Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Cell Genom ; 3(7): 100321, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37492096

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.

3.
Cell ; 184(9): 2503-2519.e17, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33838111

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.


Subject(s)
CRISPR-Cas Systems , Cellular Reprogramming , Epigenesis, Genetic , Epigenome , Gene Editing , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Cell Differentiation , CpG Islands , DNA Methylation , Gene Silencing , Histone Code , Humans , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Protein Processing, Post-Translational
4.
Cell ; 182(6): 1474-1489.e23, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32841603

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.


Subject(s)
Chromatin/metabolism , Chromosomes/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Cell Division , Cellular Senescence/genetics , Chromatin Immunoprecipitation Sequencing , Chromosomes/genetics , Cohort Studies , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Computational Biology , DNA Methylation/genetics , 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
5.
RNA ; 25(12): 1681-1695, 2019 12.
Article in English | MEDLINE | ID: mdl-31551298

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.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , RNA, Long Noncoding/genetics , 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
6.
Histochem Cell Biol ; 150(6): 579-592, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30238154

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.


Subject(s)
Cell Nucleus/genetics , Cell Nucleus/metabolism , Animals , Humans , Nuclear Lamina/genetics , Nuclear Lamina/metabolism
7.
Cell ; 174(4): 843-855.e19, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30017245

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.


Subject(s)
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 , 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
9.
Nat Med ; 22(8): 861-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27376578

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.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/genetics , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/genetics , 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
10.
RNA ; 20(12): 1844-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25316907

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.


Subject(s)
Lactation/genetics , Mammary Glands, Animal/growth & development , Morphogenesis/genetics , RNA, Long Noncoding/genetics , 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
SELECTION OF CITATIONS
SEARCH DETAIL
...