löscher BS, Nuber A, Schotta G, Neumeister V, Chavakis T, Kurth T, Lesche M, Dahl A, von Mässenhausen A, Linkermann A, Schreiber S, Aden K, Rosenstiel PC, Franke A, Hampe J, and Zeissig S
- Subjects
- Animals, Cell Differentiation, Epithelial Cells metabolism, Female, Gene Silencing, Homeostasis genetics, Humans, Loss of Function Mutation, Male, Mice, Histone-Lysine N-Methyltransferase genetics, Inflammatory Bowel Diseases genetics, Intestinal Mucosa metabolism
- Abstract
Objective: The intestinal epithelium is a rapidly renewing tissue which plays central roles in nutrient uptake, barrier function and the prevention of intestinal inflammation. Control of epithelial differentiation is essential to these processes and is dependent on cell type-specific activity of transcription factors which bind to accessible chromatin. Here, we studied the role of SET Domain Bifurcated Histone Lysine Methyltransferase 1, also known as ESET (SETDB1), a histone H3K9 methyltransferase, in intestinal epithelial homeostasis and IBD., Design: We investigated mice with constitutive and inducible intestinal epithelial deletion of Setdb1 , studied the expression of SETDB1 in patients with IBD and mouse models of IBD, and investigated the abundance of SETDB1 variants in healthy individuals and patients with IBD., Results: Deletion of intestinal epithelial Setdb1 in mice was associated with defects in intestinal epithelial differentiation, barrier disruption, inflammation and mortality. Mechanistic studies showed that loss of SETDB1 leads to de-silencing of endogenous retroviruses, DNA damage and intestinal epithelial cell death. Predicted loss-of-function variants in human SETDB1 were considerably less frequently observed than expected, consistent with a critical role of SETDB1 in human biology. While the vast majority of patients with IBD showed unimpaired mucosal SETDB1 expression, comparison of IBD and non-IBD exomes revealed over-representation of individual rare missense variants in SETDB1 in IBD, some of which are predicted to be associated with loss of function and may contribute to the pathogenesis of intestinal inflammation., Conclusion: SETDB1 plays an essential role in intestinal epithelial homeostasis. Future work is required to investigate whether rare variants in SETDB1 contribute to the pathogenesis of IBD., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
39. The MDM2 inducible promoter folds into four-tetrad antiparallel G-quadruplexes targetable to fight malignant liposarcoma.
- Author
-
Lago S, Nadai M, Ruggiero E, Tassinari M, Marušič M, Tosoni B, Frasson I, Cernilogar FM, Pirota V, Doria F, Plavec J, Schotta G, and Richter SN
- Subjects
- Apoptosis, Cell Cycle, Cell Line, Tumor, Computer Simulation, Humans, Ligands, Models, Genetic, Neoplasm Proteins metabolism, Nuclear Proteins metabolism, Protein Interaction Mapping, Proteolysis, Proto-Oncogene Proteins c-mdm2 biosynthesis, Tumor Suppressor Protein p53 metabolism, G-Quadruplexes, Gene Expression Regulation, Neoplastic genetics, Liposarcoma therapy, Molecular Targeted Therapy, Promoter Regions, Genetic genetics, Proto-Oncogene Proteins c-mdm2 genetics, Soft Tissue Neoplasms therapy
- Abstract
Well-differentiated liposarcoma (WDLPS) is a malignant neoplasia hard to diagnose and treat. Its main molecular signature is amplification of the MDM2-containing genomic region. The MDM2 oncogene is the master regulator of p53: its overexpression enhances p53 degradation and inhibits apoptosis, leading to the tumoral phenotype. Here, we show that the MDM2 inducible promoter G-rich region folds into stable G-quadruplexes both in vitro and in vivo and it is specifically recognized by cellular helicases. Cell treatment with G-quadruplex-ligands reduces MDM2 expression and p53 degradation, thus stimulating cancer cell cycle arrest and apoptosis. Structural characterization of the MDM2 G-quadruplex revealed an extraordinarily stable, unique four-tetrad antiparallel dynamic conformation, amenable to selective targeting. These data indicate the feasibility of an out-of-the-box G-quadruplex-targeting approach to defeat WDLPS and all tumours where restoration of wild-type p53 is sought. They also point to G-quadruplex-dependent genomic instability as possible cause of MDM2 expansion and WDLPS tumorigenesis., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
