1
|
Park BJ, Hua S, Casler KD, Cefaloni E, Ayers MC, Lake RF, Murphy KE, Vertino PM, O'Connell MR, Murphy PJ. CUT&Tag Identifies Repetitive Genomic Loci that are Excluded from ChIP Assays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636299. [PMID: 39974916 PMCID: PMC11838576 DOI: 10.1101/2025.02.03.636299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Determining the genomic localization of chromatin features is an essential aspect of investigating gene expression control, and ChIP-Seq has long been the gold standard technique for interrogating chromatin landscapes. Recently, the development of alternative methods, such as CUT&Tag, have provided researchers with alternative strategies that eliminate the need for chromatin purification, and allow for in situ investigation of histone modifications and chromatin bound factors. Mindful of technical differences, we set out to investigate whether distinct chromatin modifications were equally compatible with these different chromatin interrogation techniques. We found that ChIP-Seq and CUT&Tag performed similarly for modifications known to reside at gene regulatory regions, such as promoters and enhancers, but major differences were observed when we assessed enrichment over heterochromatin-associated loci. Unlike ChIP-Seq, CUT&Tag detects robust levels of H3K9me3 at a substantial number of repetitive elements, with especially high sensitivity over evolutionarily young retrotransposons. IAPEz-int elements for example, exhibited underrepresentation in mouse ChIP-Seq datasets but strong enrichment using CUT&Tag. Additionally, we identified several euchromatin-associated proteins that co-purify with repetitive loci and are similarly depleted when applying ChIP-based methods. This study reveals that our current knowledge of chromatin states across the heterochromatin portions of the mammalian genome is extensively incomplete, largely due to limitations of ChIP-Seq. We also demonstrate that newer in situ chromatin fragmentation-based techniques, such as CUT&Tag and CUT&RUN, are more suitable for studying chromatin modifications over repetitive elements and retrotransposons.
Collapse
Affiliation(s)
- Brandon J Park
- University of Rochester Medical Center, Wilmot Cancer Center, Department of Biomedical Genetics
- University of Rochester Medical Center, The Department of Biochemistry & Biophysics
| | - Shan Hua
- University of Rochester Medical Center, Wilmot Cancer Center, Department of Biomedical Genetics
- University of Rochester, Department of Biology
| | - Karli D Casler
- University of Rochester Medical Center, Wilmot Cancer Center, Department of Biomedical Genetics
- University of Rochester, Department of Biology
| | - Eric Cefaloni
- University of Rochester Medical Center, Wilmot Cancer Center, Department of Biomedical Genetics
- University of Rochester, Department of Biology
| | - Michael C Ayers
- University of Rochester Medical Center, Wilmot Cancer Center, Department of Biomedical Genetics
| | - Rahiim F Lake
- University of Rochester Medical Center, The Department of Biochemistry & Biophysics
| | - Kristin E Murphy
- University of Rochester Medical Center, Wilmot Cancer Center, Department of Biomedical Genetics
| | - Paula M Vertino
- University of Rochester Medical Center, Wilmot Cancer Center, Department of Biomedical Genetics
| | - Mitchell R O'Connell
- University of Rochester Medical Center, The Department of Biochemistry & Biophysics
| | - Patrick J Murphy
- University of Rochester Medical Center, Wilmot Cancer Center, Department of Biomedical Genetics
- University of Rochester, Department of Biology
- Cornell University, Department of Molecular Biology and Genetics
| |
Collapse
|
2
|
Sen S, Estève PO, Raman K, Beaulieu J, Chin H, Feehery G, Vishnu U, Xu SY, Samuelson J, Pradhan S. Distinct structural and functional heterochromatin partitioning of lamin B1 and lamin B2 revealed using genome-wide nicking enzyme epitope targeted DNA sequencing. Nucleic Acids Res 2025; 53:gkae1317. [PMID: 39817518 PMCID: PMC11736435 DOI: 10.1093/nar/gkae1317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 01/18/2025] Open
Abstract
Gene expression is regulated by chromatin DNA methylation and other features, including histone post-translational modifications (PTMs), chromatin remodelers and transcription factor occupancy. A complete understanding of gene regulation will require the mapping of these chromatin features in small cell number samples. Here we describe a novel genome-wide chromatin profiling technology, named as Nicking Enzyme Epitope targeted DNA sequencing (NEED-seq). NEED-seq offers antibody-targeted controlled nicking by Nt.CviPII-pGL fusion to study specific protein-DNA complexes in formaldehyde fixed cells, allowing for both visual and genomic resolution of epitope bound chromatin. When applied to nuclei, NEED-seq yielded genome-wide profile of chromatin-associated proteins and histone PTMs. Additionally, NEED-seq of lamin B1 and B2 demonstrated their association with heterochromatin. Lamin B1- and B2-associated domains (LAD) segregated to three different states, and states with stronger LAD correlated with heterochromatic marks. Hi-C analysis displayed A and B compartment with equal lamin B1 and B2 distribution, although methylated DNA remained high in B compartment. LAD clustering with Hi-C resulted in subcompartments, with lamin B1 and B2 partitioning to facultative and constitutive heterochromatin, respectively, and were associated with neuronal development. Thus, lamin B1 and B2 show structural and functional partitioning in mammalian nucleus.
Collapse
Affiliation(s)
- Sagnik Sen
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Pierre-Olivier Estève
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Karthikeyan Raman
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Julie Beaulieu
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Hang Gyeong Chin
- PMBBRC, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - George R Feehery
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | | | - Shuang-yong Xu
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - James C Samuelson
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Sriharsa Pradhan
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| |
Collapse
|
3
|
Kobayashi S, Nishiba S, Sato C, Toriumi K, Someya Y, Adachi N, Takeda S, Kurosawa A. Sulforaphane Induces Transient Reactive Oxygen Species-Mediated DNA Damage in HeLa Cells. Genes Cells 2025; 30:e13190. [PMID: 39727047 DOI: 10.1111/gtc.13190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Sulforaphane (SFN), an isothiocyanate found in plants of the Brassicaceae family, possesses antioxidant, apoptosis-inducing, and radiosensitizing effects. As one of the mechanisms of cytotoxicity by SFN, SFN has been suggested to be involved in the induction of DNA damage and inhibition of DNA repair. Recently, we reported on the potency of SFN in inducing single-ended double-strand breaks (DSBs) that are caused by the collision of replication forks with single-strand breaks (SSBs). However, the mechanism of SSB accumulation by SFN remains unclear. In this study, we examined the effect of SFN on SSB-inducing factors in HeLa cells. Although the inhibitory effect of SFN on DNA topoisomerase I was not observed, we found that the reduced form of glutathione (GSH; an antioxidant) level was decreased in an SFN concentration-dependent manner. Furthermore, the addition of ascorbic acid partially increased the viability of SFN-treated HeLa cells. We subsequently observed that poly(ADP-ribose) accumulated in SFN-treated HeLa cells, which occurs during early SSB repair. Collectively, these findings suggest that SFN may transiently induce SSBs via reactive oxygen species in HeLa cells.
