1
|
Wang Z, Jiao P. Roles of non-coding RNAs and exosomal non-coding RNAs, particularly microRNAs, long non-coding RNAs, and circular RNAs, in pathogenic mechanisms behind chronic pain: A review. Int J Biol Macromol 2025; 307:141945. [PMID: 40074135 DOI: 10.1016/j.ijbiomac.2025.141945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
Chronic pain is a significant public health concern that diminishes patients' quality of life and imposes considerable socioeconomic costs. Effective pharmacological treatments for ongoing pain are limited. Recent studies have indicated that various models of chronic pain-such as neuropathic pain, inflammatory pain, and pain associated with cancer-have abnormal levels of long noncoding RNAs (lncRNAs). Research has explored how these abnormal lncRNAs influence the activation of inflammatory cytokines, microRNAs, and other related molecules, which are crucial to the development of chronic pain. These findings suggest that these lncRNAs are vital in chronic pain mechanisms within the spinal cord and dorsal root ganglion following nerve injury. Additionally, exosomes, which can traverse the blood-brain barrier, are considered carriers of noncoding RNAs (ncRNAs) from neurons to systemic circulation. This study aims to summarize the existing knowledge on ncRNAs and exosomal ncRNAs in the context of chronic pain, highlighting potential biomarkers for diagnosis, regulatory roles in disease progression, therapeutic strategies, and clinical implications.
Collapse
Affiliation(s)
- Zhongkai Wang
- Department of Pain and Rehabilitation, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China.
| | - Pengqing Jiao
- Department of Rheumatism and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
2
|
Altendorfer E, Mundlos S, Mayer A. A transcription coupling model for how enhancers communicate with their target genes. Nat Struct Mol Biol 2025; 32:598-606. [PMID: 40217119 DOI: 10.1038/s41594-025-01523-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 02/27/2025] [Indexed: 04/16/2025]
Abstract
How enhancers communicate with their target genes to influence transcription is an unresolved question of fundamental importance. Current models of the mechanism of enhancer-target gene or enhancer-promoter (E-P) communication are transcription-factor-centric and underappreciate major findings, including that enhancers are themselves transcribed by RNA polymerase II, which correlates with enhancer activity. In this Perspective, we posit that enhancer transcription and its products, enhancer RNAs, are elementary components of enhancer-gene communication. Specifically, we discuss the possibility that transcription at enhancers and at their cognate genes are linked and that this coupling is at the basis of how enhancers communicate with their targets. This model of transcriptional coupling between enhancers and their target genes is supported by growing experimental evidence and represents a synthesis of recent key discoveries.
Collapse
Affiliation(s)
- Elisabeth Altendorfer
- Otto-Warburg-Laboratory, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Stefan Mundlos
- Development and Disease group, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute for Medical and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Mayer
- Otto-Warburg-Laboratory, Max Planck Institute for Molecular Genetics, Berlin, Germany.
| |
Collapse
|
3
|
Zeng S, Li Z, Li X, Du Q, Zhang Y, Zhong Z, Wang H, Zhang S, Li P, Li H, Chen L, Jiang A, Shang P, Li M, Long K. Inhibition of triglyceride metabolism-associated enhancers alters lipid deposition during adipocyte differentiation. FASEB J 2025; 39:e70347. [PMID: 39873971 PMCID: PMC11774232 DOI: 10.1096/fj.202401137r] [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: 05/22/2024] [Revised: 12/28/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
Triglyceride (TG) metabolism is a complex and highly coordinated biological process regulated by a series of genes, and its dysregulation can lead to the occurrence of disorders in lipid metabolism. However, the transcriptional regulatory mechanisms of crucial genes in TG metabolism mediated by enhancer-promoter interactions remain elusive. Here, we identified candidate enhancers regulating the Agpat2, Dgat1, Dgat2, Pnpla2, and Lipe genes in 3T3-L1 adipocytes by integrating epigenomic data (H3K27ac, H3K4me1, and DHS-seq) with chromatin three-dimensional interaction data. Luciferase reporter assays revealed that 11 enhancers exhibited fluorescence activity. The repression of enhancers using the dCas9-KRAB system revealed the functional roles of enhancers of Dgat2 and Pnpla2 in regulating their expression and TG metabolism. Furthermore, transcriptome analyses revealed that inhibition of Dgat2-En4 downregulated pathways associated with lipid metabolism, lipid biosynthesis, and adipocyte differentiation. Additionally, overexpression and motif mutation experiments of transcription factor found that two TFs, PPARG and RXRA, regulate the activity of Agpat2-En1, Dgat2-En4, and Pnpla2-En5. Our study identified functional enhancers regulating TG metabolism and elucidated potential regulatory mechanisms of TG deposition from enhancer-promoter interactions, providing insights into understanding lipid deposition.
Collapse
Affiliation(s)
- Sha Zeng
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
| | - Ziqi Li
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
| | - Xiaokai Li
- Chongqing Academy of Animal SciencesChongqingChina
- National Center of Technology Innovation for PigsChongqingChina
| | - Qinjiao Du
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
| | - Yu Zhang
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
| | - Zhining Zhong
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
| | - Haoming Wang
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
| | - Songling Zhang
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
| | - Penghao Li
- Jinxin Research Institute for Reproductive Medicine and GeneticsSichuan Jinxin Xi'nan Women's and Children's HospitalChengduChina
| | - Haohuan Li
- College of Veterinary MedicineSichuan Agricultural UniversityChengduChina
| | - Li Chen
- Chongqing Academy of Animal SciencesChongqingChina
- National Center of Technology Innovation for PigsChongqingChina
| | - Anan Jiang
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
| | - Peng Shang
- Animal Science CollegeTibet Agriculture and Animal Husbandry UniversityLinzhiChina
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
| | - Keren Long
- State Key Laboratory of Swine and Poultry Breeding IndustrySichuan Agricultural UniversityChengduChina
- College of Animal Science and TechnologySichuan Agricultural UniversityChengduChina
- Chongqing Academy of Animal SciencesChongqingChina
- National Center of Technology Innovation for PigsChongqingChina
| |
Collapse
|
4
|
Akiki RM, Cornbrooks RG, Magami K, Greige A, Snyder KK, Wood DJ, Herrington MC, Mace P, Blidy K, Koike N, Berto S, Cowan CW, Taniguchi M. A long noncoding eRNA forms R-loops to shape emotional experience-induced behavioral adaptation. Science 2024; 386:1282-1289. [PMID: 39666799 PMCID: PMC12071198 DOI: 10.1126/science.adp1562] [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: 03/19/2024] [Accepted: 10/09/2024] [Indexed: 12/14/2024]
Abstract
Emotional experiences often evoke neural plasticity that supports adaptive changes in behavior, including maladaptive plasticity associated with mood and substance use disorders. These adaptations are supported in part by experience-dependent activation of immediate-early response genes, such as Npas4 (neuronal PAS domain protein 4). Here we show that a conserved long noncoding enhancer RNA (lnc-eRNA), transcribed from an activity-sensitive enhancer, produces DNA:RNA hybrid R-loop structures that support three-dimensional chromatin looping between enhancer and proximal promoter and rapid Npas4 gene induction. Furthermore, in mouse models, Npas4 lnc-eRNA and its R-loop are required for the development of behavioral adaptations produced by chronic psychosocial stress or cocaine exposure, revealing a potential role for this regulatory mechanism in the transmission of emotional experiences.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Adaptation, Psychological/drug effects
- Adaptation, Psychological/physiology
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Behavior, Animal
- Chromatin/metabolism
- Cocaine/pharmacology
- Emotions/drug effects
- Emotions/physiology
- Enhancer Elements, Genetic
- Mice, Inbred C57BL
- Neuronal Plasticity
- Promoter Regions, Genetic
- R-Loop Structures
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Stress, Psychological/genetics
- Stress, Psychological/psychology
Collapse
Affiliation(s)
- Rose Marie Akiki
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
- Medical Scientist Training Program, Medical University of South Carolina; Charleston, SC, USA
| | - Rebecca G. Cornbrooks
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
| | - Kosuke Magami
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
| | - Alain Greige
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
- Medical Scientist Training Program, Medical University of South Carolina; Charleston, SC, USA
| | - Kirsten K. Snyder
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
| | - Daniel J. Wood
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
- Medical Scientist Training Program, Medical University of South Carolina; Charleston, SC, USA
| | | | - Philip Mace
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
| | - Kyle Blidy
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
| | - Nobuya Koike
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine; Kyoto, Japan
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
| | - Christopher W. Cowan
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina; Charleston, SC, USA
| |
Collapse
|
5
|
Han X, Tian W, Sun L, Wang H, Li Y, Jia H, Gao G, Mai L, Yin S, Zhang Q, Liu Y. Prognosis of colon cancer patients based on enhancer RNAs-related genes. J Cancer Res Clin Oncol 2023; 149:12443-12457. [PMID: 37438540 DOI: 10.1007/s00432-023-05130-1] [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: 06/04/2023] [Accepted: 07/04/2023] [Indexed: 07/14/2023]
Abstract
PURPOSE Colon cancer (CC) is a cancer of the large intestine with high prevalence and poor prognosis. enhancer RNAs. Therefore, valuable tools or biomarkers for predicting patient status, directing clinical practice, and reducing overtreatment are needed. Enhancer RNAs (eRNAs), a class of noncoding RNAs transcribed from enhancers, have been shown to function as regulators of oncogene or tumor suppressor gene expression. The aim of our study was to explore the potential roles of eRNAs and their target enhancer-related genes (ERGs) in the prognosis of CC. METHODS Selected CC cases (stage I-III) from The Cancer Genome Atlas database were used as a training set, and cases from the Gene Expression Omnibus were used as the validation set. ERGs associated with prognosis were screened through three steps: potential, candidate, and prognosis ERGs. Multivariate Cox proportional hazards analysis was used to identify independent prognostic factors, and a nomogram was created. Calibration curves were drawn by comparing predicted and observed survival probability. For validation, the calibration curves and ROC analysis were also applied to two external validation sets. The biological significance and clinical application of the genes obtained were investigated. RESULTS Based on the multiple tiers of strict screening, 11 prognostic ERGs were obtained, which were combined to obtain a prognosis signature. A compound nomogram integrating age, TNM classification, and the prognostic signature was constructed. The model was reliable in distinguishing the risk of patients with stage I-III CC, with AUCs of 0.78 and 0.70 at 5 and 7 years, respectively. There was good reproducibility in calibration curves. The prognostic model also yielded good prediction capability in the validation sets. CONCLUSION In this study, the usefulness and specificity of the ERGs in prognosis were described, which should be considered a key feature in the clinical guidance of CC patients with early stage. We concluded that the major implications of the eRNAs and ERGs should be valued, which would be an emerging hallmark in the prognosis of cancer.