40. HIV-1 infection activates endogenous retroviral promoters regulating antiviral gene expression.
- Author
-
Srinivasachar Badarinarayan S, Shcherbakova I, Langer S, Koepke L, Preising A, Hotter D, Kirchhoff F, Sparrer KMJ, Schotta G, and Sauter D
- Subjects
- Animals, CD4-Positive T-Lymphocytes cytology, GTP-Binding Proteins genetics, GTP-Binding Proteins immunology, HEK293 Cells, HIV Infections immunology, HIV-1, Humans, Macaca mulatta, T-Lymphocyte Subsets cytology, CD4-Positive T-Lymphocytes immunology, Endogenous Retroviruses genetics, Gene Expression Regulation immunology, HIV Infections genetics, Promoter Regions, Genetic, T-Lymphocyte Subsets immunology
- Abstract
Although endogenous retroviruses (ERVs) are known to harbor cis-regulatory elements, their role in modulating cellular immune responses remains poorly understood. Using an RNA-seq approach, we show that several members of the ERV9 lineage, particularly LTR12C elements, are activated upon HIV-1 infection of primary CD4+ T cells. Intriguingly, HIV-1-induced ERVs harboring transcription start sites are primarily found in the vicinity of immunity genes. For example, HIV-1 infection activates LTR12C elements upstream of the interferon-inducible genes GBP2 and GBP5 that encode for broad-spectrum antiviral factors. Reporter assays demonstrated that these LTR12C elements drive gene expression in primary CD4+ T cells. In line with this, HIV-1 infection triggered the expression of a unique GBP2 transcript variant by activating a cryptic transcription start site within LTR12C. Furthermore, stimulation with HIV-1-induced cytokines increased GBP2 and GBP5 expression in human cells, but not in macaque cells that naturally lack the GBP5 gene and the LTR12C element upstream of GBP2. Finally, our findings suggest that GBP2 and GBP5 have already been active against ancient viral pathogens as they suppress the maturation of the extinct retrovirus HERV-K (HML-2). In summary, our findings uncover how human cells can exploit remnants of once-infectious retroviruses to regulate antiviral gene expression., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2020
- Full Text
- View/download PDF
41. The Kidney Contains Ontogenetically Distinct Dendritic Cell and Macrophage Subtypes throughout Development That Differ in Their Inflammatory Properties.