Collapse
Affiliation(s)
- Sakine Kobayashi
- School of Science and Technology, Gunma University, Kiryu, Japan
| | - Seiya Nishiba
- School of Science and Technology, Gunma University, Kiryu, Japan
| | - Chinatsu Sato
- School of Science and Technology, Gunma University, Kiryu, Japan
| | - Kazuya Toriumi
- School of Science and Technology, Gunma University, Kiryu, Japan
| | - Yuduki Someya
- School of Science and Technology, Gunma University, Kiryu, Japan
| | - Noritaka Adachi
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Japan
| | - Shigeki Takeda
- School of Science and Technology, Gunma University, Kiryu, Japan
| | - Aya Kurosawa
- School of Science and Technology, Gunma University, Kiryu, Japan
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Japan
- Gunma University Center for Food and Science and Wellness, Gunma University, Kiryu, Japan
| |
Collapse
|
4
|
Tan Y, Yao L, Gamliel A, Nair SJ, Taylor H, Ohgi K, Aggarwal AK, Rosenfeld MG. Signal-induced enhancer activation requires Ku70 to read topoisomerase1-DNA covalent complexes. Nat Struct Mol Biol 2023; 30:148-158. [PMID: 36747093 PMCID: PMC9935399 DOI: 10.1038/s41594-022-00883-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 10/27/2022] [Indexed: 02/08/2023]
Abstract
Enhancer activation serves as the main mechanism regulating signal-dependent transcriptional programs, ensuring cellular plasticity, yet central questions persist regarding their mechanism of activation. Here, by successfully mapping topoisomerase I-DNA covalent complexes genome-wide, we find that most, if not all, acutely activated enhancers, including those induced by 17β-estradiol, dihydrotestosterone, tumor necrosis factor alpha and neuronal depolarization, are hotspots for topoisomerase I-DNA covalent complexes, functioning as epigenomic signatures read by the classic DNA damage sensor protein, Ku70. Ku70 in turn nucleates a heterochromatin protein 1 gamma (HP1γ)-mediator subunit Med26 complex to facilitate acute, but not chronic, transcriptional activation programs. Together, our data uncover a broad, unappreciated transcriptional code, required for most, if not all, acute signal-dependent enhancer activation events in both mitotic and postmitotic cells.
Collapse
Affiliation(s)
- Yuliang Tan
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Lu Yao
- Familial and Hereditary Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Amir Gamliel
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sreejith J Nair
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Oncology Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| | - Havilah Taylor
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kenny Ohgi
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Aneel K Aggarwal
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael G Rosenfeld
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
5
|
PARP1 Activation Controls Stress Granule Assembly after Oxidative Stress and DNA Damage. Cells 2022; 11:cells11233932. [PMID: 36497190 PMCID: PMC9740212 DOI: 10.3390/cells11233932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
DNA damage causes PARP1 activation in the nucleus to set up the machinery responsible for the DNA damage response. Here, we report that, in contrast to cytoplasmic PARPs, the synthesis of poly(ADP-ribose) by PARP1 opposes the formation of cytoplasmic mRNA-rich granules after arsenite exposure by reducing polysome dissociation. However, when mRNA-rich granules are pre-formed, whether in the cytoplasm or nucleus, PARP1 activation positively regulates their assembly, though without additional recruitment of poly(ADP-ribose) in stress granules. In addition, PARP1 promotes the formation of TDP-43- and FUS-rich granules in the cytoplasm, two RNA-binding proteins which form neuronal cytoplasmic inclusions observed in certain neurodegenerative diseases such as amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Together, the results therefore reveal a dual role of PARP1 activation which, on the one hand, prevents the early stage of stress granule assembly and, on the other hand, enables the persistence of cytoplasmic mRNA-rich granules in cells which may be detrimental in aging neurons.
Collapse
|
6
|
Angelov D, Boopathi R, Lone IN, Menoni H, Dimitrov S, Cadet J. Capturing Protein-Nucleic Acid Interactions by High-Intensity Laser-Induced Covalent Crosslinking. Photochem Photobiol 2022; 99:296-312. [PMID: 35997098 DOI: 10.1111/php.13699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022]
Abstract
Interactions of DNA with structural proteins such as histones, regulatory proteins, and enzymes play a crucial role in major cellular processes such as transcription, replication and repair. The in vivo mapping and characterization of the binding sites of the involved biomolecules are of primary importance for a better understanding of genomic deployment that is implicated in tissue and developmental stage-specific gene expression regulation. The most powerful and commonly used approach to date is immunoprecipitation of chemically cross-linked chromatin (XChIP) coupled with sequencing analysis (ChIP-seq). While the resolution and the sensitivity of the high-throughput sequencing techniques have been constantly improved little progress has been achieved in the crosslinking step. Because of its low efficiency the use of the conventional UVC lamps remains very limited while the formaldehyde method was established as the "gold standard" crosslinking agent. Efficient biphotonic crosslinking of directly interacting nucleic acid-protein complexes by a single short UV laser pulse has been introduced as an innovative technique for overcoming limitations of conventionally used chemical and photochemical approaches. In this survey, the main available methods including the laser approach are critically reviewed for their ability to generate DNA-protein crosslinks in vitro model systems and cells.