Collapse
Affiliation(s)
- Xinhao Han
- Department of Biostatistics, School of Public Health, Harbin Medical University, No. 157 Baojian Road, 150081, Harbin City, Heilongjiang Province, China
| | - Wei Tian
- Department of Biostatistics, School of Public Health, Harbin Medical University, No. 157 Baojian Road, 150081, Harbin City, Heilongjiang Province, China
| | - Lin Sun
- Department of Biostatistics, School of Public Health, Harbin Medical University, No. 157 Baojian Road, 150081, Harbin City, Heilongjiang Province, China
| | - Hongying Wang
- Department of Biostatistics, School of Public Health, Harbin Medical University, No. 157 Baojian Road, 150081, Harbin City, Heilongjiang Province, China
| | - Yan Li
- Department of Biostatistics, School of Public Health, Harbin Medical University, No. 157 Baojian Road, 150081, Harbin City, Heilongjiang Province, China
| | - Huixun Jia
- Department of Ophthalmology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guohong Gao
- Department of Biostatistics, School of Public Health, Harbin Medical University, No. 157 Baojian Road, 150081, Harbin City, Heilongjiang Province, China
| | - Liudan Mai
- Department of Biostatistics, School of Public Health, Harbin Medical University, No. 157 Baojian Road, 150081, Harbin City, Heilongjiang Province, China
| | - Shuwen Yin
- Department of Biostatistics, School of Public Health, Harbin Medical University, No. 157 Baojian Road, 150081, Harbin City, Heilongjiang Province, China
| | - Qiuju Zhang
- Department of Biostatistics, School of Public Health, Harbin Medical University, No. 157 Baojian Road, 150081, Harbin City, Heilongjiang Province, China.
| | - Yanlong Liu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, 150081, Harbin City, Heilongjiang Province, China.
| |
Collapse
|
6
|
Herbert A. Flipons and small RNAs accentuate the asymmetries of pervasive transcription by the reset and sequence-specific microcoding of promoter conformation. J Biol Chem 2023; 299:105140. [PMID: 37544644 PMCID: PMC10474125 DOI: 10.1016/j.jbc.2023.105140] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023] Open
Abstract
The role of alternate DNA conformations such as Z-DNA in the regulation of transcription is currently underappreciated. These structures are encoded by sequences called flipons, many of which are enriched in promoter and enhancer regions. Through a change in their conformation, flipons provide a tunable mechanism to mechanically reset promoters for the next round of transcription. They act as actuators that capture and release energy to ensure that the turnover of the proteins at promoters is optimized to cell state. Likewise, the single-stranded DNA formed as flipons cycle facilitates the docking of RNAs that are able to microcode promoter conformations and canalize the pervasive transcription commonly observed in metazoan genomes. The strand-specific nature of the interaction between RNA and DNA likely accounts for the known asymmetry of epigenetic marks present on the histone tetramers that pair to form nucleosomes. The role of these supercoil-dependent processes in promoter choice and transcriptional interference is reviewed. The evolutionary implications are examined: the resilience and canalization of flipon-dependent gene regulation is contrasted with the rapid adaptation enabled by the spread of flipon repeats throughout the genome. Overall, the current findings underscore the important role of flipons in modulating the readout of genetic information and how little we know about their biology.
Collapse
Affiliation(s)
- Alan Herbert
- Discovery Division, InsideOutBio, Charlestown, Massachusetts, USA.
| |
Collapse
|
7
|
Kumar D, Sahoo SS, Chauss D, Kazemian M, Afzali B. Non-coding RNAs in immunoregulation and autoimmunity: Technological advances and critical limitations. J Autoimmun 2023; 134:102982. [PMID: 36592512 PMCID: PMC9908861 DOI: 10.1016/j.jaut.2022.102982] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 01/02/2023]
Abstract
Immune cell function is critically dependent on precise control over transcriptional output from the genome. In this respect, integration of environmental signals that regulate gene expression, specifically by transcription factors, enhancer DNA elements, genome topography and non-coding RNAs (ncRNAs), are key components. The first three have been extensively investigated. Even though non-coding RNAs represent the vast majority of cellular RNA species, this class of RNA remains historically understudied. This is partly because of a lag in technological and bioinformatic innovations specifically capable of identifying and accurately measuring their expression. Nevertheless, recent progress in this domain has enabled a profusion of publications identifying novel sub-types of ncRNAs and studies directly addressing the function of ncRNAs in human health and disease. Many ncRNAs, including circular and enhancer RNAs, have now been demonstrated to play key functions in the regulation of immune cells and to show associations with immune-mediated diseases. Some ncRNAs may function as biomarkers of disease, aiding in diagnostics and in estimating response to treatment, while others may play a direct role in the pathogenesis of disease. Importantly, some are relatively stable and are amenable to therapeutic targeting, for example through gene therapy. Here, we provide an overview of ncRNAs and review technological advances that enable their study and hold substantial promise for the future. We provide context-specific examples by examining the associations of ncRNAs with four prototypical human autoimmune diseases, specifically rheumatoid arthritis, psoriasis, inflammatory bowel disease and multiple sclerosis. We anticipate that the utility and mechanistic roles of these ncRNAs in autoimmunity will be further elucidated in the near future.
Collapse
Affiliation(s)
- Dhaneshwar Kumar
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Subhransu Sekhar Sahoo
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Daniel Chauss
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA.
| |
Collapse
|
8
|
Niewiadomska-Cimicka A, Hache A, Le Gras S, Keime C, Ye T, Eisenmann A, Harichane I, Roux MJ, Messaddeq N, Clérin E, Léveillard T, Trottier Y. Polyglutamine-expanded ATXN7 alters a specific epigenetic signature underlying photoreceptor identity gene expression in SCA7 mouse retinopathy. J Biomed Sci 2022; 29:107. [PMID: 36539812 PMCID: PMC9768914 DOI: 10.1186/s12929-022-00892-1] [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: 08/02/2022] [Accepted: 12/11/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Spinocerebellar ataxia type 7 (SCA7) is a neurodegenerative disorder that primarily affects the cerebellum and retina. SCA7 is caused by a polyglutamine expansion in the ATXN7 protein, a subunit of the transcriptional coactivator SAGA that acetylates histone H3 to deposit narrow H3K9ac mark at DNA regulatory elements of active genes. Defective histone acetylation has been presented as a possible cause for gene deregulation in SCA7 mouse models. However, the topography of acetylation defects at the whole genome level and its relationship to changes in gene expression remain to be determined. METHODS We performed deep RNA-sequencing and chromatin immunoprecipitation coupled to high-throughput sequencing to examine the genome-wide correlation between gene deregulation and alteration of the active transcription marks, e.g. SAGA-related H3K9ac, CBP-related H3K27ac and RNA polymerase II (RNAPII), in a SCA7 mouse retinopathy model. RESULTS Our analyses revealed that active transcription marks are reduced at most gene promoters in SCA7 retina, while a limited number of genes show changes in expression. We found that SCA7 retinopathy is caused by preferential downregulation of hundreds of highly expressed genes that define morphological and physiological identities of mature photoreceptors. We further uncovered that these photoreceptor genes harbor unusually broad H3K9ac profiles spanning the entire gene bodies and have a low RNAPII pausing. This broad H3K9ac signature co-occurs with other features that delineate superenhancers, including broad H3K27ac, binding sites for photoreceptor specific transcription factors and expression of enhancer-related non-coding RNAs (eRNAs). In SCA7 retina, downregulated photoreceptor genes show decreased H3K9 and H3K27 acetylation and eRNA expression as well as increased RNAPII pausing, suggesting that superenhancer-related features are altered. CONCLUSIONS Our study thus provides evidence that distinctive epigenetic configurations underlying high expression of cell-type specific genes are preferentially impaired in SCA7, resulting in a defect in the maintenance of identity features of mature photoreceptors. Our results also suggest that continuous SAGA-driven acetylation plays a role in preserving post-mitotic neuronal identity.
