14 results on '"Adriaens C"'
Search Results
2. The long and the short of NEAT1: Roles in paraspeckle formation and cancer : NEAT1 in het kort en het lang: De rol van de twee isovormen in paraspeckles en kanker
- Author
-
Adriaens, C and Marine, J-C
- Abstract
NEAT1 is a well-conserved, highly expressed nuclear RNA that does not code for proteins. It exists in two isoforms of 3.7 and 22.7 kb, of which the longer is the essential architectural component of a nuclear body in the cell, the paraspeckle. In this thesis, we show that the tumor suppressor p53 upregulates NEAT1 transcription leading to the formation of paraspeckles in cultured cells, in mouse skin, and in human tumors of epithelial origin. We used a mouse model of chemically-induced skin carcinogenesis combined with a genetic knockout of Neat1 to demonstrate that this lncRNA contributes to tumor formation. Next, we showed, using in vitro studies, that NEAT1 knockdown sensitizes cancer cells to common chemotherapeutic agents such as doxorubicin and the PARP-inhibitor ABT-888, and that knockdown without external stress already caused an accumulation of DNA damage on its own. In particular, knockdown of the long isoform NEAT1_2, the central building block of the paraspeckle nuclear body, is sufficient to do so, indicating that it may be the paraspeckle which is the functional unit in this process. Knockdown of NEAT1 prevented proper activation of the ATR-Chk1 pathway, decreasing the appropriate DNA repair. This decreased signaling could potentially lead to an increase in collapsed replication forks, causing an increase in S139-phosphorylated Histone 2A.X (ɣ-H2A.X) and an arrest of the cells in the next G1 phase. Finally, higher expression levels of the long isoform in a cohort of cisplatinum-treated ovarian cancer patients could predict poor survival, whereas considering both isoforms together did not have predictive value. The isoform specificity of these latter observations (i.e. knockdown of the long isoform alone was sufficient to induce a growth phenotype) prompted us to follow up on the individual roles of NEAT1_1 and NEAT1_2. We found that despite a common regulation by p53 at the NEAT1 promoter, both isoforms are differentially expressed upon different forms of stress, a process mediated by alternative processing of the NEAT1_1 short isoform at its 3' end. Nutlin-3a, which causes pharmacological activation of p53 and arrests the cells in the G1 phase, led to significant amounts of NEAT1_1 found outside of the paraspeckle nuclear bodies. In contrast, hydroxyurea stalls the cells mid-S phase in addition to activating p53. When treated with this agent, we failed to detect NEAT1_1 outside of nuclear bodies in the majority of the cells. In a non-treated, mixed population, the proportion of cells expressing NEAT1_1 outside of paraspeckles varies greatly. These observations suggested that NEAT1 isoform configurations in the cell could be cell cycle dependent. We subsequently found that NEAT1_1 is the predominant isoform in quiescent (G0) and G1 cells, whereas this isoform disappeared and NEAT1_2 and paraspeckles became primarily expressed from replication (S phase) onwards. Moreover, CRISPR-mediated knockout of the polyadenylation signal revealed that the short isoform does not contribute to the observed replication stress and cell cycle arrest upon knockdown of both. In addition, this isoform does not contribute to NEAT1_2 and paraspeckle function, at least when considering its role of protecting against replication stress. In fact, the data presented here suggest that a long-standing paradigm in paraspeckle research may need to be challenged: from our work it appears that NEAT1_1 does not contribute, at all, to paraspeckle function or formation, and that the hypothesis of NEAT1_1 recruitment into the paraspeckle may be wrong. In effect, in addition to our estimate that NEAT1_1 expression is almost zero in conditions where only NEAT1_2 is observed, we also found a mechanism of NEAT1_1 degradation by the RNA exosome. Here, further studies are needed to establish a cell cycle stage-specific role of the RNA exosome-mediated degradation process. Finally, repeating our short-term skin carcinogenesis experiment in mice lacking both Neat1 isoforms as before, but now also using a knockout mouse model of only Neat1_1 indicated that Neat1_2 competent cells (WT and knock out for Neat1_1) behave in