Collapse
Affiliation(s)
- Dimitar Angelov
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS, Laboratoire de Biologie et de Modélisation de la Cellule LBMC, CNRS UMR 5239, 46 Allée d'Italie, 69007, Lyon, France.,Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Balçova, Izmir 35330, Turkey
| | - Ramachandran Boopathi
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS, Laboratoire de Biologie et de Modélisation de la Cellule LBMC, CNRS UMR 5239, 46 Allée d'Italie, 69007, Lyon, France.,Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), 38000, Grenoble, France
| | - Imtiaz Nisar Lone
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Balçova, Izmir 35330, Turkey
| | - Hervé Menoni
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Stefan Dimitrov
- Université Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences (IAB), Site Santé - Allée des Alpes, 38700, La Tronche, France
| | - Jean Cadet
- Département de Médecine nucléaire et Radiobiologie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| |
Collapse
|
7
|
Li X, Zhou J, Zhao W, Wen Q, Wang W, Peng H, Gao Y, Bouchonville KJ, Offer SM, Chan K, Wang Z, Li N, Gan H. Defining Proximity Proteomics of Histone Modifications by Antibody-mediated Protein A-APEX2 Labeling. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 20:87-100. [PMID: 34555496 PMCID: PMC9510856 DOI: 10.1016/j.gpb.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/02/2022]
Abstract
Proximity labeling catalyzed by promiscuous enzymes, such as APEX2, has emerged as a powerful approach to characterize multiprotein complexes and protein–protein interactions. However, current methods depend on the expression of exogenous fusion proteins and cannot be applied to identify proteins surrounding post-translationally modified proteins. To address this limitation, we developed a new method to label proximal proteins of interest by antibody-mediated protein A-ascorbate peroxidase 2 (pA-APEX2) labeling (AMAPEX). In this method, a modified protein is bound in situ by a specific antibody, which then tethers a pA-APEX2 fusion protein. Activation of APEX2 labels the nearby proteins with biotin; the biotinylated proteins are then purified using streptavidin beads and identified by mass spectrometry. We demonstrated the utility of this approach by profiling the proximal proteins of histone modifications including H3K27me3, H3K9me3, H3K4me3, H4K5ac, and H4K12ac, as well as verifying the co-localization of these identified proteins with bait proteins by published ChIP-seq analysis and nucleosome immunoprecipitation. Overall, AMAPEX is an efficient method to identify proteins that are proximal to modified histones.
Collapse
Affiliation(s)
- Xinran Li
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiaqi Zhou
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenjuan Zhao
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qing Wen
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Weijie Wang
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Huipai Peng
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuan Gao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kelly J Bouchonville
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Steven M Offer
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Mayo Clinic Cancer Center, Rochester, MN 55905, USA
| | - Kuiming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region 999077, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518172, China
| | - Zhiquan Wang
- Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | - Nan Li
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Haiyun Gan
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
8
|
Poly(ADP-ribose) Polymerase 1 (PARP1) restrains MyoD-dependent gene expression during muscle differentiation. Sci Rep 2020; 10:15086. [PMID: 32934320 PMCID: PMC7493885 DOI: 10.1038/s41598-020-72155-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
The myogenic factor MyoD regulates skeletal muscle differentiation by interacting with a variety of chromatin-modifying complexes. Although MyoD can induce and maintain chromatin accessibility at its target genes, its binding and trans-activation ability can be limited by some types of not fully characterized epigenetic constraints. In this work we analysed the role of PARP1 in regulating MyoD-dependent gene expression. PARP1 is a chromatin-associated enzyme, playing a well recognized role in DNA repair and that is implicated in transcriptional regulation. PARP1 affects gene expression through multiple mechanisms, often involving the Poly(ADP-ribosyl)ation of chromatin proteins. In line with PARP1 down-regulation during differentiation, we observed that PARP1 depletion boosts the up-regulation of MyoD targets, such as p57, myogenin, Mef2C and p21, while its re-expression reverts this effect. We also found that PARP1 interacts with some MyoD-binding regions and that its presence, independently of the enzymatic activity, interferes with MyoD recruitment and gene induction. We finally suggest a relationship between the binding of PARP1 and the loss of the activating histone modification H3K4me3 at MyoD-binding regions. This work highlights not only a novel player in the epigenetic control of myogenesis, but also a repressive and catalytic-independent mechanisms by which PARP1 regulates transcription.
Collapse
|
9
|
Ummethum H, Hamperl S. Proximity Labeling Techniques to Study Chromatin. Front Genet 2020; 11:450. [PMID: 32477404 PMCID: PMC7235407 DOI: 10.3389/fgene.2020.00450] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/14/2020] [Indexed: 12/19/2022] Open
Abstract
Mammals contain over 200 different cell types, yet nearly all have the same genomic DNA sequence. It is a key question in biology how the genetic instructions in DNA are selectively interpreted by cells to specify various transcriptional programs and therefore cellular identity. The structural and functional organization of chromatin governs the transcriptional state of individual genes. To understand how genomic loci adopt different levels of gene expression, it is critical to characterize all local chromatin factors as well as long-range interactions in the 3D nuclear compartment. Much of our current knowledge regarding protein interactions in a chromatin context is based on affinity purification of chromatin components coupled to mass spectrometry (AP-MS). AP-MS has been invaluable to map strong protein-protein interactions in the nucleus. However, the interaction is detected after cell lysis and biochemical enrichment, allowing for loss or gain of false positive or negative interaction partners. Recently, proximity-dependent labeling methods have emerged as powerful tools for studying chromatin in its native context. These methods take advantage of engineered enzymes that are fused to a chromatin factor of interest and can directly label all factors in proximity. Subsequent pull-down assays followed by mass spectrometry or sequencing approaches provide a comprehensive snapshot of the proximal chromatin interactome. By combining this method with dCas9, this approach can also be extended to study chromatin at specific genomic loci. Here, we review and compare current proximity-labeling approaches available for studying chromatin, with a particular focus on new emerging technologies that can provide important insights into the transcriptional and chromatin interaction networks essential for cellular identity.
Collapse
Affiliation(s)
- Henning Ummethum
- Chromosome Dynamics and Genome Stability, Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| | - Stephan Hamperl
- Chromosome Dynamics and Genome Stability, Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
10
|
Stapleton K, Das S, Reddy MA, Leung A, Amaram V, Lanting L, Chen Z, Zhang L, Palanivel R, Deiuliis JA, Natarajan R. Novel Long Noncoding RNA, Macrophage Inflammation-Suppressing Transcript ( MIST), Regulates Macrophage Activation During Obesity. Arterioscler Thromb Vasc Biol 2020; 40:914-928. [PMID: 32078363 PMCID: PMC7098442 DOI: 10.1161/atvbaha.119.313359] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Supplemental Digital Content is available in the text. Objective: Systemic low-grade inflammation associated with obesity and metabolic syndrome is a strong risk factor for the development of diabetes mellitus and associated cardiovascular complications. This inflammatory state is caused by release of proinflammatory cytokines by macrophages, especially in adipose tissue. Long noncoding RNAs regulate macrophage activation and inflammatory gene networks, but their role in macrophage dysfunction during diet-induced obesity has been largely unexplored. Approach and Results: We sequenced total RNA from peritoneal macrophages isolated from mice fed either high-fat diet or standard diet and performed de novo transcriptome assembly to identify novel differentially expressed mRNAs and long noncoding RNAs. A top candidate long noncoding RNA, macrophage inflammation-suppressing transcript (Mist), was downregulated in both peritoneal macrophages and adipose tissue macrophages from high-fat diet–fed mice. GapmeR-mediated Mist knockdown in vitro and in vivo upregulated expression of genes associated with immune response and inflammation and increased modified LDL (low-density lipoprotein) uptake in macrophages. Conversely, Mist overexpression decreased basal and LPS (lipopolysaccharide)-induced expression of inflammatory response genes and decreased modified LDL uptake. RNA-pull down coupled with mass spectrometry showed that Mist interacts with PARP1 (poly [ADP]-ribose polymerase-1). Disruption of this RNA-protein interaction increased PARP1 recruitment and chromatin PARylation at promoters of inflammatory genes, resulting in increased gene expression. Furthermore, human orthologous MIST was also downregulated by proinflammatory stimuli, and its expression in human adipose tissue macrophages inversely correlated with obesity and insulin resistance. Conclusions: Mist is a novel protective long noncoding RNA, and its loss during obesity contributes to metabolic dysfunction and proinflammatory phenotype of macrophages via epigenetic mechanisms.