Collapse
Affiliation(s)
- Anna Niewiadomska-Cimicka
- grid.11843.3f0000 0001 2157 9291Institute of Genetics and Molecular and Cellular Biology (IGBMC), INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France
| | - Antoine Hache
- grid.11843.3f0000 0001 2157 9291Institute of Genetics and Molecular and Cellular Biology (IGBMC), INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France
| | - Stéphanie Le Gras
- grid.11843.3f0000 0001 2157 9291Institute of Genetics and Molecular and Cellular Biology (IGBMC), INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France
| | - Céline Keime
- grid.11843.3f0000 0001 2157 9291Institute of Genetics and Molecular and Cellular Biology (IGBMC), INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France
| | - Tao Ye
- grid.11843.3f0000 0001 2157 9291Institute of Genetics and Molecular and Cellular Biology (IGBMC), INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France
| | - Aurelie Eisenmann
- grid.11843.3f0000 0001 2157 9291Institute of Genetics and Molecular and Cellular Biology (IGBMC), INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France
| | - Imen Harichane
- grid.462844.80000 0001 2308 1657Department of Genetics, INSERM, CNRS, Institut de la Vision, Sorbonne University, 75012 Paris, France
| | - Michel J. Roux
- grid.11843.3f0000 0001 2157 9291Institute of Genetics and Molecular and Cellular Biology (IGBMC), INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France
| | - Nadia Messaddeq
- grid.11843.3f0000 0001 2157 9291Institute of Genetics and Molecular and Cellular Biology (IGBMC), INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France
| | - Emmanuelle Clérin
- grid.462844.80000 0001 2308 1657Department of Genetics, INSERM, CNRS, Institut de la Vision, Sorbonne University, 75012 Paris, France
| | - Thierry Léveillard
- grid.462844.80000 0001 2308 1657Department of Genetics, INSERM, CNRS, Institut de la Vision, Sorbonne University, 75012 Paris, France
| | - Yvon Trottier
- grid.11843.3f0000 0001 2157 9291Institute of Genetics and Molecular and Cellular Biology (IGBMC), INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France
| |
Collapse
|
9
|
Integrative analysis reveals histone demethylase LSD1 promotes RNA polymerase II pausing. iScience 2022; 25:105049. [PMID: 36124234 PMCID: PMC9482124 DOI: 10.1016/j.isci.2022.105049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/18/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Lysine-specific demethylase 1 (LSD1) is well-known for its role in decommissioning enhancers during mouse embryonic stem cell (ESC) differentiation. Its role in gene promoters remains poorly understood despite its widespread presence at these sites. Here, we report that LSD1 promotes RNA polymerase II (RNAPII) pausing, a rate-limiting step in transcription regulation, in ESCs. We found the knockdown of LSD1 preferentially affects genes with higher RNAPII pausing. Next, we demonstrate that the co-localization sites of LSD1 and MYC, a factor known to regulate pause-release, are enriched for other RNAPII pausing factors. We show that LSD1 and MYC directly interact and MYC recruitment to genes co-regulated with LSD1 is dependent on LSD1 but not vice versa. The co-regulated gene set is significantly enriched for housekeeping processes and depleted of transcription factors compared to those bound by LSD1 alone. Collectively, our integrative analysis reveals a pleiotropic role of LSD1 in promoting RNAPII pausing. LSD1 promotes RNA polymerase II pausing in mouse embryonic stem cells LSD1 knockdown causes global reduction of RNAPII pausing Co-localized sites of LSD1 and MYC are enriched for RNAPII pausing and releasing factors MYC recruitment to co-regulated genes is dependent on LSD1 but not vice versa
Collapse
|
10
|
Paranjapye A, NandyMazumdar M, Harris A. Krüppel-Like Factor 5 Regulates CFTR Expression Through Repression by Maintaining Chromatin Architecture Coupled with Direct Enhancer Activation. J Mol Biol 2022; 434:167561. [PMID: 35341742 PMCID: PMC9086126 DOI: 10.1016/j.jmb.2022.167561] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023]
Abstract
Single cell RNA-sequencing has accurately identified cell types within the human airway that express the Cystic Fibrosis Transmembrane Conductance regulator (CFTR) gene. Low abundance CFTR transcripts are seen in many secretory cells, while high levels are restricted to rare pulmonary ionocytes. Here we focus on the mechanisms coordinating basal CFTR expression in the secretory compartment. Cell-selective regulation of CFTR is achieved within its invariant topologically associating domain by the recruitment of cis-regulatory elements (CREs). CRE activity is coordinated by cell-type-selective transcription factors. One such factor, Krüppel-Like Factor 5 (KLF5), profoundly represses CFTR transcript and protein in primary human airway epithelial cells and airway cell lines. Here we reveal the mechanism of action of KLF5 upon the CFTR gene. We find that depletion or ablation of KLF5 from airway epithelial cells changes higher order chromatin structure at the CFTR locus. Critical looping interactions that are required for normal gene expression are altered, the H3K27ac active chromatin mark is redistributed, and CTCF occupancy is modified. However, mutation of a single KLF5 binding site within a pivotal airway cell CRE abolishes CFTR expression. Hence, KLF5 has both direct activating and indirect repressive effects, which together coordinate CFTR expression in the airway.
Collapse
Affiliation(s)
- Alekh Paranjapye
- Department of Genetics and Genome Sciences, and Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Monali NandyMazumdar
- Department of Genetics and Genome Sciences, and Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ann Harris
- Department of Genetics and Genome Sciences, and Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
11
|
Lozano R, Booth GT, Omar BY, Li B, Buckler ES, Lis JT, del Carpio DP, Jannink JL. RNA polymerase mapping in plants identifies intergenic regulatory elements enriched in causal variants. G3 (BETHESDA, MD.) 2021; 11:jkab273. [PMID: 34499719 PMCID: PMC8527479 DOI: 10.1093/g3journal/jkab273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/04/2021] [Indexed: 12/14/2022]
Abstract
Control of gene expression is fundamental at every level of cell function. Promoter-proximal pausing and divergent transcription at promoters and enhancers, which are prominent features in animals, have only been studied in a handful of research experiments in plants. PRO-Seq analysis in cassava (Manihot esculenta) identified peaks of transcriptionally engaged RNA polymerase at both the 5' and 3' end of genes, consistent with paused or slowly moving Polymerase. In addition, we identified divergent transcription at intergenic sites. A full genome search for bi-directional transcription using an algorithm for enhancer detection developed in mammals (dREG) identified many intergenic regulatory element (IRE) candidates. These sites showed distinct patterns of methylation and nucleotide conservation based on genomic evolutionary rate profiling (GERP). SNPs within these IRE candidates explained significantly more variation in fitness and root composition than SNPs in chromosomal segments randomly ascertained from the same intergenic distribution, strongly suggesting a functional importance of these sites. Maize GRO-Seq data showed RNA polymerase occupancy at IREs consistent with patterns in cassava. Furthermore, these IREs in maize significantly overlapped with sites previously identified on the basis of open chromatin, histone marks, and methylation, and were enriched for reported eQTL. Our results suggest that bidirectional transcription can identify intergenic genomic regions in plants that play an important role in transcription regulation and whose identification has the potential to aid crop improvement.
Collapse
Affiliation(s)
- Roberto Lozano
- Plant Breeding and Genetics, School of Integrative Plant Science, Cornell University, Ithaca, NY 14853, USA
| | - Gregory T Booth
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | | | - Bo Li
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Science, Beijing 100101, China
| | - Edward S Buckler
- Plant Breeding and Genetics, School of Integrative Plant Science, Cornell University, Ithaca, NY 14853, USA
- Institute for Genomic Diversity, Cornell University, Ithaca, NY 14853, USA
- United States Department of Agriculture, Agricultural Research Service (USDA-ARS) R.W. Holley Center for Agriculture and Health, Ithaca, NY 14853, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Dunia Pino del Carpio
- Plant Breeding and Genetics, School of Integrative Plant Science, Cornell University, Ithaca, NY 14853, USA
| | - Jean-Luc Jannink
- Plant Breeding and Genetics, School of Integrative Plant Science, Cornell University, Ithaca, NY 14853, USA
- United States Department of Agriculture, Agricultural Research Service (USDA-ARS) R.W. Holley Center for Agriculture and Health, Ithaca, NY 14853, USA
| |
Collapse
|
12
|
Probiotics and Trained Immunity. Biomolecules 2021; 11:biom11101402. [PMID: 34680035 PMCID: PMC8533468 DOI: 10.3390/biom11101402] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 12/17/2022] Open
Abstract
The characteristics of innate immunity have recently been investigated in depth in several research articles, and original findings suggest that innate immunity also has a memory capacity, which has been named “trained immunity”. This notion has revolutionized our knowledge of the innate immune response. Thus, stimulation of trained immunity represents a therapeutic alternative that is worth exploring. In this context, probiotics, live microorganisms which when administered in adequate amounts confer a health benefit on the host, represent attractive candidates for the stimulation of trained immunity; however, although numerous studies have documented the beneficial proprieties of these microorganisms, their mechanisms of action are not yet fully understood. In this review, we propose to explore the putative connection between probiotics and stimulation of trained immunity.
