1
|
Liu C, Zhang C, Glatt SJ. Psychiatric Genomics 2025: State of the Art and the Path Forward. Psychiatr Clin North Am 2025; 48:217-240. [PMID: 40348414 DOI: 10.1016/j.psc.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Psychiatric genetics has evolved from candidate-gene studies to whole-genome sequencing efforts. With hundreds of disease-associated loci now identified, functional interpretation of the associated loci becomes the critical next step toward translational applications. The article discusses achievements, challenges, and opportunities in psychiatric genomics associated with complexity and heterogeneity. Brain expression quantitative trait loci, single-cell ribonucleic acid-sequence, and functional genomics technologies are highlighted. It also covers newly developed techniques with improved spatiotemporal resolution, quality and sensitivity, coupled with advanced analytical methods and artificial intelligence. The power of collaborative research and inclusion of diverse populations will ensure a bright future for precision psychiatry.
Collapse
Affiliation(s)
- Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA.
| | - Chunling Zhang
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| | - Stephen J Glatt
- Department of Psychiatry, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| |
Collapse
|
2
|
Beernink BM, Vogel JP, Lei L. Enhancers in Plant Development, Adaptation and Evolution. PLANT & CELL PHYSIOLOGY 2025; 66:461-476. [PMID: 39412125 DOI: 10.1093/pcp/pcae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/13/2024] [Accepted: 10/09/2024] [Indexed: 05/18/2025]
Abstract
Understanding plant responses to developmental and environmental cues is crucial for studying morphological divergence and local adaptation. Gene expression changes, governed by cis-regulatory modules (CRMs) including enhancers, are a major source of plant phenotypic variation. However, while genome-wide approaches have revealed thousands of putative enhancers in mammals, far fewer have been identified and functionally characterized in plants. This review provides an overview of how enhancers function to control gene regulation, methods to predict DNA sequences that may have enhancer activity, methods utilized to functionally validate enhancers and the current knowledge of enhancers in plants, including how they impact plant development, response to environment and evolutionary adaptation.
Collapse
Affiliation(s)
- Bliss M Beernink
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - John P Vogel
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Li Lei
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| |
Collapse
|
3
|
Barbadilla-Martínez L, Klaassen N, van Steensel B, de Ridder J. Predicting gene expression from DNA sequence using deep learning models. Nat Rev Genet 2025:10.1038/s41576-025-00841-2. [PMID: 40360798 DOI: 10.1038/s41576-025-00841-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2025] [Indexed: 05/15/2025]
Abstract
Transcription of genes is regulated by DNA elements such as promoters and enhancers, the activity of which are in turn controlled by many transcription factors. Owing to the highly complex combinatorial logic involved, it has been difficult to construct computational models that predict gene activity from DNA sequence. Recent advances in deep learning techniques applied to data from epigenome mapping and high-throughput reporter assays have made substantial progress towards addressing this complexity. Such models can capture the regulatory grammar with remarkable accuracy and show great promise in predicting the effects of non-coding variants, uncovering detailed molecular mechanisms of gene regulation and designing synthetic regulatory elements for biotechnology. Here, we discuss the principles of these approaches, the types of training data sets that are available and the strengths and limitations of different approaches.
Collapse
Affiliation(s)
- Lucía Barbadilla-Martínez
- Oncode Institute, Utrecht, The Netherlands
- Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Noud Klaassen
- Oncode Institute, Utrecht, The Netherlands
- Division of Molecular Genetics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bas van Steensel
- Oncode Institute, Utrecht, The Netherlands.
- Division of Molecular Genetics, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Jeroen de Ridder
- Oncode Institute, Utrecht, The Netherlands.
- Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
4
|
Williams RM. Leveraging Chicken Embryos for Studying Human Enhancers. Dev Biol 2025:S0012-1606(25)00132-0. [PMID: 40368318 DOI: 10.1016/j.ydbio.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 04/30/2025] [Accepted: 05/12/2025] [Indexed: 05/16/2025]
Abstract
The dynamic activity of complex gene regulatory networks stands at the core of all cellular functions that define cell identity and behaviour. Gene regulatory networks comprise transcriptional enhancers, acted upon by cell-specific transcription factors to control gene expression in a spatial and temporal specific manner. Enhancers are found in the non-coding genome; pathogenic variants can disrupt enhancer activity and lead to disease. Correlating non-coding variants with aberrant enhancer activity remains a significant challenge. Due to their clinical significance, there is a longstanding interest in understanding enhancer function during early embryogenesis. With the onset of the omics era, it is now feasible to identify putative tissue-specific enhancers from epigenome data. However, such predictions in vivo require validation. The early stages of chick embryogenesis closely parallel those of human, offering an accessible in vivo model in which to assess the activity of putative human enhancer sequences. This review explores the unique advantages and recent advancements in employing chicken embryos to elucidate the activity of human transcriptional enhancers and the potential implications of these findings in human disease.