Collapse
Affiliation(s)
- Kenneth Stapleton
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA.,Irell and Manella Graduate School of Biological Sciences (K.S., V.A., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Sadhan Das
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Marpadga A Reddy
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Amy Leung
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Vishnu Amaram
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA.,Irell and Manella Graduate School of Biological Sciences (K.S., V.A., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Linda Lanting
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Zhuo Chen
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Lingxiao Zhang
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| | - Rengasamy Palanivel
- Cardiovascular Research Institute of the Case Western Reserve University, Cleveland, OH (R.P., J.A.D.)
| | - Jeffrey A Deiuliis
- Cardiovascular Research Institute of the Case Western Reserve University, Cleveland, OH (R.P., J.A.D.)
| | - Rama Natarajan
- From the Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute (K.S, S.D., M.A.R., A.L., V.A., L.L., Z.C., L.Z., R.N.), Beckman Research Institute of City of Hope, Duarte, CA.,Irell and Manella Graduate School of Biological Sciences (K.S., V.A., R.N.), Beckman Research Institute of City of Hope, Duarte, CA
| |
Collapse
|
11
|
Krastev DB, Pettitt SJ, Campbell J, Song F, Tanos BE, Stoynov SS, Ashworth A, Lord CJ. Coupling bimolecular PARylation biosensors with genetic screens to identify PARylation targets. Nat Commun 2018; 9:2016. [PMID: 29789535 PMCID: PMC5964205 DOI: 10.1038/s41467-018-04466-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/01/2018] [Indexed: 12/26/2022] Open
Abstract
Poly (ADP-ribose)ylation is a dynamic protein modification that regulates multiple cellular processes. Here, we describe a system for identifying and characterizing PARylation events that exploits the ability of a PBZ (PAR-binding zinc finger) protein domain to bind PAR with high-affinity. By linking PBZ domains to bimolecular fluorescent complementation biosensors, we developed fluorescent PAR biosensors that allow the detection of temporal and spatial PARylation events in live cells. Exploiting transposon-mediated recombination, we integrate the PAR biosensor en masse into thousands of protein coding genes in living cells. Using these PAR-biosensor "tagged" cells in a genetic screen we carry out a large-scale identification of PARylation targets. This identifies CTIF (CBP80/CBP20-dependent translation initiation factor) as a novel PARylation target of the tankyrase enzymes in the centrosomal region of cells, which plays a role in the distribution of the centrosomal satellites.
Collapse
Affiliation(s)
- Dragomir B Krastev
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Stephen J Pettitt
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - James Campbell
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Feifei Song
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Barbara E Tanos
- The Cancer Therapeutics Division, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Stoyno S Stoynov
- The Institute of Molecular Biology, Bulgarian Academy of Sciences, 1113, Sofia, Bulgaria
| | - Alan Ashworth
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
- UCSF Helen Diller Family Comprehensive Cancer Center, 1450 3rd Street, San Francisco, CA, 94158, USA.
| | - Christopher J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
| |
Collapse
|
12
|
Michelena J, Altmeyer M. Cell Cycle Resolved Measurements of Poly(ADP-Ribose) Formation and DNA Damage Signaling by Quantitative Image-Based Cytometry. Methods Mol Biol 2018; 1608:57-68. [PMID: 28695503 DOI: 10.1007/978-1-4939-6993-7_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Formation of poly(ADP-ribose) (PAR) marks intracellular stress signaling and is notably induced upon DNA damage. PAR polymerases (PARPs) catalyze PAR synthesis upon genotoxic stress and thereby recruit multiple proteins to damaged chromatin. PAR induction is transient and antagonized by the action of PAR glycohydrolase (PARG). Given that poly(ADP-ribosyl)ation (PARylation) is involved in genome integrity maintenance and other vital cellular functions, but also in light of the recent approval of PARP inhibitors for cancer treatments, reliable measurements of intracellular PAR formation have gained importance. Here we provide a detailed protocol for PAR measurements by quantitative image-based cytometry. This technique combines the high spatial resolution of single-cell microscopy with the advantages of cell population measurements through automated high-content imaging. Such upscaling of immunofluorescence-based PAR detection not only increases the robustness of the measurements through averaging across large cell populations but also allows for the discrimination of subpopulations and thus enables multivariate measurements of PAR levels and DNA damage signaling. We illustrate how this technique can be used to assess the dynamics of the cellular response to oxidative damage as well as to PARP inhibitor-induced genotoxicity in a cell cycle resolved manner. Due to the possibility to use any automated microscope for quantitative image-based cytometry, the presented method has widespread applicability in the area of PARP biology and beyond.
Collapse
Affiliation(s)
- Jone Michelena
- Department of Molecular Mechanisms of Disease, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Matthias Altmeyer
- Department of Molecular Mechanisms of Disease, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| |
Collapse
|
13
|
Imre L, Simándi Z, Horváth A, Fenyőfalvi G, Nánási P, Niaki EF, Hegedüs É, Bacsó Z, Weyemi U, Mauser R, Ausio J, Jeltsch A, Bonner W, Nagy L, Kimura H, Szabó G. Nucleosome stability measured in situ by automated quantitative imaging. Sci Rep 2017; 7:12734. [PMID: 28986581 PMCID: PMC5630628 DOI: 10.1038/s41598-017-12608-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023] Open
Abstract
Current approaches have limitations in providing insight into the functional properties of particular nucleosomes in their native molecular environment. Here we describe a simple and powerful method involving elution of histones using intercalators or salt, to assess stability features dependent on DNA superhelicity and relying mainly on electrostatic interactions, respectively, and measurement of the fraction of histones remaining chromatin-bound in the individual nuclei using histone type- or posttranslational modification- (PTM-) specific antibodies and automated, quantitative imaging. The method has been validated in H3K4me3 ChIP-seq experiments, by the quantitative assessment of chromatin loop relaxation required for nucleosomal destabilization, and by comparative analyses of the intercalator and salt induced release from the nucleosomes of different histones. The accuracy of the assay allowed us to observe examples of strict association between nucleosome stability and PTMs across cell types, differentiation state and throughout the cell-cycle in close to native chromatin context, and resolve ambiguities regarding the destabilizing effect of H2A.X phosphorylation. The advantages of the in situ measuring scenario are demonstrated via the marked effect of DNA nicking on histone eviction that underscores the powerful potential of topological relaxation in the epigenetic regulation of DNA accessibility.