- Author
-
Salei N, Rambichler S, Salvermoser J, Papaioannou NE, Schuchert R, Pakalniškytė D, Li N, Marschner JA, Lichtnekert J, Stremmel C, Cernilogar FM, Salvermoser M, Walzog B, Straub T, Schotta G, Anders HJ, Schulz C, and Schraml BU
- Subjects
- Acute Kidney Injury immunology, Age Factors, Animals, CD11b Antigen analysis, CX3C Chemokine Receptor 1 analysis, Calcium-Binding Proteins analysis, Cisplatin pharmacology, Histocompatibility Antigens Class II analysis, Kidney drug effects, Kidney metabolism, Lectins, C-Type analysis, Mice, Mice, Inbred C57BL, Receptors, G-Protein-Coupled analysis, Receptors, Immunologic analysis, Dendritic Cells immunology, Kidney immunology, Macrophages immunology, Nephritis immunology
- Abstract
Background: Mononuclear phagocytes (MPs), including macrophages, monocytes, and dendritic cells (DCs), are phagocytic cells with important roles in immunity. The developmental origin of kidney DCs has been highly debated because of the large phenotypic overlap between macrophages and DCs in this tissue., Methods: We used fate mapping, RNA sequencing, flow cytometry, confocal microscopy, and histo-cytometry to assess the origin and phenotypic and functional properties of renal DCs in healthy kidney and of DCs after cisplatin and ischemia reperfusion-induced kidney injury., Results: Adult kidney contains at least four subsets of MPs with prominent Clec9a -expression history indicating a DC origin. We demonstrate that these populations are phenotypically, functionally, and transcriptionally distinct from each other. We also show these kidney MPs exhibit unique age-dependent developmental heterogeneity. Kidneys from newborn mice contain a prominent population of embryonic-derived MHCII
neg F4/80hi CD11blow macrophages that express T cell Ig and mucin domain containing 4 (TIM-4) and MER receptor tyrosine kinase (MERTK). These macrophages are replaced within a few weeks after birth by phenotypically similar cells that express MHCII but lack TIM-4 and MERTK. MHCII+ F4/80hi cells exhibit prominent Clec9a- expression history in adulthood but not early life, indicating additional age-dependent developmental heterogeneity. In AKI, MHCIIneg F4/80hi cells reappear in adult kidneys as a result of MHCII downregulation by resident MHCII+ F4/80hi cells, possibly in response to prostaglandin E2 (PGE2). RNA sequencing further suggests MHCII+ F4/80hi cells help coordinate the recruitment of inflammatory cells during renal injury., Conclusions: Distinct developmental programs contribute to renal DC and macrophage populations throughout life, which could have important implications for therapies targeting these cells., (Copyright © 2020 by the American Society of Nephrology.)- Published
- 2020
- Full Text
- View/download PDF
42. Loss of KDM6A confers drug resistance in acute myeloid leukemia.
- Author
-
Stief SM, Hanneforth AL, Weser S, Mattes R, Carlet M, Liu WH, Bartoschek MD, Domínguez Moreno H, Oettle M, Kempf J, Vick B, Ksienzyk B, Tizazu B, Rothenberg-Thurley M, Quentmeier H, Hiddemann W, Vosberg S, Greif PA, Metzeler KH, Schotta G, Bultmann S, Jeremias I, Leonhardt H, and Spiekermann K
- Subjects
- Animals, Heterografts, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Mice, Mutation, Drug Resistance, Neoplasm physiology, Histone Demethylases genetics, Histone Demethylases metabolism, Leukemia, Myeloid, Acute pathology
- Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic neoplasm resulting from the malignant transformation of myeloid progenitors. Despite intensive chemotherapy leading to initial treatment responses, relapse caused by intrinsic or acquired drug resistance represents a major challenge. Here, we report that histone 3 lysine 27 demethylase KDM6A (UTX) is targeted by inactivating mutations and mutation-independent regulation in relapsed AML. Analyses of matched diagnosis and relapse specimens from individuals with KDM6A mutations showed an outgrowth of the KDM6A mutated tumor population at relapse. KDM6A expression is heterogeneously regulated and relapse-specific loss of KDM6A was observed in 45.7% of CN-AML patients. KDM6A-null myeloid leukemia cells were more resistant to treatment with the chemotherapeutic agents cytarabine (AraC) and daunorubicin. Inducible re-expression of KDM6A in KDM6A-null cell lines suppressed proliferation and sensitized cells again to AraC treatment. RNA expression analysis and functional studies revealed that resistance to AraC was conferred by downregulation of the nucleoside membrane transporter ENT1 (SLC29A1) by reduced H3K27 acetylation at the ENT1 locus. Our results show that loss of KDM6A provides cells with a selective advantage during chemotherapy, which ultimately leads to the observed outgrowth of clones with KDM6A mutations or reduced KDM6A expression at relapse.