a similar fashion, whereas mice that are Neat1_2 and paraspeckle incompetent show a decrease in epidermal hyperplasia and an increase in DNA damage as shown in the first part of this work. Similarly, phenotypes described by our lab and others affecting the mammary gland and other aspects of mouse fertility could not be recapitulated in mice lacking only Neat1_1, indicating that it is effectively the long isoform alone that confers a survival and growth advantage when cells need to withstand the internal and external stressors. In summary, the work presented in this thesis revealed that cancer cells can hijack the protective function of NEAT1_2/paraspeckles, but, that the more abundant and better conserved NEAT1_1 short isoform does not seem to contribute to this process, and that, although well studied, many aspects of NEAT1 and paraspeckle biology still remain to be understood. status: published
- Published
- 2019
3. Mechanism of Coordinated Access to Orphan Drugs
- Author
-
DeRidder, H., primary, Arickx, F., additional, Adriaens, C., additional, Quanten, A., additional, Mortier, M., additional, and Kleinermans, D., additional
- Published
- 2013
- Full Text
- View/download PDF
4. PHP218 - Mechanism of Coordinated Access to Orphan Drugs
- Author
-
DeRidder, H., Arickx, F., Adriaens, C., Quanten, A., Mortier, M., and Kleinermans, D.
- Published
- 2013
- Full Text
- View/download PDF
5. Publisher Correction: p53 induces formation of NEAT1 lncRNA-containing paraspeckles that modulate replication stress response and chemosensitivity.
- Author
-
Adriaens C, Standaert L, Barra J, Latil M, Verfaillie A, Kalev P, Boeckx B, Wijnhoven PWG, Radaelli E, Vermi W, Leucci E, Lapouge G, Beck B, van den Oord J, Nakagawa S, Hirose T, Sablina AA, Lambrechts D, Aerts S, Blanpain C, and Marine JC
- Published
- 2024
- Full Text
- View/download PDF
6. Impact of supraphysiologic MDM2 expression on chromatin networks and therapeutic responses in sarcoma.
- Author
-
Bevill SM, Casaní-Galdón S, El Farran CA, Cytrynbaum EG, Macias KA, Oldeman SE, Oliveira KJ, Moore MM, Hegazi E, Adriaens C, Najm FJ, Demetri GD, Cohen S, Mullen JT, Riggi N, Johnstone SE, and Bernstein BE
- Abstract
Amplification of MDM2 on supernumerary chromosomes is a common mechanism of P53 inactivation across tumors. Here, we investigated the impact of MDM2 overexpression on chromatin, gene expression, and cellular phenotypes in liposarcoma. Three independent regulatory circuits predominate in aggressive, dedifferentiated tumors. RUNX and AP-1 family transcription factors bind mesenchymal gene enhancers. P53 and MDM2 co-occupy enhancers and promoters associated with P53 signaling. When highly expressed, MDM2 also binds thousands of P53-independent growth and stress response genes, whose promoters engage in multi-way topological interactions. Overexpressed MDM2 concentrates within nuclear foci that co-localize with PML and YY1 and could also contribute to P53-independent phenotypes associated with supraphysiologic MDM2. Importantly, we observe striking cell-to-cell variability in MDM2 copy number and expression in tumors and models. Whereas liposarcoma cells are generally sensitive to MDM2 inhibitors and their combination with pro-apoptotic drugs, MDM2-high cells tolerate them and may underlie the poor clinical efficacy of these agents., Competing Interests: B.E.B. declares outside interests in Fulcrum Therapeutics, Arsenal Biosciences, HiFiBio, Cell Signaling Technologies, Design Pharmaceuticals, and Chroma Medicine. G.D.D. reports leadership as co-founder of IDRX; stocks/options/shares in IDRX, Blueprint Medicines, G1 Therapeutics, Caris Life Sciences, Erasca Pharmaceuticals, RELAY Therapeutics, Bessor Pharmaceuticals, CellCarta, IKENA Oncology, and Kojin Therapeutics; paid consulting fees from Bayer, Pfizer, Novartis, Roche/Genentech, GSK, PharmaMar, Daiichi Sankyo, EMD-Serono/Merck KGaA, Mirati, WCG/Arsenal Capital, G1 Therapeutics, Caris Life Sciences, RELAY Therapeutics, CellCarta, IKENA Oncology, Kojin Therapeutics, RAIN Therapeutics, Jazz Pharmaceuticals, Aadi Biosciences, and IDRX; royalties, patents, or licenses from Novartis to Dana-Farber Cancer Institute for “use patent” of imatinib in GIST; and non-financial interests in AACR Science Policy and Government Affairs Committee and Alexandria Real Estate Equities summit conference series., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