Collapse
Affiliation(s)
- László Imre
- Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | - Zoltán Simándi
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, H-4032, Hungary.,Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Attila Horváth
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | - György Fenyőfalvi
- Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | - Péter Nánási
- Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | - Erfaneh Firouzi Niaki
- Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | - Éva Hegedüs
- Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | - Zsolt Bacsó
- Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | - Urbain Weyemi
- Center for Cancer Research National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Rebekka Mauser
- Institute of Biochemistry, Stuttgart University, Stuttgart, Germany
| | - Juan Ausio
- University of Victoria, Department of Biochemistry, Victoria, BC, V8W 3P6, Canada
| | - Albert Jeltsch
- Institute of Biochemistry, Stuttgart University, Stuttgart, Germany
| | - William Bonner
- Center for Cancer Research National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - László Nagy
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, H-4032, Hungary.,Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA.,MTA-DE "Lendulet" Immunogenomics Research Group, University of Debrecen, Debrecen, Hungary
| | - Hiroshi Kimura
- Cell Biology Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Gábor Szabó
- Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, H-4032, Hungary.
| |
Collapse
|
14
|
ADP-ribose-specific chromatin-affinity purification for investigating genome-wide or locus-specific chromatin ADP-ribosylation. Nat Protoc 2017; 12:1951-1961. [PMID: 28837130 DOI: 10.1038/nprot.2017.072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Protein ADP-ribosylation is a structurally heterogeneous post-translational modification (PTM) that influences the physicochemical and biological properties of the modified protein. ADP-ribosylation of chromatin changes its structural properties, thereby regulating important nuclear functions. A lack of suitable antibodies for chromatin immunoprecipitation (ChIP) has so far prevented a comprehensive analysis of DNA-associated protein ADP-ribosylation. To analyze chromatin ADP-ribosylation, we recently developed a novel ADP-ribose-specific chromatin-affinity purification (ADPr-ChAP) methodology that uses the recently identified ADP-ribose-binding domains RNF146 WWE and Af1521. In this protocol, we describe how to use this robust and versatile method for genome-wide and loci-specific localization of chromatin ADP-ribosylation. ADPr-ChAP enables bioinformatic comparisons of ADP-ribosylation with other chromatin modifications and is useful for understanding how ADP-ribosylation regulates biologically important cellular processes. ADPr-ChAP takes 1 week and requires standard skills in molecular biology and biochemistry. Although not covered in detail here, this technique can also be combined with conventional ChIP or DNA analysis to define the histone marks specifically associated with the ADP-ribosylated chromatin fractions and dissect the molecular mechanism and functional role of chromatin ADP-ribosylation.
Collapse
|
15
|
Halász L, Karányi Z, Boros-Oláh B, Kuik-Rózsa T, Sipos É, Nagy É, Mosolygó-L Á, Mázló A, Rajnavölgyi É, Halmos G, Székvölgyi L. RNA-DNA hybrid (R-loop) immunoprecipitation mapping: an analytical workflow to evaluate inherent biases. Genome Res 2017; 27:1063-1073. [PMID: 28341774 PMCID: PMC5453320 DOI: 10.1101/gr.219394.116] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/23/2017] [Indexed: 12/16/2022]
Abstract
The impact of R-loops on the physiology and pathology of chromosomes has been demonstrated extensively by chromatin biology research. The progress in this field has been driven by technological advancement of R-loop mapping methods that largely relied on a single approach, DNA-RNA immunoprecipitation (DRIP). Most of the DRIP protocols use the experimental design that was developed by a few laboratories, without paying attention to the potential caveats that might affect the outcome of RNA-DNA hybrid mapping. To assess the accuracy and utility of this technology, we pursued an analytical approach to estimate inherent biases and errors in the DRIP protocol. By performing DRIP-sequencing, qPCR, and receiver operator characteristic (ROC) analysis, we tested the effect of formaldehyde fixation, cell lysis temperature, mode of genome fragmentation, and removal of free RNA on the efficacy of RNA-DNA hybrid detection and implemented workflows that were able to distinguish complex and weak DRIP signals in a noisy background with high confidence. We also show that some of the workflows perform poorly and generate random answers. Furthermore, we found that the most commonly used genome fragmentation method (restriction enzyme digestion) led to the overrepresentation of lengthy DRIP fragments over coding ORFs, and this bias was enhanced at the first exons. Biased genome sampling severely compromised mapping resolution and prevented the assignment of precise biological function to a significant fraction of R-loops. The revised workflow presented herein is established and optimized using objective ROC analyses and provides reproducible and highly specific RNA-DNA hybrid detection.
Collapse
Affiliation(s)
- László Halász
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Research Centre for Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Zsolt Karányi
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Research Centre for Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Internal Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Beáta Boros-Oláh
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Research Centre for Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tímea Kuik-Rózsa
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Research Centre for Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Éva Sipos
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Research Centre for Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Biopharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Éva Nagy
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Research Centre for Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Ágnes Mosolygó-L
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Research Centre for Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, University of Debrecen, 4032 Debrecen, Hungary
| | - Éva Rajnavölgyi
- Department of Immunology, University of Debrecen, 4032 Debrecen, Hungary
| | - Gábor Halmos
- Department of Biopharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Lóránt Székvölgyi
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Research Centre for Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
16
|
Brant L, Georgomanolis T, Nikolic M, Brackley CA, Kolovos P, van Ijcken W, Grosveld FG, Marenduzzo D, Papantonis A. Exploiting native forces to capture chromosome conformation in mammalian cell nuclei. Mol Syst Biol 2016; 12:891. [PMID: 27940490 PMCID: PMC5199122 DOI: 10.15252/msb.20167311] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mammalian interphase chromosomes fold into a multitude of loops to fit the confines of cell nuclei, and looping is tightly linked to regulated function. Chromosome conformation capture (3C) technology has significantly advanced our understanding of this structure‐to‐function relationship. However, all 3C‐based methods rely on chemical cross‐linking to stabilize spatial interactions. This step remains a “black box” as regards the biases it may introduce, and some discrepancies between microscopy and 3C studies have now been reported. To address these concerns, we developed “i3C”, a novel approach for capturing spatial interactions without a need for cross‐linking. We apply i3C to intact nuclei of living cells and exploit native forces that stabilize chromatin folding. Using different cell types and loci, computational modeling, and a methylation‐based orthogonal validation method, “TALE‐iD”, we show that native interactions resemble cross‐linked ones, but display improved signal‐to‐noise ratios and are more focal on regulatory elements and CTCF sites, while strictly abiding to topologically associating domain restrictions.