- Published
- 2020
- Full Text
- View/download PDF
43. Pre-marked chromatin and transcription factor co-binding shape the pioneering activity of Foxa2.
- Author
-
Cernilogar FM, Hasenöder S, Wang Z, Scheibner K, Burtscher I, Sterr M, Smialowski P, Groh S, Evenroed IM, Gilfillan GD, Lickert H, and Schotta G
- Subjects
- Animals, Binding Sites genetics, Cell Differentiation genetics, Chromatin Assembly and Disassembly genetics, Endoderm cytology, GATA4 Transcription Factor genetics, GATA4 Transcription Factor metabolism, Gene Expression Regulation, Developmental genetics, Hepatocyte Nuclear Factor 3-beta genetics, Histone Code, Histones metabolism, Mice, Mice, Knockout, Models, Genetic, Mouse Embryonic Stem Cells cytology, Signal Transduction, Chromatin metabolism, Hepatocyte Nuclear Factor 3-beta metabolism, Mouse Embryonic Stem Cells metabolism
- Abstract
Pioneer transcription factors (PTF) can recognize their binding sites on nucleosomal DNA and trigger chromatin opening for recruitment of other non-pioneer transcription factors. However, critical properties of PTFs are still poorly understood, such as how these transcription factors selectively recognize cell type-specific binding sites and under which conditions they can initiate chromatin remodelling. Here we show that early endoderm binding sites of the paradigm PTF Foxa2 are epigenetically primed by low levels of active chromatin modifications in embryonic stem cells (ESC). Priming of these binding sites is supported by preferential recruitment of Foxa2 to endoderm binding sites compared to lineage-inappropriate binding sites, when ectopically expressed in ESCs. We further show that binding of Foxa2 is required for chromatin opening during endoderm differentiation. However, increased chromatin accessibility was only detected on binding sites which are synergistically bound with other endoderm transcription factors. Thus, our data suggest that binding site selection of PTFs is directed by the chromatin environment and that chromatin opening requires collaboration of PTFs with additional transcription factors., (© The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2019
- Full Text
- View/download PDF
44. Point mutations in the PDX1 transactivation domain impair human β-cell development and function.
- Author
-
Wang X, Sterr M, Ansarullah, Burtscher I, Böttcher A, Beckenbauer J, Siehler J, Meitinger T, Häring HU, Staiger H, Cernilogar FM, Schotta G, Irmler M, Beckers J, Wright CVE, Bakhti M, and Lickert H
- Subjects
- Adult, Carboxylic Ester Hydrolases genetics, Carboxylic Ester Hydrolases metabolism, Cell Line, Female, Homeodomain Proteins chemistry, Homeodomain Proteins metabolism, Humans, Insulin-Secreting Cells cytology, Loss of Function Mutation, Male, Protein Domains, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, Trans-Activators chemistry, Trans-Activators metabolism, Transcription Factors genetics, Transcription Factors metabolism, Cell Differentiation, Diabetes Mellitus genetics, Homeodomain Proteins genetics, Insulin Secretion, Insulin-Secreting Cells metabolism, Point Mutation, Trans-Activators genetics
- Abstract
Objective: Hundreds of missense mutations in the coding region of PDX1 exist; however, if these mutations predispose to diabetes mellitus is unknown., Methods: In this study, we screened a large cohort of subjects with increased risk for diabetes and identified two subjects with impaired glucose tolerance carrying common, heterozygous, missense mutations in the PDX1 coding region leading to single amino acid exchanges (P33T, C18R) in its transactivation domain. We generated iPSCs from patients with heterozygous PDX1
P33T/+ , PDX1C18R/+ mutations and engineered isogenic cell lines carrying homozygous PDX1P33T/P33T , PDX1C18R/C18R mutations and a heterozygous PDX1 loss-of-function mutation (PDX1+/- )., Results: Using an in vitro β-cell differentiation protocol, we demonstrated that both, heterozygous PDX1P33T/+ , PDX1C18R/+ and homozygous PDX1P33T/P33T , PDX1C18R/C18R mutations impair β-cell differentiation and function. Furthermore, PDX1+/- and PDX1P33T/P33T mutations reduced differentiation efficiency of pancreatic progenitors (PPs), due to downregulation of PDX1-bound genes, including transcription factors MNX1 and PDX1 as well as insulin resistance gene CES1. Additionally, both PDX1P33T/+ and PDX1P33T/P33T mutations in PPs reduced the expression of PDX1-bound genes including the long-noncoding RNA, MEG3 and the imprinted gene NNAT, both involved in insulin synthesis and secretion., Conclusions: Our results reveal mechanistic details of how common coding mutations in PDX1 impair human pancreatic endocrine lineage formation and β-cell function and contribute to the predisposition for diabetes., (Copyright © 2019 The Authors. Published by Elsevier GmbH.. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