Collapse
|
13
|
Yan H, Tian S, Kleinstern G, Wang Z, Lee JH, Boddicker NJ, Cerhan JR, Kay NE, Braggio E, Slager SL. Chronic lymphocytic leukemia (CLL) risk is mediated by multiple enhancer variants within CLL risk loci. Hum Mol Genet 2021; 29:2761-2774. [PMID: 32744316 DOI: 10.1093/hmg/ddaa165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/02/2020] [Accepted: 07/25/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in Western countries. It has a strong genetic basis, showing a ~ 8-fold increased risk of CLL in first-degree relatives. Genome-wide association studies (GWAS) have identified 41 risk variants across 41 loci. However, for a majority of the loci, the functional variants and the mechanisms underlying their causal roles remain undefined. Here, we examined the genetic and epigenetic features associated with 12 index variants, along with any correlated (r2 ≥ 0.5) variants, at the CLL risk loci located outside of gene promoters. Based on publicly available ChIP-seq and chromatin accessibility data as well as our own ChIP-seq data from CLL patients, we identified six candidate functional variants at six loci and at least two candidate functional variants at each of the remaining six loci. The functional variants are predominantly located within enhancers or super-enhancers, including bi-directionally transcribed enhancers, which are often restricted to immune cell types. Furthermore, we found that, at 78% of the functional variants, the alternative alleles altered the transcription factor binding motifs or histone modifications, indicating the involvement of these variants in the change of local chromatin state. Finally, the enhancers carrying functional variants physically interacted with genes enriched in the type I interferon signaling pathway, apoptosis, or TP53 network that are known to play key roles in CLL. These results support the regulatory roles for inherited noncoding variants in the pathogenesis of CLL.
Collapse
Affiliation(s)
- Huihuang Yan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Shulan Tian
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Geffen Kleinstern
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhiquan Wang
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jeong-Heon Lee
- Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - James R Cerhan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Neil E Kay
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Esteban Braggio
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Susan L Slager
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
14
|
Blasiak J, Hyttinen JMT, Szczepanska J, Pawlowska E, Kaarniranta K. Potential of Long Non-Coding RNAs in Age-Related Macular Degeneration. Int J Mol Sci 2021; 22:9178. [PMID: 34502084 PMCID: PMC8431062 DOI: 10.3390/ijms22179178] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of visual impairment in the aging population with poorly known pathogenesis and lack of effective treatment. Age and family history are the strongest AMD risk factors, and several loci were identified to contribute to AMD. Recently, also the epigenetic profile was associated with AMD, and some long non-coding RNAs (lncRNAs) were shown to involve in AMD pathogenesis. The Vax2os1/2 (ventral anterior homeobox 2 opposite strand isoform 1) lncRNAs may modulate the balance between pro- and anti-angiogenic factors in the eye contributing to wet AMD. The stress-induced dedifferentiation of retinal pigment epithelium cells can be inhibited by the ZNF503-AS1 (zinc finger protein 503 antisense RNA 2) and LINC00167 lncRNAs. Overexpression of the PWRN2 (Prader-Willi region non-protein-coding RNA 2) lncRNA aggravated RPE cells apoptosis and mitochondrial impairment induced by oxidative stress. Several other lncRNAs were reported to exert protective or detrimental effects in AMD. However, many studies are limited to an association between lncRNA and AMD in patients or model systems with bioinformatics. Therefore, further works on lncRNAs in AMD are rational, and they should be enriched with mechanistic and clinical studies to validate conclusions obtained in high-throughput in vitro research.
Collapse
Affiliation(s)
- Janusz Blasiak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Juha M. T. Hyttinen
- Department of Ophthalmology, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Joanna Szczepanska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-216 Lodz, Poland;
| | - Elzbieta Pawlowska
- Department of Orthodontics, Medical University of Lodz, 92-217 Lodz, Poland;
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, 70210 Kuopio, Finland;
- Department of Ophthalmology, Kuopio University Hospital, 70210 Kuopio, Finland
| |
Collapse
|
15
|
Fu W, Zhao J, Hu W, Dai L, Jiang Z, Zhong S, Deng B, Huang Y, Wu W, Yin J. LINC01224/ZNF91 Promote Stem Cell-Like Properties and Drive Radioresistance in Non-Small Cell Lung Cancer. Cancer Manag Res 2021; 13:5671-5681. [PMID: 34285587 PMCID: PMC8286114 DOI: 10.2147/cmar.s313744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/13/2021] [Indexed: 12/22/2022] Open
Abstract
Background Radioresistance is the main reason for the failure of radiotherapy in non-small-cell lung cancer (NSCLC); however, the molecular mechanism of radioresistance is still unclear. Methods An RNA-Seq assay was used to screen differentially expressed long non-coding RNAs (lncRNAs) and genes in irradiation-resistant NSCLC cells. RT-PCR and Western blotting assays were performed to analyze the expressions of lncRNAs and genes. The chromosome conformation capture (3C) assay was performed to measure chromatin interactions. Cell cytotoxicity, cell apoptosis, sphere formation and Transwell assays were performed to assess cellular function. Results In this study, it was found that LINC01224 increased during the induction of radioresistance in NSCLC cells. LINC01224 was located within the enhancer of ZNF91, and LINC01224 could affect the transcription of ZNF91 by regulating the long-range interactions between the ZNF91 enhancer and promoter. Moreover, upregulation of LINC01224 and ZNF91 could promote irradiation resistance by regulating the stem cell-like properties of NSCLC cells. In addition, high expression levels of LINC01224 and ZNF91 in tissue samples were associated with radioresistance in NSCLC patients. Conclusion Our findings demonstrated that LINC01224/ZNF91 drove radioresistance regulation by promoting the stem cell-like properties in NSCLC.
Collapse
Affiliation(s)
- Wenfan Fu
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| | - Jian Zhao
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| | - Weimin Hu
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| | - Lu Dai
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| | - Zeyong Jiang
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| | - Shengpeng Zhong
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| | - Boyun Deng
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| | - Yun Huang
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| | - Wenjie Wu
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| | - Jun Yin
- Departments of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, People's Republic of China
| |
Collapse
|
16
|
Jasenosky LD, Nambu A, Tsytsykova AV, Ranjbar S, Haridas V, Kruidenier L, Tough DF, Goldfeld AE. Identification of a Distal Locus Enhancer Element That Controls Cell Type-Specific TNF and LTA Gene Expression in Human T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:2479-2488. [PMID: 32978279 DOI: 10.4049/jimmunol.1901311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 08/24/2020] [Indexed: 12/16/2022]
Abstract
The human TNF/LT locus genes TNF, LTA, and LTB are expressed in a cell type-specific manner. In this study, we show that a highly conserved NFAT binding site within the distal noncoding element hHS-8 coordinately controls TNF and LTA gene expression in human T cells. Upon activation of primary human CD4+ T cells, hHS-8 and the TNF and LTA promoters display increased H3K27 acetylation and nuclease sensitivity and coordinate induction of TNF, LTA, and hHS-8 enhancer RNA transcription occurs. Functional analyses using CRISPR/dead(d)Cas9 targeting of the hHS-8-NFAT site in the human T cell line CEM demonstrate significant reduction of TNF and LTA mRNA synthesis and of RNA polymerase II recruitment to their promoters. These studies elucidate how a distal element regulates the inducible cell type-specific gene expression program of the human TNF/LT locus and provide an approach for modulation of TNF and LTA transcription in human disease using CRISPR/dCas9.
Collapse
Affiliation(s)
- Luke D Jasenosky
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Aya Nambu
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Alla V Tsytsykova
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115.,Program in Hematology, Boston Children's Hospital, Boston, MA 02115
| | - Shahin Ranjbar
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Viraga Haridas
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | | | - David F Tough
- Adaptive Immunity Research Unit, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage SG1 2NY, United Kingdom
| | - Anne E Goldfeld
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115;
| |
Collapse
|
17
|
Adam MA, Harwell CC. Epigenetic regulation of cortical neurogenesis; orchestrating fate switches at the right time and place. Curr Opin Neurobiol 2020; 63:146-153. [PMID: 32428815 PMCID: PMC7483903 DOI: 10.1016/j.conb.2020.03.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/21/2022]
Abstract
Over the last several decades the field has made tremendous progress in understanding the proliferative behavior of cortical progenitors and the lineage relationships of their clonal progeny. The genetic and epigenetic mechanisms that control the dynamic patterns of gene expression during cortical development are only beginning to be characterized. In this review we highlight the most well characterized epigenetic modifications and their influence on progenitor proliferation and cortical neuron cell fate.