Collapse
Affiliation(s)
- Lilija Brant
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | | | - Milos Nikolic
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Chris A Brackley
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Petros Kolovos
- Department of Cell Biology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Frank G Grosveld
- Department of Cell Biology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Davide Marenduzzo
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
17
|
ADPr-ChAP: Mapping ADP-Ribosylation onto the Genome. Mol Cell 2016; 61:327-328. [PMID: 26849191 DOI: 10.1016/j.molcel.2016.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In this issue of Molecular Cell, Bartolomei et al. (2016) describe a chromatin affinity precipitation method using well-characterized ADP-ribose binding domains to provide the first genome-wide view of ADP-ribosylated chromatin. Here, we discuss its potential applications and the remaining challenges ahead.
Collapse
|
18
|
Moritz B, Woltering L, Becker PB, Göpfert U. High levels of histone H3 acetylation at the CMV promoter are predictive of stable expression in Chinese hamster ovary cells. Biotechnol Prog 2016; 32:776-86. [DOI: 10.1002/btpr.2271] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/11/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Benjamin Moritz
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich; Germany
- Biomedical Center and Center for Integrated Protein Science, Molecular Biology Division, Ludwig-Maximilians-University; Munich Germany
| | - Laura Woltering
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich; Germany
| | - Peter B. Becker
- Biomedical Center and Center for Integrated Protein Science, Molecular Biology Division, Ludwig-Maximilians-University; Munich Germany
| | - Ulrich Göpfert
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich; Germany
| |
Collapse
|
19
|
Bartolomei G, Leutert M, Manzo M, Baubec T, Hottiger MO. Analysis of Chromatin ADP-Ribosylation at the Genome-wide Level and at Specific Loci by ADPr-ChAP. Mol Cell 2016; 61:474-485. [PMID: 26833088 DOI: 10.1016/j.molcel.2015.12.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 10/21/2015] [Accepted: 12/23/2015] [Indexed: 01/04/2023]
Abstract
Chromatin ADP-ribosylation regulates important cellular processes. However, the exact location and magnitude of chromatin ADP-ribosylation are largely unknown. A robust and versatile method for assessing chromatin ADP-ribosylation is therefore crucial for further understanding its function. Here, we present a chromatin affinity precipitation method based on the high specificity and avidity of two well-characterized ADP-ribose binding domains to map chromatin ADP-ribosylation at the genome-wide scale and at specific loci. Our ADPr-ChAP method revealed that in cells exposed to oxidative stress, ADP-ribosylation of chromatin scales with histone density, with highest levels at heterochromatic sites and depletion at active promoters. Furthermore, in growth factor-induced adipocyte differentiation, increased chromatin ADP-ribosylation was observed at PPARγ target genes, whose expression is ADP-ribosylation dependent. In combination with deep-sequencing and conventional chromatin immunoprecipitation, the established ADPr-ChAP provides a valuable resource for the bioinformatic comparison of ADP-ribosylation with other chromatin modifications and for addressing its role in other biologically important processes.
Collapse
Affiliation(s)
- Giody Bartolomei
- Department of Molecular Mechanisms of Disease, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Molecular Life Science PhD Program of the Life Science Zurich Graduate School, University of Zurich, 8057 Zurich, Switzerland
| | - Mario Leutert
- Department of Molecular Mechanisms of Disease, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Molecular Life Science PhD Program of the Life Science Zurich Graduate School, University of Zurich, 8057 Zurich, Switzerland
| | - Massimiliano Manzo
- Department of Molecular Mechanisms of Disease, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Molecular Life Science PhD Program of the Life Science Zurich Graduate School, University of Zurich, 8057 Zurich, Switzerland
| | - Tuncay Baubec
- Department of Molecular Mechanisms of Disease, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Michael O Hottiger
- Department of Molecular Mechanisms of Disease, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
20
|
Abstract
Chromatin immunoprecipitation (ChIP) is a valuable method to investigate protein-DNA interactions in vivo. Since its discovery it has been indispensable to identify binding sites and patterns of a variety of DNA-interacting proteins, such as transcription factors and regulators, modified histones, and epigenetic modifiers. The Polycomb repressors were the first proteins that have been mapped using this technique, which provided the mechanistic basis for the understanding of their biological function. Cross-linked (XChIP) or native (NChIP) chromatin from tissues or cultured cells is fragmented and the protein of interest is immunoprecipitated using a specific antibody. The co-precipitated DNA is then purified and subjected to analysis by region-specific PCR, DNA microarray (ChIP-on-chip), or next-generation sequencing (ChIP-seq). The assay can therefore produce information about the localization of the analyzed protein at specific candidate loci or throughout the entire genome. In this chapter, we provide a detailed protocol of the basic standard ChIP assay and some remarks about variations.
Collapse
Affiliation(s)
- Laura Wiehle
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany.
| | - Achim Breiling
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| |
Collapse
|
21
|
ChIP bias as a function of cross-linking time. Chromosome Res 2015; 24:175-81. [PMID: 26685864 DOI: 10.1007/s10577-015-9509-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 11/12/2015] [Accepted: 11/18/2015] [Indexed: 01/01/2023]
Abstract
The chromatin immunoprecipitation (ChIP) assay is widely used to capture interactions between chromatin and regulatory proteins in vivo. Formaldehyde cross-linking of DNA and proteins is a critical step required to trap their interactions inside the cells before immunoprecipitation and analysis. Yet insufficient attention has been given to variables that might give rise to artifacts in this procedure, such as the duration of cross-linking. We analyzed the dependence of the ChIP signal on the duration of formaldehyde cross-linking time for two proteins: DNA topoisomerase 1 (Top1) that is functionally associated with the double helix in vivo, especially with active chromatin, and green fluorescent protein (GFP) that has no known bona fide interactions with DNA. With short time of formaldehyde fixation, only Top1 immunoprecipation efficiently recovered DNA from active promoters, whereas prolonged fixation augmented non-specific recovery of GFP dramatizing the need to optimize ChIP protocols to minimize the time of cross-linking, especially for abundant nuclear proteins. Thus, ChIP is a powerful approach to study the localization of protein on the genome when care is taken to manage potential artifacts.