45. BZLF1 interacts with chromatin remodelers promoting escape from latent infections with EBV.
- Author
-
Schaeffner M, Mrozek-Gorska P, Buschle A, Woellmer A, Tagawa T, Cernilogar FM, Schotta G, Krietenstein N, Lieleg C, Korber P, and Hammerschmidt W
- Subjects
- ATPases Associated with Diverse Cellular Activities genetics, Adenosine Triphosphatases metabolism, Binding Sites, Cell Survival, Chromosomal Proteins, Non-Histone metabolism, DNA, Viral metabolism, DNA-Binding Proteins genetics, Gene Expression Regulation, Viral, Gene Knockdown Techniques, HEK293 Cells, Histones metabolism, Humans, RNA, Small Interfering genetics, THP-1 Cells, Transfection, Virus Activation physiology, ATPases Associated with Diverse Cellular Activities metabolism, Chromatin Assembly and Disassembly physiology, DNA-Binding Proteins metabolism, Epstein-Barr Virus Infections virology, Herpesvirus 4, Human physiology, Trans-Activators genetics, Trans-Activators metabolism, Virus Latency
- Abstract
A hallmark of EBV infections is its latent phase, when all viral lytic genes are repressed. Repression results from a high nucleosome occupancy and epigenetic silencing by cellular factors such as the Polycomb repressive complex 2 (PRC2) and DNA methyltransferases that, respectively, introduce repressive histone marks and DNA methylation. The viral transcription factor BZLF1 acts as a molecular switch to induce transition from the latent to the lytic or productive phase of EBV's life cycle. It is unknown how BZLF1 can bind to the epigenetically silenced viral DNA and whether it directly reactivates the viral genome through chromatin remodeling. We addressed these fundamental questions and found that BZLF1 binds to nucleosomal DNA motifs both in vivo and in vitro. BZLF1 co-precipitates with cellular chromatin remodeler ATPases, and the knock-down of one of them, INO80, impaired lytic reactivation and virus synthesis. In Assay for Transposase-Accessible Chromatin-seq experiments, non-accessible chromatin opens up locally when BZLF1 binds to its cognate sequence motifs in viral DNA. We conclude that BZLF1 reactivates the EBV genome by directly binding to silenced chromatin and recruiting cellular chromatin-remodeling enzymes, which implement a permissive state for lytic viral transcription. BZLF1 shares this mode of action with a limited number of cellular pioneer factors, which are instrumental in transcriptional activation, differentiation, and reprogramming in all eukaryotic cells., (© 2019 Schaeffner et al.)