Collapse
Affiliation(s)
- Manal A Adam
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, United States
| | - Corey C Harwell
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
18
|
Pfleger J, Coleman RC, Ibetti J, Roy R, Kyriazis ID, Gao E, Drosatos K, Koch WJ. Genomic Binding Patterns of Forkhead Box Protein O1 Reveal Its Unique Role in Cardiac Hypertrophy. Circulation 2020; 142:882-898. [PMID: 32640834 DOI: 10.1161/circulationaha.120.046356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cardiac hypertrophic growth is mediated by robust changes in gene expression and changes that underlie the increase in cardiomyocyte size. The former is regulated by RNA polymerase II (pol II) de novo recruitment or loss; the latter involves incremental increases in the transcriptional elongation activity of pol II that is preassembled at the transcription start site. The differential regulation of these distinct processes by transcription factors remains unknown. Forkhead box protein O1 (FoxO1) is an insulin-sensitive transcription factor that is also regulated by hypertrophic stimuli in the heart. However, the scope of its gene regulation remains unexplored. METHODS To address this, we performed FoxO1 chromatin immunoprecipitation-deep sequencing in mouse hearts after 7 days of isoproterenol injections (3 mg·kg-1·mg-1), transverse aortic constriction, or vehicle injection/sham surgery. RESULTS Our data demonstrate increases in FoxO1 chromatin binding during cardiac hypertrophic growth, which positively correlate with extent of hypertrophy. To assess the role of FoxO1 on pol II dynamics and gene expression, the FoxO1 chromatin immunoprecipitation-deep sequencing results were aligned with those of pol II chromatin immunoprecipitation-deep sequencing across the chromosomal coordinates of sham- or transverse aortic constriction-operated mouse hearts. This uncovered that FoxO1 binds to the promoters of 60% of cardiac-expressed genes at baseline and 91% after transverse aortic constriction. FoxO1 binding is increased in genes regulated by pol II de novo recruitment, loss, or pause-release. In vitro, endothelin-1- and, in vivo, pressure overload-induced cardiomyocyte hypertrophic growth is prevented with FoxO1 knockdown or deletion, which was accompanied by reductions in inducible genes, including Comtd1 in vitro and Fstl1 and Uck2 in vivo. CONCLUSIONS Together, our data suggest that FoxO1 may mediate cardiac hypertrophic growth via regulation of pol II de novo recruitment and pause-release; the latter represents the majority (59%) of FoxO1-bound, pol II-regulated genes after pressure overload. These findings demonstrate the breadth of transcriptional regulation by FoxO1 during cardiac hypertrophy, information that is essential for its therapeutic targeting.
Collapse
Affiliation(s)
- Jessica Pfleger
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Ryan C Coleman
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Jessica Ibetti
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Rajika Roy
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Ioannis D Kyriazis
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Erhe Gao
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Konstantinos Drosatos
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
19
|
NandyMazumdar M, Yin S, Paranjapye A, Kerschner JL, Swahn H, Ge A, Leir SH, Harris A. Looping of upstream cis-regulatory elements is required for CFTR expression in human airway epithelial cells. Nucleic Acids Res 2020; 48:3513-3524. [PMID: 32095812 PMCID: PMC7144911 DOI: 10.1093/nar/gkaa089] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 01/14/2020] [Accepted: 02/03/2020] [Indexed: 12/14/2022] Open
Abstract
The CFTR gene lies within an invariant topologically associated domain (TAD) demarcated by CTCF and cohesin, but shows cell-type specific control mechanisms utilizing different cis-regulatory elements (CRE) within the TAD. Within the respiratory epithelium, more than one cell type expresses CFTR and the molecular mechanisms controlling its transcription are likely divergent between them. Here, we determine how two extragenic CREs that are prominent in epithelial cells in the lung, regulate expression of the gene. We showed earlier that these CREs, located at -44 and -35 kb upstream of the promoter, have strong cell-type-selective enhancer function. They are also responsive to inflammatory mediators and to oxidative stress, consistent with a key role in CF lung disease. Here, we use CRISPR/Cas9 technology to remove these CREs from the endogenous locus in human bronchial epithelial cells. Loss of either site extinguished CFTR expression and abolished long-range interactions between these sites and the gene promoter, suggesting non-redundant enhancers. The deletions also greatly reduced promoter interactions with the 5' TAD boundary. We show substantial recruitment of RNAPII to the -35 kb element and identify CEBPβ as a key activator of airway expression of CFTR, likely through occupancy at this CRE and the gene promoter.
Collapse
Affiliation(s)
- Monali NandyMazumdar
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44116, USA
| | - Shiyi Yin
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44116, USA
| | - Alekh Paranjapye
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44116, USA
| | - Jenny L Kerschner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44116, USA
| | - Hannah Swahn
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44116, USA
| | - Alex Ge
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44116, USA
| | - Shih-Hsing Leir
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44116, USA
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44116, USA
| |
Collapse
|
20
|
Chang CY, Hung JH, Huang LW, Li J, Fung KS, Kao CF, Chen L. Epigenetic Regulation of WNT3A Enhancer during Regeneration of Injured Cortical Neurons. Int J Mol Sci 2020; 21:ijms21051891. [PMID: 32164275 PMCID: PMC7084788 DOI: 10.3390/ijms21051891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022] Open
Abstract
Traumatic brain injury is known to reprogram the epigenome. Chromatin immunoprecipitation-sequencing of histone H3 lysine 27 acetylation (H3K27ac) and tri-methylation of histone H3 at lysine 4 (H3K4me3) marks was performed to address the transcriptional regulation of candidate regeneration-associated genes. In this study, we identify a novel enhancer region for induced WNT3A transcription during regeneration of injured cortical neurons. We further demonstrated an increased mono-methylation of histone H3 at lysine 4 (H3K4me1) modification at this enhancer concomitant with a topological interaction between sub-regions of this enhancer and with promoter of WNT3A gene. Together, this study reports a novel mechanism for WNT3A gene transcription and reveals a potential therapeutic intervention for neuronal regeneration.
Collapse
Affiliation(s)
- Chu-Yuan Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (K.S.F.)
| | - Jui-Hung Hung
- Department of Computer Science, National Chiao Tung University, Hsinchu 30010, Taiwan; (J.-H.H.); (J.L.)
| | - Liang-Wei Huang
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Joye Li
- Department of Computer Science, National Chiao Tung University, Hsinchu 30010, Taiwan; (J.-H.H.); (J.L.)
| | - Ka Shing Fung
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (K.S.F.)
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11574, Taiwan;
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (K.S.F.)
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: ; Tel.: +886-3-574-2775; Fax: +886-3-571-5934
| |
Collapse
|
21
|
Zizza P, Dinami R, Porru M, Cingolani C, Salvati E, Rizzo A, D'Angelo C, Petti E, Amoreo CA, Mottolese M, Sperduti I, Chambery A, Russo R, Ostano P, Chiorino G, Blandino G, Sacconi A, Cherfils-Vicini J, Leonetti C, Gilson E, Biroccio A. TRF2 positively regulates SULF2 expression increasing VEGF-A release and activity in tumor microenvironment. Nucleic Acids Res 2019; 47:3365-3382. [PMID: 30698737 PMCID: PMC6468246 DOI: 10.1093/nar/gkz041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/16/2019] [Indexed: 12/04/2022] Open
Abstract
The telomeric protein TRF2 is overexpressed in several human malignancies and contributes to tumorigenesis even though the molecular mechanism is not completely understood. By using a high-throughput approach based on the multiplexed Luminex X-MAP technology, we demonstrated that TRF2 dramatically affects VEGF-A level in the secretome of cancer cells, promoting endothelial cell-differentiation and angiogenesis. The pro-angiogenic effect of TRF2 is independent from its role in telomere capping. Instead, TRF2 binding to a distal regulatory element promotes the expression of SULF2, an endoglucosamine-6-sulfatase that impairs the VEGF-A association to the plasma membrane by inducing post-synthetic modification of heparan sulfate proteoglycans (HSPGs). Finally, we addressed the clinical relevance of our findings showing that TRF2/SULF2 expression is a worse prognostic biomarker in colorectal cancer (CRC) patients.