Collapse
|
22
|
Ciccarone F, Valentini E, Bacalini MG, Zampieri M, Calabrese R, Guastafierro T, Mariano G, Reale A, Franceschi C, Caiafa P. Poly(ADP-ribosyl)ation is involved in the epigenetic control of TET1 gene transcription. Oncotarget 2015; 5:10356-67. [PMID: 24939750 PMCID: PMC4279378 DOI: 10.18632/oncotarget.1905] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 04/16/2014] [Indexed: 12/24/2022] Open
Abstract
TET enzymes are the epigenetic factors involved in the formation of the sixth DNA base 5-hydroxymethylcytosine, whose deregulation has been associated with tumorigenesis. In particular, TET1 acts as tumor suppressor preventing cell proliferation and tumor metastasis and it has frequently been found down-regulated in cancer. Thus, considering the importance of a tight control of TET1 expression, the epigenetic mechanisms involved in the transcriptional regulation of TET1 gene are here investigated. The involvement of poly(ADP-ribosyl)ation in the control of DNA and histone methylation on TET1 gene was examined. PARP activity is able to positively regulate TET1 expression maintaining a permissive chromatin state characterized by DNA hypomethylation of TET1 CpG island as well as high levels of H3K4 trimethylation. These epigenetic modifications were affected by PAR depletion causing TET1 down-regulation and in turn reduced recruitment of TET1 protein on HOXA9 target gene. In conclusion, this work shows that PARP activity is a transcriptional regulator of TET1 gene through the control of epigenetic events and it suggests that deregulation of these mechanisms could account for TET1 repression in cancer.
Collapse
Affiliation(s)
- Fabio Ciccarone
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy. Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| | - Elisabetta Valentini
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy. Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| | - Maria Giulia Bacalini
- Department of Experimental Pathology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Michele Zampieri
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy. Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| | - Roberta Calabrese
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy. Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| | - Tiziana Guastafierro
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy. Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| | - Germano Mariano
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy. Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| | - Anna Reale
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy. Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| | - Claudio Franceschi
- Department of Experimental Pathology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Paola Caiafa
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy. Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
23
|
Improving chromatin immunoprecipitation (ChIP) by suppression of method-induced DNA-damage signaling. Methods Mol Biol 2015; 1228:67-81. [PMID: 25311123 DOI: 10.1007/978-1-4939-1680-1_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Genomic DNA is always associated with proteins that modulate the accessibility of the genetic information. This chromatin is the essential structure in which all nuclear activity from regulation to replication, transcription, and repair takes place. This dynamic structure can be most efficiently analyzed by using the method of chromatin immunoprecipitation (ChIP), where application of cell-permeable cross-linkers to living cells induces covalent bridging between proteins and adjacent DNA in the nucleus. After fragmentation of the DNA, the complexed proteins are isolated by binding to specific antibodies. The attached DNA is isolated and can be analyzed. This method has been improved multiple times and adjusted to different experimental needs. This chapter describes a further advance based on the observation that the current standard method itself induces alterations in the chromatin.
Collapse
|
24
|
Gavrilov A, Razin SV, Cavalli G. In vivo formaldehyde cross-linking: it is time for black box analysis. Brief Funct Genomics 2014; 14:163-5. [PMID: 25241225 PMCID: PMC6090872 DOI: 10.1093/bfgp/elu037] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Formaldehyde cross-linking is an important component of many technologies, including chromatin immunoprecipitation and chromosome conformation capture. The procedure remains empirical and poorly characterized, however, despite a long history of its use in research. Little is known about the specificity of in vivo cross-linking, its efficiency and chemical adducts induced by the procedure. It is time to search this black box.
Collapse
|
25
|
Tetievsky A, Assayag M, Ben-Hamo R, Efroni S, Cohen G, Abbas A, Horowitz M. Heat acclimation memory: do the kinetics of the deacclimated transcriptome predispose to rapid reacclimation and cytoprotection? J Appl Physiol (1985) 2014; 117:1262-77. [PMID: 25237184 DOI: 10.1152/japplphysiol.00422.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Faster reinduction of heat acclimation (AC) after its decline indicates "AC memory." Our previous results revealed involvement of epigenetic mechanisms of transcriptional regulation. We hypothesized that the decline of AC (DeAC) is a period of "dormant memory" during which many processes are alerted to enable rapid reacclimation (ReAC). Using a genomewide approach we studied the AC, DeAC, and ReAC transcriptomes, to uncover hallmark pathways linked to "molecular memory" in the cardioacclimatome. Fifty rats subjected to heat acclimation [34°C for 2d (AC2d) or 30d (AC30)], DeAC (24°C, 30 days), ReAC (34°C, 2 days), and untreated controls were used. The GeneChip Rat Gene 1.0 ST Array was employed for left ventricular (cardiac) mRNA hybridization. Three independent bioinformatic analyses showed that 1) during AC2d enrichment of DNA impair/repair-linked genes is seen, and this is the molecular on-switch of acclimation; 2) genes activated in AC30 underlie the qualitative physiological adaptations of cardiac performance; 3) particular molecular programs encompassing constitutive upregulation of p38 MAPK, Jak/Stat, and Akt pathways and targets are specifically activated during DeAC and ReAC; and 4) epigenetic markers such as linker histones (histones H1 cluster), associated with nucleosome spacing, transcriptional chromatin modifiers, poly-(ADP-ribose) polymerase-1 (PARP1) linked to chromatin compaction, and microRNAs are only altered during DeAC/ReAC. The latter are newcomers to the AC/DeAC puzzle. We suggest that these transcriptional responses maintain euchromatin and proteostasis and enable faster physiological recovery upon ReAC by rapidly reestablishing the protected acclimated cardiophenotype. We propose that the cardiac AC model can be applied to acclimation processes in general.