- Published
- 2019
- Full Text
- View/download PDF
46. Blimp1 Prevents Methylation of Foxp3 and Loss of Regulatory T Cell Identity at Sites of Inflammation.
- Author
-
Garg G, Muschaweckh A, Moreno H, Vasanthakumar A, Floess S, Lepennetier G, Oellinger R, Zhan Y, Regen T, Hiltensperger M, Peter C, Aly L, Knier B, Palam LR, Kapur R, Kaplan MH, Waisman A, Rad R, Schotta G, Huehn J, Kallies A, and Korn T
- Subjects
- Animals, DNA (Cytosine-5-)-Methyltransferases antagonists & inhibitors, DNA (Cytosine-5-)-Methyltransferases immunology, DNA (Cytosine-5-)-Methyltransferases metabolism, DNA Methyltransferase 3A, Encephalomyelitis, Autoimmune, Experimental genetics, Encephalomyelitis, Autoimmune, Experimental metabolism, Epigenesis, Genetic, Female, Forkhead Transcription Factors biosynthesis, Forkhead Transcription Factors immunology, Genomic Imprinting, Interleukin-6 immunology, Male, Mice, Mice, Inbred C57BL, T-Lymphocytes, Regulatory metabolism, DNA Methylation, Encephalomyelitis, Autoimmune, Experimental immunology, Forkhead Transcription Factors genetics, Positive Regulatory Domain I-Binding Factor 1 genetics, Positive Regulatory Domain I-Binding Factor 1 immunology, T-Lymphocytes, Regulatory immunology
- Abstract
Foxp3
+ regulatory T (Treg) cells restrict immune pathology in inflamed tissues; however, an inflammatory environment presents a threat to Treg cell identity and function. Here, we establish a transcriptional signature of central nervous system (CNS) Treg cells that accumulate during experimental autoimmune encephalitis (EAE) and identify a pathway that maintains Treg cell function and identity during severe inflammation. This pathway is dependent on the transcriptional regulator Blimp1, which prevents downregulation of Foxp3 expression and "toxic" gain-of-function of Treg cells in the inflamed CNS. Blimp1 negatively regulates IL-6- and STAT3-dependent Dnmt3a expression and function restraining methylation of Treg cell-specific conserved non-coding sequence 2 (CNS2) in the Foxp3 locus. Consequently, CNS2 is heavily methylated when Blimp1 is ablated, leading to a loss of Foxp3 expression and severe disease. These findings identify a Blimp1-dependent pathway that preserves Treg cell stability in inflamed non-lymphoid tissues., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
47. The Aryl Hydrocarbon Receptor Pathway Defines the Time Frame for Restorative Neurogenesis.
- Author
-
Di Giaimo R, Durovic T, Barquin P, Kociaj A, Lepko T, Aschenbroich S, Breunig CT, Irmler M, Cernilogar FM, Schotta G, Barbosa JS, Trümbach D, Baumgart EV, Neuner AM, Beckers J, Wurst W, Stricker SH, and Ninkovic J
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Cell Survival, Ependymoglial Cells cytology, Ependymoglial Cells metabolism, Mitosis, Neurons cytology, Time Factors, Zebrafish, Neurogenesis, Receptors, Aryl Hydrocarbon metabolism, Signal Transduction
- Abstract
Zebrafish have a high capacity to replace lost neurons after brain injury. New neurons involved in repair are generated by a specific set of glial cells, known as ependymoglial cells. We analyze changes in the transcriptome of ependymoglial cells and their progeny after injury to infer the molecular pathways governing restorative neurogenesis. We identify the aryl hydrocarbon receptor (AhR) as a regulator of ependymoglia differentiation toward post-mitotic neurons. In vivo imaging shows that high AhR signaling promotes the direct conversion of a specific subset of ependymoglia into post-mitotic neurons, while low AhR signaling promotes ependymoglial proliferation. Interestingly, we observe the inactivation of AhR signaling shortly after injury followed by a return to the basal levels 7 days post injury. Interference with timely AhR regulation after injury leads to aberrant restorative neurogenesis. Taken together, we identify AhR signaling as a crucial regulator of restorative neurogenesis timing in the zebrafish brain., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