Collapse
Affiliation(s)
- Pasquale Zizza
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Roberto Dinami
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Manuela Porru
- SAFU, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Chiara Cingolani
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Erica Salvati
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Angela Rizzo
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Carmen D'Angelo
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Eleonora Petti
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Carla Azzurra Amoreo
- Pathology, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Marcella Mottolese
- Pathology, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Isabella Sperduti
- Department of Biostatistics Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Angela Chambery
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università della Campania Luigi Vanvitelli, via Vivaldi 43, 80100 Caserta
| | - Rosita Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università della Campania Luigi Vanvitelli, via Vivaldi 43, 80100 Caserta
| | - Paola Ostano
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900 Biella
| | - Giovanna Chiorino
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900 Biella
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Andrea Sacconi
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Julien Cherfils-Vicini
- Université Côte d'Azur, CNRS UMR 7284/INSERM U108, Institute for Research on Cancer and Aging, Nice (IRCAN), Medical School, Nice, France
| | - Carlo Leonetti
- SAFU, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | - Eric Gilson
- Université Côte d'Azur, CNRS UMR 7284/INSERM U108, Institute for Research on Cancer and Aging, Nice (IRCAN), Medical School, Nice, France.,Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Annamaria Biroccio
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| |
Collapse
|
22
|
Ha SD, Cho W, DeKoter RP, Kim SO. The transcription factor PU.1 mediates enhancer-promoter looping that is required for IL-1β eRNA and mRNA transcription in mouse melanoma and macrophage cell lines. J Biol Chem 2019; 294:17487-17500. [PMID: 31586032 DOI: 10.1074/jbc.ra119.010149] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/11/2019] [Indexed: 01/08/2023] Open
Abstract
The DNA-binding protein PU.1 is a myeloid lineage-determining and pioneering transcription factor due to its ability to bind "closed" genomic sites and maintain "open" chromatin state for myeloid lineage-specific genes. The precise mechanism of PU.1 in cell type-specific programming is yet to be elucidated. The melanoma cell line B16BL6, although it is nonmyeloid lineage, expressed Toll-like receptors and activated the transcription factor NF-κB upon stimulation by the bacterial cell wall component lipopolysaccharide. However, it did not produce cytokines, such as IL-1β mRNA. Ectopic PU.1 expression induced remodeling of a novel distal enhancer (located ∼10 kbp upstream of the IL-1β transcription start site), marked by nucleosome depletion, enhancer-promoter looping, and histone H3 lysine 27 acetylation (H3K27ac). PU.1 induced enhancer-promoter looping and H3K27ac through two distinct PU.1 regions. These PU.1-dependent events were independently required for subsequent signal-dependent and co-dependent events: NF-κB recruitment and further H3K27ac, both of which were required for enhancer RNA (eRNA) transcription. In murine macrophage RAW264.7 cells, these PU.1-dependent events were constitutively established and readily expressed eRNA and subsequently IL-1β mRNA by lipopolysaccharide stimulation. In summary, this study showed a sequence of epigenetic events in programming IL-1β transcription by the distal enhancer priming and eRNA production mediated by PU.1 and the signal-dependent transcription factor NF-κB.
Collapse
Affiliation(s)
- Soon-Duck Ha
- Department of Microbiology and Immunology and Infectious Diseases Research Group, Siebens-Drake Research Institute, University of Western Ontario, London, Ontario N6G 2V4, Canada
| | - Woohyun Cho
- Department of Microbiology and Immunology and Infectious Diseases Research Group, Siebens-Drake Research Institute, University of Western Ontario, London, Ontario N6G 2V4, Canada
| | - Rodney P DeKoter
- Department of Microbiology and Immunology and Infectious Diseases Research Group, Siebens-Drake Research Institute, University of Western Ontario, London, Ontario N6G 2V4, Canada
| | - Sung Ouk Kim
- Department of Microbiology and Immunology and Infectious Diseases Research Group, Siebens-Drake Research Institute, University of Western Ontario, London, Ontario N6G 2V4, Canada
| |
Collapse
|
23
|
Prestel M, Prell-Schicker C, Webb T, Malik R, Lindner B, Ziesch N, Rex-Haffner M, Röh S, Viturawong T, Lehm M, Mokry M, den Ruijter H, Haitjema S, Asare Y, Söllner F, Najafabadi MG, Aherrahrou R, Civelek M, Samani NJ, Mann M, Haffner C, Dichgans M. The Atherosclerosis Risk Variant rs2107595 Mediates Allele-Specific Transcriptional Regulation of HDAC9 via E2F3 and Rb1. Stroke 2019; 50:2651-2660. [PMID: 31500558 DOI: 10.1161/strokeaha.119.026112] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background and Purpose- Genome-wide association studies have identified the HDAC9 (histone deacetylase 9) gene region as a major risk locus for atherosclerotic stroke and coronary artery disease in humans. Previous results suggest a role of altered HDAC9 expression levels as the underlying disease mechanism. rs2107595, the lead single nucleotide polymorphism for stroke and coronary artery disease resides in noncoding DNA and colocalizes with histone modification marks suggestive of enhancer elements. Methods- To determine the mechanisms by which genetic variation at rs2107595 regulates HDAC9 expression and thus vascular risk we employed targeted resequencing, proteome-wide search for allele-specific nuclear binding partners, chromatin immunoprecipitation, genome-editing, reporter assays, circularized chromosome conformation capture, and gain- and loss-of-function experiments in cultured human cell lines and primary immune cells. Results- Targeted resequencing of the HDAC9 locus in patients with atherosclerotic stroke and controls supported candidacy of rs2107595 as the causative single nucleotide polymorphism. A proteomic search for nuclear binding partners revealed preferential binding of the E2F3/TFDP1/Rb1 complex (E2F transcription factor 3/transcription factor Dp-1/Retinoblastoma 1) to the rs2107595 common allele, consistent with the disruption of an E2F3 consensus site by the risk allele. Gain- and loss-of-function studies showed a regulatory effect of E2F/Rb proteins on HDAC9 expression. Compared with the common allele, the rs2107595 risk allele exhibited higher transcriptional capacity in luciferase assays and was associated with higher HDAC9 mRNA levels in primary macrophages and genome-edited Jurkat cells. Circularized chromosome conformation capture revealed a genomic interaction of the rs2107595 region with the HDAC9 promoter, which was stronger for the common allele as was the in vivo interaction with E2F3 and Rb1 determined by chromatin immunoprecipitation. Gain-of-function experiments in isogenic Jurkat cells demonstrated a key role of E2F3 in mediating rs2107595-dependent transcriptional regulation of HDAC9. Conclusions- Collectively, our findings imply allele-specific transcriptional regulation of HDAC9 via E2F3 and Rb1 as a major mechanism mediating vascular risk at rs2107595.
Collapse
Affiliation(s)
- Matthias Prestel
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
| | - Caroline Prell-Schicker
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
| | - Tom Webb
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (T.W., M.G.N., N.J.S.)
| | - Rainer Malik
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
| | - Barbara Lindner
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
| | - Natalie Ziesch
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
| | - Monika Rex-Haffner
- Department of Translational Research in Psychiatry, Max-Planck-Institute for Psychiatry, Germany (M.R.H., S.R.)
| | - Simone Röh
- Department of Translational Research in Psychiatry, Max-Planck-Institute for Psychiatry, Germany (M.R.H., S.R.)
| | - Thanatip Viturawong
- Department of Proteomics and Signal Transduction, Max-Planck-Institute for Biochemistry, Martinsried, Germany (T.V., M.L., M. Mann)
| | - Manuel Lehm
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
- Department of Proteomics and Signal Transduction, Max-Planck-Institute for Biochemistry, Martinsried, Germany (T.V., M.L., M. Mann)
- Abteilung für Diagnostische und Interventionelle Neuroradiologie, Klinikum rechts der Isar, Munich, Germany (M.L.)
| | - Michal Mokry
- Department of Pediatrics (M. Mokry), University Medical Center Utrecht, the Netherlands
| | - Hester den Ruijter
- Laboratory of Experimental Cardiology (H.d.R., S.H.), University Medical Center Utrecht, the Netherlands
| | - Saskia Haitjema
- Laboratory of Experimental Cardiology (H.d.R., S.H.), University Medical Center Utrecht, the Netherlands
| | - Yaw Asare
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
| | - Flavia Söllner
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
- Department of Physiological Chemistry, Biomedical Center Munich, Ludwig-Maximilians-Universität München, Germany (F.S.)
| | - Maryam Ghaderi Najafabadi
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (T.W., M.G.N., N.J.S.)
| | - Rédouane Aherrahrou
- Center for Public Health Genomics, Department of Biomedical Engineering, University of Virginia, Charlottesville, (R.A., M.C.)
| | - Mete Civelek
- Center for Public Health Genomics, Department of Biomedical Engineering, University of Virginia, Charlottesville, (R.A., M.C.)
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (T.W., M.G.N., N.J.S.)
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck-Institute for Biochemistry, Martinsried, Germany (T.V., M.L., M. Mann)
| | - Christof Haffner
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
| | - Martin Dichgans
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Germany (M.P., C.P.S., R.M., B.L., N.Z., M.L., Y.A., F.S., C.H., M.D.)
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| |
Collapse
|
24
|
Dysregulated Transcriptional Control in Prostate Cancer. Int J Mol Sci 2019; 20:ijms20122883. [PMID: 31200487 PMCID: PMC6627928 DOI: 10.3390/ijms20122883] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/24/2022] Open
Abstract
Recent advances in whole-genome and transcriptome sequencing of prostate cancer at different stages indicate that a large number of mutations found in tumors are present in non-protein coding regions of the genome and lead to dysregulated gene expression. Single nucleotide variations and small mutations affecting the recruitment of transcription factor complexes to DNA regulatory elements are observed in an increasing number of cases. Genomic rearrangements may position coding regions under the novel control of regulatory elements, as exemplified by the TMPRSS2-ERG fusion and the amplified enhancer identified upstream of the androgen receptor (AR) gene. Super-enhancers are increasingly found to play important roles in aberrant oncogenic transcription. Several players involved in these processes are currently being evaluated as drug targets and may represent new vulnerabilities that can be exploited for prostate cancer treatment. They include factors involved in enhancer and super-enhancer function such as bromodomain proteins and cyclin-dependent kinases. In addition, non-coding RNAs with an important gene regulatory role are being explored. The rapid progress made in understanding the influence of the non-coding part of the genome and of transcription dysregulation in prostate cancer could pave the way for the identification of novel treatment paradigms for the benefit of patients.