Collapse
Affiliation(s)
- Anna Tetievsky
- Laboratory of Environmental Physiology, Faculty of Dentistry, The Hebrew University, Jerusalem, Israel; and
| | - Miri Assayag
- Laboratory of Environmental Physiology, Faculty of Dentistry, The Hebrew University, Jerusalem, Israel; and
| | - Rotem Ben-Hamo
- The Mina and Everard Goodman Faculty of Life Science Bar Ilan University, Ramat Gan, Israel
| | - Sol Efroni
- The Mina and Everard Goodman Faculty of Life Science Bar Ilan University, Ramat Gan, Israel
| | - Gal Cohen
- Laboratory of Environmental Physiology, Faculty of Dentistry, The Hebrew University, Jerusalem, Israel; and
| | - Atallah Abbas
- Laboratory of Environmental Physiology, Faculty of Dentistry, The Hebrew University, Jerusalem, Israel; and
| | - Michal Horowitz
- Laboratory of Environmental Physiology, Faculty of Dentistry, The Hebrew University, Jerusalem, Israel; and
| |
Collapse
|
26
|
Proteome-wide Identification of Poly(ADP-Ribosyl)ation Targets in Different Genotoxic Stress Responses. Mol Cell 2013; 52:272-85. [DOI: 10.1016/j.molcel.2013.08.026] [Citation(s) in RCA: 259] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 06/28/2013] [Accepted: 08/13/2013] [Indexed: 01/01/2023]
|
27
|
Martello R, Mangerich A, Sass S, Dedon PC, Bürkle A. Quantification of cellular poly(ADP-ribosyl)ation by stable isotope dilution mass spectrometry reveals tissue- and drug-dependent stress response dynamics. ACS Chem Biol 2013; 8:1567-75. [PMID: 23631432 DOI: 10.1021/cb400170b] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Poly(ADP-ribosyl)ation is an essential post-translational modification with the biopolymer poly(ADP-ribose) (PAR). The reaction is catalyzed by poly(ADP-ribose) polymerases (PARPs) and plays key roles in cellular physiology and stress response. PARP inhibitors are currently being tested in clinical cancer treatment, in combination therapy, or as monotherapeutic agents by inducing synthetic lethality. We have developed an accurate and sensitive bioanalytical platform based on isotope dilution mass spectrometry in order to quantify steady-state and stress-induced PAR levels in cells and tissues and to characterize pharmacological properties of PARP inhibitors. In contrast to existing PAR-detection techniques, the LC-MS/MS method uses authentic isotope-labeled standards, which provide unequivocal chemical specificity to quantify cellular PAR in absolute terms with femtomol sensitivity. Using this platform we analyzed steady-state levels as well as stress-induced dynamics of poly(ADP-ribosyl)ation in a series of biological systems including cancer cell lines, mouse tissues, and primary human lymphocytes. Our results demonstrate a rapid and transient stress-induced increase in PAR levels by >100-fold in a dose- and time-dependent manner with significant differences between cell types and individual human lymphocyte donors. Furthermore, ex vivo pharmacodynamic studies in human lymphocytes provide new insight into pharmacological properties of clinically relevant PARP inhibitors. Finally, we adapted the LC-MS/MS method to quantify poly(ADP-ribosyl)ation in solid tissues and identified tissue-dependent associations between PARP1 expression and PAR levels in a series of different mouse organs. In conclusion, this study demonstrates that mass spectrometric quantification of cellular poly(ADP-ribosyl)ation has a wide range of applications in basic research as well as in drug development.
Collapse
Affiliation(s)
- Rita Martello
- Molecular
Toxicology Group and ‡Konstanz Research School Chemical Biology, University of Konstanz, Konstanz 78464, Germany
- Department
of Biological Engineering and ∥Center for Environmental Health Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02193, United States
| | - Aswin Mangerich
- Molecular
Toxicology Group and ‡Konstanz Research School Chemical Biology, University of Konstanz, Konstanz 78464, Germany
- Department
of Biological Engineering and ∥Center for Environmental Health Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02193, United States
| | - Sabine Sass
- Molecular
Toxicology Group and ‡Konstanz Research School Chemical Biology, University of Konstanz, Konstanz 78464, Germany
- Department
of Biological Engineering and ∥Center for Environmental Health Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02193, United States
| | - Peter C. Dedon
- Molecular
Toxicology Group and ‡Konstanz Research School Chemical Biology, University of Konstanz, Konstanz 78464, Germany
- Department
of Biological Engineering and ∥Center for Environmental Health Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02193, United States
| | - Alexander Bürkle
- Molecular
Toxicology Group and ‡Konstanz Research School Chemical Biology, University of Konstanz, Konstanz 78464, Germany
- Department
of Biological Engineering and ∥Center for Environmental Health Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02193, United States
| |
Collapse
|
28
|
Bornhorst J, Meyer S, Weber T, Böker C, Marschall T, Mangerich A, Beneke S, Bürkle A, Schwerdtle T. Molecular mechanisms of Mn induced neurotoxicity: RONS generation, genotoxicity, and DNA-damage response. Mol Nutr Food Res 2013; 57:1255-69. [DOI: 10.1002/mnfr.201200758] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/14/2013] [Accepted: 01/23/2013] [Indexed: 12/29/2022]
Affiliation(s)
- Julia Bornhorst
- Graduate School of Chemistry; University of Münster; Münster Germany
- Institute of Food Chemistry; University of Münster; Münster Germany
| | - Sören Meyer
- Graduate School of Chemistry; University of Münster; Münster Germany
- Institute of Food Chemistry; University of Münster; Münster Germany
| | - Till Weber
- Institute of Food Chemistry; University of Münster; Münster Germany
| | - Carolina Böker
- Institute of Food Chemistry; University of Münster; Münster Germany
| | - Talke Marschall
- Institute of Food Chemistry; University of Münster; Münster Germany
| | - Aswin Mangerich
- Department of Biology, Molecular Toxicology Group; University of Konstanz; Konstanz Germany
| | - Sascha Beneke
- Institute of Veterinary Pharmacology and Toxicology; University of Zürich; Zürich Switzerland
| | - Alexander Bürkle
- Department of Biology, Molecular Toxicology Group; University of Konstanz; Konstanz Germany
| | - Tanja Schwerdtle
- Institute of Food Chemistry; University of Münster; Münster Germany
| |
Collapse
|
29
|
Beneke S. Regulation of chromatin structure by poly(ADP-ribosyl)ation. Front Genet 2012; 3:169. [PMID: 22969794 PMCID: PMC3432497 DOI: 10.3389/fgene.2012.00169] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/17/2012] [Indexed: 12/23/2022] Open
Abstract
The interaction of DNA with proteins in the context of chromatin has to be tightly regulated to achieve so different tasks as packaging, transcription, replication and repair. The very rapid and transient post-translational modification of proteins by poly(ADP-ribose) has been shown to take part in all four. Originally identified as immediate cellular answer to a variety of genotoxic stresses, already early data indicated the ability of this highly charged nucleic acid-like polymer to modulate nucleosome structure, the basic unit of chromatin. At the same time the enzyme responsible for synthesizing poly(ADP-ribose), the zinc-finger protein poly(ADP-ribose) polymerase-1 (PARP1), was shown to control transcription initiation as basic factor TFIIC within the RNA-polymerase II machinery. Later research focused more on PARP-mediated regulation of DNA repair and cell death, but in the last few years, transcription as well as chromatin modulation has re-appeared on the scene. This review will discuss the impact of PARP1 on transcription and transcription factors, its implication in chromatin remodeling for DNA repair and probably also replication, and its role in controlling epigenetic events such as DNA methylation and the functionality of the insulator protein CCCTC-binding factor.
Collapse
Affiliation(s)
- Sascha Beneke
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich Zurich, Switzerland
| |
Collapse
|