48. Genome-wide analysis of PDX1 target genes in human pancreatic progenitors.
- Author
-
Wang X, Sterr M, Burtscher I, Chen S, Hieronimus A, Machicao F, Staiger H, Häring HU, Lederer G, Meitinger T, Cernilogar FM, Schotta G, Irmler M, Beckers J, Hrabě de Angelis M, Ray M, Wright CVE, Bakhti M, and Lickert H
- Subjects
- Calcium-Binding Proteins, Cell Differentiation, Cells, Cultured, Chromatin Assembly and Disassembly, Diabetes Mellitus, Type 2 metabolism, Enhancer Elements, Genetic, Genome-Wide Association Study, Hepatocyte Nuclear Factor 1-beta genetics, Hepatocyte Nuclear Factor 1-beta metabolism, Homeodomain Proteins metabolism, Humans, Induced Pluripotent Stem Cells cytology, Insulin-Secreting Cells cytology, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Myeloid Ecotropic Viral Integration Site 1 Protein genetics, Myeloid Ecotropic Viral Integration Site 1 Protein metabolism, Polymorphism, Single Nucleotide, Protein Binding, Regulatory Factor X Transcription Factors genetics, Regulatory Factor X Transcription Factors metabolism, Trans-Activators metabolism, Transcription Factor 7-Like 2 Protein genetics, Transcription Factor 7-Like 2 Protein metabolism, Diabetes Mellitus, Type 2 genetics, Homeodomain Proteins genetics, Induced Pluripotent Stem Cells metabolism, Insulin-Secreting Cells metabolism, Trans-Activators genetics
- Abstract
Objective: Homozygous loss-of-function mutations in the gene coding for the homeobox transcription factor (TF) PDX1 leads to pancreatic agenesis, whereas heterozygous mutations can cause Maturity-Onset Diabetes of the Young 4 (MODY4). Although the function of Pdx1 is well studied in pre-clinical models during insulin-producing β-cell development and homeostasis, it remains elusive how this TF controls human pancreas development by regulating a downstream transcriptional program. Also, comparative studies of PDX1 binding patterns in pancreatic progenitors and adult β-cells have not been conducted so far. Furthermore, many studies reported the association between single nucleotide polymorphisms (SNPs) and T2DM, and it has been shown that islet enhancers are enriched in T2DM-associated SNPs. Whether regions, harboring T2DM-associated SNPs are PDX1 bound and active at the pancreatic progenitor stage has not been reported so far., Methods: In this study, we have generated a novel induced pluripotent stem cell (iPSC) line that efficiently differentiates into human pancreatic progenitors (PPs). Furthermore, PDX1 and H3K27ac chromatin immunoprecipitation sequencing (ChIP-seq) was used to identify PDX1 transcriptional targets and active enhancer and promoter regions. To address potential differences in the function of PDX1 during development and adulthood, we compared PDX1 binding profiles from PPs and adult islets. Moreover, combining ChIP-seq and GWAS meta-analysis data we identified T2DM-associated SNPs in PDX1 binding sites and active chromatin regions., Results: ChIP-seq for PDX1 revealed a total of 8088 PDX1-bound regions that map to 5664 genes in iPSC-derived PPs. The PDX1 target regions include important pancreatic TFs, such as PDX1 itself, RFX6, HNF1B, and MEIS1, which were activated during the differentiation process as revealed by the active chromatin mark H3K27ac and mRNA expression profiling, suggesting that auto-regulatory feedback regulation maintains PDX1 expression and initiates a pancreatic TF program. Remarkably, we identified several PDX1 target genes that have not been reported in the literature in human so far, including RFX3, required for ciliogenesis and endocrine differentiation in mouse, and the ligand of the Notch receptor DLL1, which is important for endocrine induction and tip-trunk patterning. The comparison of PDX1 profiles from PPs and adult human islets identified sets of stage-specific target genes, associated with early pancreas development and adult β-cell function, respectively. Furthermore, we found an enrichment of T2DM-associated SNPs in active chromatin regions from iPSC-derived PPs. Two of these SNPs fall into PDX1 occupied sites that are located in the intronic regions of TCF7L2 and HNF1B. Both of these genes are key transcriptional regulators of endocrine induction and mutations in cis-regulatory regions predispose to diabetes., Conclusions: Our data provide stage-specific target genes of PDX1 during in vitro differentiation of stem cells into pancreatic progenitors that could be useful to identify pathways and molecular targets that predispose for diabetes. In addition, we show that T2DM-associated SNPs are enriched in active chromatin regions at the pancreatic progenitor stage, suggesting that the susceptibility to T2DM might originate from imperfect execution of a β-cell developmental program., (Copyright © 2018 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