7. Genome-wide programmable transcriptional memory by CRISPR-based epigenome editing.
- Author
-
Nuñez JK, Chen J, Pommier GC, Cogan JZ, Replogle JM, Adriaens C, Ramadoss GN, Shi Q, Hung KL, Samelson AJ, Pogson AN, Kim JYS, Chung A, Leonetti MD, Chang HY, Kampmann M, Bernstein BE, Hovestadt V, Gilbert LA, and Weissman JS
- Subjects
- Cell Differentiation, CpG Islands, DNA Methylation, Gene Silencing, Histone Code, Humans, Induced Pluripotent Stem Cells metabolism, Neurons metabolism, Protein Processing, Post-Translational, CRISPR-Cas Systems, Cellular Reprogramming, Epigenesis, Genetic, Epigenome, Gene Editing, Induced Pluripotent Stem Cells cytology, Neurons cytology
- Abstract
A general approach for heritably altering gene expression has the potential to enable many discovery and therapeutic efforts. Here, we present CRISPRoff-a programmable epigenetic memory writer consisting of a single dead Cas9 fusion protein that establishes DNA methylation and repressive histone modifications. Transient CRISPRoff expression initiates highly specific DNA methylation and gene repression that is maintained through cell division and differentiation of stem cells to neurons. Pairing CRISPRoff with genome-wide screens and analysis of chromatin marks establishes rules for heritable gene silencing. We identify single guide RNAs (sgRNAs) capable of silencing the large majority of genes including those lacking canonical CpG islands (CGIs) and reveal a wide targeting window extending beyond annotated CGIs. The broad ability of CRISPRoff to initiate heritable gene silencing even outside of CGIs expands the canonical model of methylation-based silencing and enables diverse applications including genome-wide screens, multiplexed cell engineering, enhancer silencing, and mechanistic exploration of epigenetic inheritance., Competing Interests: Declaration of interests J.K.N., J.C., G.C.P., L.A.G., and J.S.W. have filed patent applications related to CRISPRoff, CRISPRon, and CRISPRi/a screening. J.M.R. consults for Maze Therapeutics. L.A.G., J.S.W., H.Y.C., and B.E.B. consult for and hold equity in Chroma Medicine. J.S.W. declares outside interest in KSQ Therapeutics, Maze Therapeutics, Amgen, and Tessera Therapeutics. M.K. serves on the Scientific Advisory Boards of Engine Biosciences, Casma Therapeutics, and Cajal Neuroscience. B.E.B. declares outside interests in Fulcrum Therapeutics, Arsenal Biosciences, HiFiBio, and Cell Signaling Technologies. H.Y.C. is a co-founder of Accent Therapeutics, Boundless Bio, and is an advisor for 10x Genomics, Arsenal Biosciences, and Spring Discovery., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