Collapse
|
25
|
Scheidegger A, Dunn CJ, Samarakkody A, Koney NKK, Perley D, Saha RN, Nechaev S. Genome-wide RNA pol II initiation and pausing in neural progenitors of the rat. BMC Genomics 2019; 20:477. [PMID: 31185909 PMCID: PMC6558777 DOI: 10.1186/s12864-019-5829-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/23/2019] [Indexed: 12/29/2022] Open
Abstract
Background Global RNA sequencing technologies have revealed widespread RNA polymerase II (Pol II) transcription outside of gene promoters. Small 5′-capped RNA sequencing (Start-seq) originally developed for the detection of promoter-proximal Pol II pausing has helped improve annotation of Transcription Start Sites (TSSs) of genes as well as identification of non-genic regulatory elements. However, apart from the most well studied genomes of human and mouse, mammalian transcription has not been profiled with sufficiently high precision. Results We prepared and sequenced Start-seq libraries from rat (Rattus norgevicus) primary neural progenitor cells. Over 48 million uniquely mappable reads from two independent biological replicates allowed us to define the TSSs of 7365 known genes in the rn6 genome, reannotating 2503 TSSs by more than 5 base pairs, characterize promoter-associated antisense transcription, and profile Pol II pausing. By combining TSS data with polyA-selected RNA sequencing, we also identified thousands of potential new genes producing stable RNA as well as non-genic transcripts representing possible regulatory elements. Conclusions Our study has produced the first Start-seq dataset for the rat. Apart from profiling transcription initiation, our data reaffirm the prevalence of Pol II pausing across the rat genome and indicate conservation of pausing mechanisms across metazoan genomes. We suggest that pausing location, at least in mammals, is constrained by a distance from initiation of transcription, whether it occurs at or outside of a gene promoter. Abundant antisense transcription initiation around protein coding genes indicates that Pol II recruited to the vicinity of a promoter is distributed to available start sites of transcription at either DNA strand. Transcriptome profiling of neural progenitors presented here will facilitate further studies of other rat cell types as well as other organisms. Electronic supplementary material The online version of this article (10.1186/s12864-019-5829-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adam Scheidegger
- Department of Biomedical Sciences, University of North Dakota School of Medicine, Grand Forks, ND, 58202, USA.,Present address: Omega Therapeutics, Cambridge, MA, 02139, USA
| | - Carissa J Dunn
- Molecular and Cell Biology Department, School of Natural Sciences, University of California Merced, Merced, CA, 95343, USA
| | - Ann Samarakkody
- Department of Biomedical Sciences, University of North Dakota School of Medicine, Grand Forks, ND, 58202, USA.,Present address: Department of Pediatric Hematology-Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Nii Koney-Kwaku Koney
- Department of Biomedical Sciences, University of North Dakota School of Medicine, Grand Forks, ND, 58202, USA
| | - Danielle Perley
- Department of Biomedical Sciences, University of North Dakota School of Medicine, Grand Forks, ND, 58202, USA
| | - Ramendra N Saha
- Molecular and Cell Biology Department, School of Natural Sciences, University of California Merced, Merced, CA, 95343, USA
| | - Sergei Nechaev
- Department of Biomedical Sciences, University of North Dakota School of Medicine, Grand Forks, ND, 58202, USA.
| |
Collapse
|
26
|
Herre M, Korb E. The chromatin landscape of neuronal plasticity. Curr Opin Neurobiol 2019; 59:79-86. [PMID: 31174107 DOI: 10.1016/j.conb.2019.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/18/2019] [Indexed: 01/27/2023]
Abstract
Examining the links between neuronal activity, transcriptional output, and synaptic function offers unique insights into how neurons adapt to changing environments and form memories. Epigenetic markers, such as DNA methylation and histone modifications, have been implicated in the formation of not only cellular memories such as cell fate, but also memories of experience at the organismal level. Here, we review recent advances in chromatin regulation that contribute to synaptic plasticity and drive adaptive behaviors through dynamic and precise regulation of transcription output in neurons. We discuss chromatin-associated proteins, histone variant proteins, the contribution of cis-regulatory elements and their interaction with histone modifications, and how these mechanisms are integrated into distinct behavior and environmental response paradigms.
Collapse
Affiliation(s)
- Margaret Herre
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Erica Korb
- Department of Genetics, Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
27
|
Machado Xavier A, Belhocine S, Gosselin D. Essential contributions of enhancer genomic regulatory elements to microglial cell identity and functions. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2019; 11:e1449. [PMID: 31016893 DOI: 10.1002/wsbm.1449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 11/09/2022]
Abstract
Microglia are the specialized macrophages of the brain and play essential roles in ensuring its proper functioning. Accumulating evidence suggests that these cells coordinate the inflammatory response that accompanies various clinical brain conditions, including neurodegenerative diseases and psychiatric disorders. Therefore, investigating the functions of these cells and how these are regulated have become important areas of research in neuroscience over the past decade. In this regards, recent efforts to characterize the epigenomic mechanisms underlying microglial gene transcription have provided significant insights into the mechanisms that control the ontogeny and the cellular competences of microglia. In particular, these studies have established that a substantial proportion of the microglial repertoire of promoter-distal genomic regulatory elements, or enhancers, is relatively specific to these cells compared to other tissue-resident macrophages. Notably, this specificity is under the regulation of factors present in the brain that modulate activity of target axes of signaling pathways-transcription factors in microglia. Thus, the microglial enhancer repertoire is highly responsive to perturbations in the cerebral tissue microenvironment and this responsiveness has profound implications on the activity of these cells in brain diseases. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Models of Systems Properties and Processes > Mechanistic Models Biological Mechanisms > Cell Fates Developmental Biology > Lineages.
Collapse
Affiliation(s)
- André Machado Xavier
- CHU de Québec Research Center - Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Sarah Belhocine
- CHU de Québec Research Center - Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - David Gosselin
- CHU de Québec Research Center - Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
28
|
Sulaiman SA, Muhsin NIA, Jamal R. Regulatory Non-coding RNAs Network in Non-alcoholic Fatty Liver Disease. Front Physiol 2019; 10:279. [PMID: 30941061 PMCID: PMC6433939 DOI: 10.3389/fphys.2019.00279] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/04/2019] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) spectrum comprises simple steatosis and non-alcoholic steatohepatitis (NASH) that can lead to fibrosis and cirrhosis. The patients usually have no history of excessive alcohol consumption and other etiologies that can cause fatty liver. Understanding of the pathophysiology of NAFLD has revealed that non-coding RNAs (ncRNAs) play significant roles in modulating the disease susceptibility, pathogenesis and progression. Currently, the ncRNAs are grouped according to their sizes and their regulatory or housekeeping functions. Each of these ncRNAs has a wide range of involvement in the regulation of the genes and biological pathways. Here, we briefly review the current literature the regulatory ncRNAs in NAFLD pathogenesis and progression, mainly the microRNAs, long non-coding RNAs and circular RNAs. We also discuss the co-regulatory functions and interactions between these ncRNAs in modulating the disease pathogenesis. Elucidation of ncRNAs in NAFLD may facilitate the identification of early diagnostic biomarkers and development of therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
- Siti Aishah Sulaiman
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | | |
Collapse
|
29
|
Agirre X, Meydan C, Jiang Y, Garate L, Doane AS, Li Z, Verma A, Paiva B, Martín-Subero JI, Elemento O, Mason CE, Prosper F, Melnick A. Long non-coding RNAs discriminate the stages and gene regulatory states of human humoral immune response. Nat Commun 2019; 10:821. [PMID: 30778059 PMCID: PMC6379396 DOI: 10.1038/s41467-019-08679-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
lncRNAs make up a majority of the human transcriptome and have key regulatory functions. Here we perform unbiased de novo annotation of transcripts expressed during the human humoral immune response to find 30% of the human genome transcribed during this process, yet 58% of these transcripts manifest striking differential expression, indicating an lncRNA phylogenetic relationship among cell types that is more robust than that of coding genes. We provide an atlas of lncRNAs in naive and GC B-cells that indicates their partition into ten functionally categories based on chromatin features, DNase hypersensitivity and transcription factor localization, defining lncRNAs classes such as enhancer-RNAs (eRNA), bivalent-lncRNAs, and CTCF-associated, among others. Specifically, eRNAs are transcribed in 8.6% of regular enhancers and 36.5% of super enhancers, and are associated with coding genes that participate in critical immune regulatory pathways, while plasma cells have uniquely high levels of circular-RNAs accounted for by and reflecting the combinatorial clonal state of the Immunoglobulin loci. Long non-coding RNAs (lncRNA) constitute a large fraction of human transcriptome, and are reported, individually, for context-specific regulatory functions. Here the authors expand our understanding by providing a systemic, unbiased annotation of lncRNA to establish an atlas of lncRNA landscape during the induction of human humoral immune responses.
Collapse
Affiliation(s)
- Xabier Agirre
- Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 10021, USA. .,Division of Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, IDISNA, Ciberonc, Pamplona, 31008, Spain.