49. Mammalian HP1 Isoforms Have Specific Roles in Heterochromatin Structure and Organization.
- Author
-
Bosch-Presegué L, Raurell-Vila H, Thackray JK, González J, Casal C, Kane-Goldsmith N, Vizoso M, Brown JP, Gómez A, Ausió J, Zimmermann T, Esteller M, Schotta G, Singh PB, Serrano L, and Vaquero A
- Subjects
- Amino Acid Sequence genetics, Animals, Chromatin metabolism, Chromobox Protein Homolog 5, HeLa Cells, Humans, Mammals metabolism, Protein Binding genetics, Protein Binding immunology, Protein Isoforms genetics, Protein Isoforms metabolism, Transcription Factors genetics, Transcription Factors metabolism, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism
- Abstract
HP1 is a structural component of heterochromatin. Mammalian HP1 isoforms HP1α, HP1β, and HP1γ play different roles in genome stability, but their precise role in heterochromatin structure is unclear. Analysis of Hp1α
-/- , Hp1β-/- , and Hp1γ-/- MEFs show that HP1 proteins have both redundant and unique functions within pericentric heterochromatin (PCH) and also act globally throughout the genome. HP1α confines H4K20me3 and H3K27me3 to regions within PCH, while its absence results in a global hyper-compaction of chromatin associated with a specific pattern of mitotic defects. In contrast, HP1β is functionally associated with Suv4-20h2 and H4K20me3, and its loss induces global chromatin decompaction and an abnormal enrichment of CTCF in PCH and other genomic regions. Our work provides insight into the roles of HP1 proteins in heterochromatin structure and genome stability., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
50. Histone H4K20 tri-methylation at late-firing origins ensures timely heterochromatin replication.
- Author
-
Brustel J, Kirstein N, Izard F, Grimaud C, Prorok P, Cayrou C, Schotta G, Abdelsamie AF, Déjardin J, Méchali M, Baldacci G, Sardet C, Cadoret JC, Schepers A, and Julien E
- Subjects
- Humans, Methylation, DNA Replication, Heterochromatin metabolism, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Lysine metabolism, Protein Processing, Post-Translational
- Abstract
Among other targets, the protein lysine methyltransferase PR-Set7 induces histone H4 lysine 20 monomethylation (H4K20me1), which is the substrate for further methylation by the Suv4-20h methyltransferase. Although these enzymes have been implicated in control of replication origins, the specific contribution of H4K20 methylation to DNA replication remains unclear. Here, we show that H4K20 mutation in mammalian cells, unlike in Drosophila , partially impairs S-phase progression and protects from DNA re-replication induced by stabilization of PR-Set7. Using Epstein-Barr virus-derived episomes, we further demonstrate that conversion of H4K20me1 to higher H4K20me2/3 states by Suv4-20h is not sufficient to define an efficient origin per se , but rather serves as an enhancer for MCM2-7 helicase loading and replication activation at defined origins. Consistent with this, we find that Suv4-20h-mediated H4K20 tri-methylation (H4K20me3) is required to sustain the licensing and activity of a subset of ORCA/LRWD1-associated origins, which ensure proper replication timing of late-replicating heterochromatin domains. Altogether, these results reveal Suv4-20h-mediated H4K20 tri-methylation as a critical determinant in the selection of active replication initiation sites in heterochromatin regions of mammalian genomes., (© 2017 The Authors.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.