8. Large-Scale Topological Changes Restrain Malignant Progression in Colorectal Cancer.
- Author
-
Johnstone SE, Reyes A, Qi Y, Adriaens C, Hegazi E, Pelka K, Chen JH, Zou LS, Drier Y, Hecht V, Shoresh N, Selig MK, Lareau CA, Iyer S, Nguyen SC, Joyce EF, Hacohen N, Irizarry RA, Zhang B, Aryee MJ, and Bernstein BE
- Subjects
- Cell Division, Cellular Senescence genetics, Chromatin Immunoprecipitation Sequencing, Chromosomes genetics, Cohort Studies, Colorectal Neoplasms mortality, Colorectal Neoplasms pathology, Computational Biology, Epigenomics, HCT116 Cells, Humans, In Situ Hybridization, Fluorescence, Microscopy, Electron, Transmission, Molecular Dynamics Simulation, RNA-Seq, Spatial Analysis, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Chromatin metabolism, Chromosomes metabolism, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, DNA Methylation genetics, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic genetics
- Abstract
Widespread changes to DNA methylation and chromatin are well documented in cancer, but the fate of higher-order chromosomal structure remains obscure. Here we integrated topological maps for colon tumors and normal colons with epigenetic, transcriptional, and imaging data to characterize alterations to chromatin loops, topologically associated domains, and large-scale compartments. We found that spatial partitioning of the open and closed genome compartments is profoundly compromised in tumors. This reorganization is accompanied by compartment-specific hypomethylation and chromatin changes. Additionally, we identify a compartment at the interface between the canonical A and B compartments that is reorganized in tumors. Remarkably, similar shifts were evident in non-malignant cells that have accumulated excess divisions. Our analyses suggest that these topological changes repress stemness and invasion programs while inducing anti-tumor immunity genes and may therefore restrain malignant progression. Our findings call into question the conventional view that tumor-associated epigenomic alterations are primarily oncogenic., Competing Interests: Declaration of Interests N.H. is an equity holder of BioNTech and a consultant for Related Sciences. M.J.A. declares outside interest in Excelsior Genomics. B.E.B. declares outside interests in Fulcrum Therapeutics, 1CellBio, HiFiBio, Arsenal Biosciences, Cell Signaling Technologies, BioMillenia, and Nohla Therapeutics., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
9. The long noncoding RNA NEAT1_1 is seemingly dispensable for normal tissue homeostasis and cancer cell growth.
- Author
-
Adriaens C, Rambow F, Bervoets G, Silla T, Mito M, Chiba T, Asahara H, Hirose T, Nakagawa S, Jensen TH, and Marine JC
- Subjects
- Alternative Splicing, Animals, Cell Cycle drug effects, Cell Line, Cell Proliferation, Exosomes metabolism, Gene Knockout Techniques, Homeostasis, Humans, In Situ Hybridization, Fluorescence, Mice, Neoplasms metabolism, Neoplasms pathology, RNA Stability, Stress, Physiological genetics, Transcriptome, Gene Expression Regulation, Neoplastic, Neoplasms genetics, RNA, Long Noncoding genetics
- Abstract
NEAT1 is one of the most studied lncRNAs, in part because its silencing in mice causes defects in mammary gland development and corpus luteum formation and protects them from skin cancer development. Moreover, depleting NEAT1 in established cancer cell lines reduces growth and sensitizes cells to DNA damaging agents. However, NEAT1 produces two isoforms and because the short isoform, NEAT1_1 , completely overlaps the 5' part of the long NEAT1_2 isoform; the respective contributions of each of the isoforms to these phenotypes has remained unclear. Whereas NEAT1_1 is highly expressed in most tissues, NEAT1_2 is the central architectural component of paraspeckles, which are nuclear bodies that assemble in specific tissues and cells exposed to various forms of stress. Using dual RNA-FISH to detect both NEAT1_1 outside of the paraspeckles and NEAT1_2/NEAT1 inside this nuclear body, we report herein that NEAT1_1 levels are dynamically regulated during the cell cycle and targeted for degradation by the nuclear RNA exosome. Unexpectedly, however, cancer cells engineered to lack NEAT1_1 , but not NEAT1_2 , do not exhibit cell cycle defects. Moreover, Neat1_1 -specific knockout mice do not exhibit the phenotypes observed in Neat1 -deficient mice. We propose that NEAT1 functions are mainly, if not exclusively, attributable to NEAT1_2 and, by extension, to paraspeckles., (© 2019 Adriaens et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2019