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA.,The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Yanwen Jiang
- Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Leire Garate
- Division of Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, IDISNA, Ciberonc, Pamplona, 31008, Spain.,Department of Hematology, Clínica Universidad de Navarra, IDISNA, Pamplona, 31008, Spain
| | - Ashley S Doane
- The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Zhuoning Li
- Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Akanksha Verma
- The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Bruno Paiva
- Division of Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, IDISNA, Ciberonc, Pamplona, 31008, Spain
| | - José I Martín-Subero
- Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, 08036, Spain
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA.,The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA. .,The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA. .,The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| | - Felipe Prosper
- Division of Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, IDISNA, Ciberonc, Pamplona, 31008, Spain. .,Department of Hematology, Clínica Universidad de Navarra, IDISNA, Pamplona, 31008, Spain.
| | - Ari Melnick
- Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
30
|
Cremer M, Cremer T. Nuclear compartmentalization, dynamics, and function of regulatory DNA sequences. Genes Chromosomes Cancer 2019; 58:427-436. [DOI: 10.1002/gcc.22714] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/23/2018] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Affiliation(s)
- Marion Cremer
- Biocenter, Department Biology II; Ludwig Maximilians-Universität (LMU Munich); Munich Germany
| | - Thomas Cremer
- Biocenter, Department Biology II; Ludwig Maximilians-Universität (LMU Munich); Munich Germany
| |
Collapse
|
31
|
Potter KC, Wang J, Schaller GE, Kieber JJ. Cytokinin modulates context-dependent chromatin accessibility through the type-B response regulators. NATURE PLANTS 2018; 4:1102-1111. [PMID: 30420712 DOI: 10.1038/s41477-018-0290-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 09/26/2018] [Indexed: 05/18/2023]
Abstract
The phytohormone cytokinin regulates diverse aspects of plant growth and development, probably through context-dependent transcriptional regulation that relies on a dynamic interplay between regulatory proteins and chromatin. We employed the assay for transposase accessible chromatin with sequencing to profile changes in the chromatin landscape of Arabidopsis roots and shoots in response to cytokinin. Our results reveal differentially accessible chromatin regions indicative of dynamic regulation in response to cytokinin. These changes in chromatin occur preferentially upstream of cytokinin-regulated genes. The changes also largely overlap with binding sites for the type-B ARABIDOPSIS RESPONSE REGULATORS (ARRs), transcription factors that mediate the primary response to cytokinin. Furthermore, the type-B ARRs were found to be necessary for the changes in chromatin state in response to cytokinin. Last, we identified context-dependent responses by comparing root and shoot profiles. This study provides new insight into the dynamics between cytokinin and chromatin with regard to directing transcriptional programmes and how cytokinin mediates its pleiotropic effects.
Collapse
Affiliation(s)
- Kevin C Potter
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Judy Wang
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
| | - G Eric Schaller
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Joseph J Kieber
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
32
|
Blokhin I, Khorkova O, Hsiao J, Wahlestedt C. Developments in lncRNA drug discovery: where are we heading? Expert Opin Drug Discov 2018; 13:837-849. [PMID: 30078338 DOI: 10.1080/17460441.2018.1501024] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The central dogma of molecular biology, which states that the only role of long RNA transcripts is to convey information from gene to protein, was brought into question in recent years due to discovery of the extensive presence and complex roles of long noncoding RNAs (lncRNAs). Furthermore, lncRNAs were found to be involved in pathogenesis of multiple diseases and thus represent a new class of therapeutic targets. Translational efforts in the lncRNA field have been augmented by progress in optimizing the chemistry and delivery platforms of lncRNA-targeting modalities, including oligonucleotide-based drugs and CRISPR-Cas9. Areas covered: This review covers the current advances in characterizing diversity and biological functions of lncRNA focusing on their therapeutic potential in selected therapeutic areas. Expert opinion: Due to accelerating parallel progress in lncRNA biology and lncRNA-compatible therapeutic modalities, it is likely that lncRNA-dependent mechanisms of pathogenesis will soon be targeted in various disorders, including neurological, psychiatric, cardiovascular, infectious diseases, and cancer. Significant efforts, however, are still required to better understand the biology of both lncRNAs and lncRNA-targeting drugs. Further work is needed in the areas of lncRNA nomenclature, database representation, intra/interfield communication, and education of the community at large.
Collapse
Affiliation(s)
- Ilya Blokhin
- a Center for Therapeutic Innovation and Department of Psychiatry and Behavioral Sciences , University of Miami Miller School of Medicine , Miami , FL , USA
| | | | | | - Claes Wahlestedt
- a Center for Therapeutic Innovation and Department of Psychiatry and Behavioral Sciences , University of Miami Miller School of Medicine , Miami , FL , USA
| |
Collapse
|
33
|
Gottesfeld JM, Carey MF. Introduction to the Thematic Minireview Series: Chromatin and transcription. J Biol Chem 2018; 293:13775-13777. [PMID: 30068547 DOI: 10.1074/jbc.tm118.004544] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Regulation of transcription in eukaryotic cells is a dynamic interplay between chromatin structure and recruitment of a plethora of transcription factors to enhancers, upstream activator sequences, and proximal promoter elements. These factors serve to recruit RNA polymerase to the core promoter for productive transcription. In this Thematic Minireview Series on chromatin and transcription, five reviews summarize current knowledge of diverse aspects of transcriptional regulation and the role of chromatin structure in transcription and development.
Collapse
Affiliation(s)
- Joel M Gottesfeld
- From the Departments of Molecular Medicine and Chemistry, The Scripps Research Institute, La, Jolla, California 92037 and
| | - Michael F Carey
- the Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| |
Collapse
|
34
|
Webber JL, Zhang J, Massey A, Sanchez-Luege N, Rebay I. Collaborative repressive action of the antagonistic ETS transcription factors Pointed and Yan fine-tunes gene expression to confer robustness in Drosophila. Development 2018; 145:dev.165985. [PMID: 29848501 DOI: 10.1242/dev.165985] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/17/2018] [Indexed: 01/29/2023]
Abstract
The acquisition of cellular identity during development depends on precise spatiotemporal regulation of gene expression, with combinatorial interactions between transcription factors, accessory proteins and the basal transcription machinery together translating complex signaling inputs into appropriate gene expression outputs. The opposing repressive and activating inputs of the Drosophila ETS family transcription factors Yan and Pointed orchestrate numerous cell fate transitions downstream of receptor tyrosine kinase signaling, providing one of the premier systems for studying this process. Current models describe the differentiative transition as a switch from Yan-mediated repression to Pointed-mediated activation of common target genes. We describe here a new layer of regulation whereby Yan and Pointed co-occupy regulatory elements to repress gene expression in a coordinated manner, with Pointed being unexpectedly required for the genome-wide occupancy of both Yan and the co-repressor Groucho. Using even skipped as a test-case, synergistic genetic interactions between Pointed, Groucho, Yan and components of the RNA polymerase II pausing machinery suggest that Pointed integrates multiple scales of repressive regulation to confer robustness. We speculate that this mechanism may be used broadly to fine-tune the expression of many genes crucial for development.
Collapse
Affiliation(s)
- Jemma L Webber
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Jie Zhang
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Alex Massey
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Nicelio Sanchez-Luege
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Ilaria Rebay
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
35
|
Abstract
Transcriptional enhancers constitute a subclass of regulatory elements that facilitate transcription. Such regions are generally organized by short stretches of DNA enriched in transcription factor-binding sites but also can include very large regions containing clusters of enhancers, termed super-enhancers. These regions increase the probability or the rate (or both) of transcription generally in
cis and sometimes over very long distances by altering chromatin states and the activity of Pol II machinery at promoters. Although enhancers were discovered almost four decades ago, their inner workings remain enigmatic. One important opening into the underlying principle has been provided by observations that enhancers make physical contacts with their target promoters to facilitate the loading of the RNA polymerase complex. However, very little is known about how such chromatin loops are regulated and how they govern transcription in the three-dimensional context of the nuclear architecture. Here, we present current themes of how enhancers may boost gene expression in three dimensions and we identify currently unresolved key questions.
Collapse
Affiliation(s)
- Anita Göndör
- Department of Oncology and Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Rolf Ohlsson
- Department of Oncology and Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
36
|
Current Advances on the Important Roles of Enhancer RNAs in Gene Regulation and Cancer. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2405351. [PMID: 29951530 PMCID: PMC5987348 DOI: 10.1155/2018/2405351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/05/2018] [Accepted: 04/19/2018] [Indexed: 12/27/2022]
Abstract
Revealing the gene regulation networks governing cancer initiation and development is necessary while it remains uncompleted. In recent years, enhancers have been reported to be widely transcribed, resulting in the generation of enhancer RNAs (eRNAs). Previous studies have reported that eRNAs are a subclass of long noncoding RNAs (lncRNAs), which play a critical role in gene regulation and cancer development. These eRNAs can promote enhancer-promoter (E-P) looping formation by binding to other protein factors or propel expression of downstream protein-coding gene. In this review, we have focused on the characteristics of eRNAs and illustrated the biological function and potential mechanism of eRNAs in regulating gene expression and cancer development.
Collapse
|