- Full Text
- View/download PDF
10. Blank spots on the map: some current questions on nuclear organization and genome architecture.
- Author
-
Adriaens C, Serebryannyy LA, Feric M, Schibler A, Meaburn KJ, Kubben N, Trzaskoma P, Shachar S, Vidak S, Finn EH, Sood V, Pegoraro G, and Misteli T
- Subjects
- Animals, Humans, Nuclear Lamina genetics, Nuclear Lamina metabolism, Cell Nucleus genetics, Cell Nucleus metabolism
- Abstract
The past decades have provided remarkable insights into how the eukaryotic cell nucleus and the genome within it are organized. The combined use of imaging, biochemistry and molecular biology approaches has revealed several basic principles of nuclear architecture and function, including the existence of chromatin domains of various sizes, the presence of a large number of non-membranous intranuclear bodies, non-random positioning of genes and chromosomes in 3D space, and a prominent role of the nuclear lamina in organizing genomes. Despite this tremendous progress in elucidating the biological properties of the cell nucleus, many questions remain. Here, we highlight some of the key open areas of investigation in the field of nuclear organization and genome architecture with a particular focus on the mechanisms and principles of higher-order genome organization, the emerging role of liquid phase separation in cellular organization, and the functional role of the nuclear lamina in physiological processes.
- Published
- 2018
- Full Text
- View/download PDF
11. Toward Minimal Residual Disease-Directed Therapy in Melanoma.
- Author
-
Rambow F, Rogiers A, Marin-Bejar O, Aibar S, Femel J, Dewaele M, Karras P, Brown D, Chang YH, Debiec-Rychter M, Adriaens C, Radaelli E, Wolter P, Bechter O, Dummer R, Levesque M, Piris A, Frederick DT, Boland G, Flaherty KT, van den Oord J, Voet T, Aerts S, Lund AW, and Marine JC
- Subjects
- Animals, Biomarkers, Tumor, Drug Resistance, Neoplasm drug effects, Female, Humans, MAP Kinase Kinase 1 antagonists & inhibitors, MAP Kinase Kinase 1 genetics, Male, Melanoma metabolism, Melanoma pathology, Mice, SCID, Mutation, Neoplasm, Residual metabolism, Neoplasm, Residual pathology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Neural Stem Cells metabolism, Neural Stem Cells pathology, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf genetics, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Gene Expression Regulation, Neoplastic drug effects, Melanoma drug therapy, Neoplasm, Residual drug therapy, Neoplastic Stem Cells drug effects, Neural Stem Cells drug effects, Protein Kinase Inhibitors pharmacology, Retinoid X Receptor gamma antagonists & inhibitors
- Abstract
Many patients with advanced cancers achieve dramatic responses to a panoply of therapeutics yet retain minimal residual disease (MRD), which ultimately results in relapse. To gain insights into the biology of MRD, we applied single-cell RNA sequencing to malignant cells isolated from BRAF mutant patient-derived xenograft melanoma cohorts exposed to concurrent RAF/MEK-inhibition. We identified distinct drug-tolerant transcriptional states, varying combinations of which co-occurred within MRDs from PDXs and biopsies of patients on treatment. One of these exhibited a neural crest stem cell (NCSC) transcriptional program largely driven by the nuclear receptor RXRG. An RXR antagonist mitigated accumulation of NCSCs in MRD and delayed the development of resistance. These data identify NCSCs as key drivers of resistance and illustrate the therapeutic potential of MRD-directed therapy. They also highlight how gene regulatory network architecture reprogramming may be therapeutically exploited to limit cellular heterogeneity, a key driver of disease progression and therapy resistance., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
12. NEAT1-containing paraspeckles: Central hubs in stress response and tumor formation.
- Author
-
Adriaens C and Marine JC
- Subjects
- DNA Damage, Humans, Models, Biological, RNA, Long Noncoding genetics, Intranuclear Inclusion Bodies metabolism, Neoplasms metabolism, RNA, Long Noncoding metabolism, Stress, Physiological
- Published
- 2017
- Full Text
- View/download PDF
13. p53 induces formation of NEAT1 lncRNA-containing paraspeckles that modulate replication stress response and chemosensitivity.
- Author
-
Adriaens C, Standaert L, Barra J, Latil M, Verfaillie A, Kalev P, Boeckx B, Wijnhoven PW, Radaelli E, Vermi W, Leucci E, Lapouge G, Beck B, van den Oord J, Nakagawa S, Hirose T, Sablina AA, Lambrechts D, Aerts S, Blanpain C, and Marine JC
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Cell Line, Tumor, DNA Damage, Flow Cytometry, Humans, Immunoblotting, Immunohistochemistry, In Situ Hybridization, Fluorescence, Kaplan-Meier Estimate, MCF-7 Cells, Mice, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Precancerous Conditions genetics, Proportional Hazards Models, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Skin Neoplasms genetics, Survival Analysis, Carcinogenesis genetics, Gene Expression Regulation, Neoplastic genetics, RNA, Long Noncoding genetics, Tumor Suppressor Protein p53 genetics
- Abstract
In a search for mediators of the p53 tumor suppressor pathway, which induces pleiotropic and often antagonistic cellular responses, we identified the long noncoding RNA (lncRNA) NEAT1. NEAT1 is an essential architectural component of paraspeckle nuclear bodies, whose pathophysiological relevance remains unclear. Activation of p53, pharmacologically or by oncogene-induced replication stress, stimulated the formation of paraspeckles in mouse and human cells. Silencing Neat1 expression in mice, which prevents paraspeckle formation, sensitized preneoplastic cells to DNA-damage-induced cell death and impaired skin tumorigenesis. We provide mechanistic evidence that NEAT1 promotes ATR signaling in response to replication stress and is thereby engaged in a negative feedback loop that attenuates oncogene-dependent activation of p53. NEAT1 targeting in established human cancer cell lines induced synthetic lethality with genotoxic chemotherapeutics, including PARP inhibitors, and nongenotoxic activation of p53. This study establishes a key genetic link between NEAT1 paraspeckles, p53 biology and tumorigenesis and identifies NEAT1 as a promising target to enhance sensitivity of cancer cells to both chemotherapy and p53 reactivation therapy.
- Published
- 2016
- Full Text
- View/download PDF
14. The long noncoding RNA Neat1 is required for mammary gland development and lactation.
- Author
-
Standaert L, Adriaens C, Radaelli E, Van Keymeulen A, Blanpain C, Hirose T, Nakagawa S, and Marine JC
- Subjects
- Animals, Cell Proliferation genetics, Female, Gene Expression Regulation, Developmental, Humans, Intranuclear Inclusion Bodies genetics, Intranuclear Inclusion Bodies metabolism, Mammary Glands, Animal metabolism, Mice, Nuclear Proteins genetics, RNA, Long Noncoding metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Lactation genetics, Mammary Glands, Animal growth & development, Morphogenesis genetics, RNA, Long Noncoding genetics
- Abstract
The lncRNA Neat1 is an essential architectural component of paraspeckle nuclear bodies. Although cell-based studies identified Neat1-paraspeckles as key regulators of gene expression through retention of hyperdited mRNAs and/or transcription factors, it is unclear under which specific physiological conditions paraspeckles are formed in vivo and whether they have any biological relevance. Herein, we show that paraspeckles are assembled in luminal epithelial cells during mammary gland development. Importantly, genetic ablation of Neat1 results in aberrant mammary gland morphogenesis and lactation defects. We provide evidence that the lactation defect is caused by a decreased ability of Neat1-mutant cells to sustain high rates of proliferation during lobular-alveolar development. This study is the first to assign an important biological function to the lncRNA Neat1 and to link it to the presence of paraspeckles nuclear bodies in vivo., (© 2014 Standaert et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.