Collapse
Affiliation(s)
- Ruth M Williams
- University of Manchester, Faculty of Biology, Medicine and Health, Michael Smith building, Oxford Road, Manchester, United Kingdom.
| |
Collapse
|
5
|
Bevan MW, Messerer M, Gundlach H, Kamal N, Hall A, Spannagl M, Mayer KFX. Integrating Arabidopsis and crop species gene discovery for crop improvement. THE PLANT CELL 2025; 37:koaf087. [PMID: 40251981 PMCID: PMC12079385 DOI: 10.1093/plcell/koaf087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 03/19/2025] [Indexed: 04/21/2025]
Abstract
Genome sequence assemblies form a durable and precise framework supporting nearly all areas of biological research, including evolutionary biology, taxonomy and conservation science, pathogen population diversity, crop domestication, and biochemistry. In the early days of plant genomics, resources were limited to a handful of tractable genomes, leading to a tension between focus on discovering mechanisms in experimental species such as Arabidopsis thaliana (Arabidopsis) and on trait analyses in crop species. This tension arose from challenges in translating knowledge of gene function across the large evolutionary distances between Arabidopsis and diverse crop species in the absence of comparative genome support. For some time, these clashing interests influenced funding priorities in plant science that limited both the acquisition of knowledge of mechanisms in Arabidopsis and the timely development of the capacity of crop science to incorporate emerging knowledge of genes and their mechanisms. In this review we show how advances in genomics analysis technologies are revealing a high degree of conservation of molecular mechanisms between evolutionarily distant plant species. This progress is bridging the model-species-to-crop barrier, resulting in ever-increasing unification of plant science that is now accelerating progress in understanding mechanisms underlying diverse traits in crops and improving their performance. We lay out some examples of important priorities and outcomes arising from these new opportunities.
Collapse
Affiliation(s)
- Michael W Bevan
- Cell and Developmental Biology Dept, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, UK
| | - Maxim Messerer
- Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Heidrun Gundlach
- Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Nadia Kamal
- Technical University München, Graduate Centre of Life Sciences, Alte Akademie 8a, 85354 Freising, Germany
| | - Anthony Hall
- The Earlham Institute, Norwich Research Park, Norwich NR4 7UH, UK
| | - Manuel Spannagl
- Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Klaus F X Mayer
- Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| |
Collapse
|
6
|
Psatha N, Sova P, Georgolopoulos G, Paschoudi K, Iwata M, Bloom J, Ulyanova T, Wang H, Kirtsou A, Vasiloudis NI, Wilken MS, Stamatoyannopoulos JA, Yannaki E, Papayanopoulou T, Stamatoyannopoulos G, Vierstra J. Large-scale discovery of potent, compact and erythroid specific enhancers for gene therapy vectors. Nat Commun 2025; 16:4325. [PMID: 40346084 PMCID: PMC12064758 DOI: 10.1038/s41467-025-59235-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
Gene expression during cell development and differentiation is orchestrated by distal regulatory elements that precisely modulate cell selective gene activity. Gene therapy vectors leverage these elements for precise spatiotemporal transgene expression. Here, we develop a one-shot approach to screen candidate regulatory sequences from large-scale epigenomics data for programmable transgene expression within gene therapy viral vectors. We assess a library of 15,000 short sequences derived from developmentally active elements during erythropoiesis using a clinically relevant reporter vector. These elements display a gradient of transcriptional enhancer activity in erythroid cells, with high cell type restriction and developmental stage specificity. Finally, replacing the canonical β-globin μLCR with a compact enhancer in a β-thalassemia lentiviral vector successfully corrects the thalassemic phenotype in patient-derived hematopoietic and stem and progenitor cells (HSPCs), while increasing viral titers and cell transducibility. Our approach provides further insights into enhancer biology with wider implications for human gene therapy.
Collapse
Affiliation(s)
- Nikoletta Psatha
- Altius Institute for Biomedical Sciences, Seattle, WA, USA.
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Pavel Sova
- Altius Institute for Biomedical Sciences, Seattle, WA, USA
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Kiriaki Paschoudi
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Gene and Cell Therapy Center, Hematology Clinic-BMTU, "George Papanikolaou" Hospital, Thessaloniki, Greece
| | - Mineo Iwata
- Altius Institute for Biomedical Sciences, Seattle, WA, USA
| | - Jordan Bloom
- Altius Institute for Biomedical Sciences, Seattle, WA, USA
| | - Tatyana Ulyanova
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Hao Wang
- Altius Institute for Biomedical Sciences, Seattle, WA, USA
| | - Alexandra Kirtsou
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Gene and Cell Therapy Center, Hematology Clinic-BMTU, "George Papanikolaou" Hospital, Thessaloniki, Greece
| | - Ninos-Ioannis Vasiloudis
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Gene and Cell Therapy Center, Hematology Clinic-BMTU, "George Papanikolaou" Hospital, Thessaloniki, Greece
| | | | | | - Evangelia Yannaki
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
- Gene and Cell Therapy Center, Hematology Clinic-BMTU, "George Papanikolaou" Hospital, Thessaloniki, Greece
| | - Thalia Papayanopoulou
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Jeff Vierstra
- Altius Institute for Biomedical Sciences, Seattle, WA, USA.
| |
Collapse
|
7
|
Dincer TU, Ernst J. ChromActivity: integrative epigenomic and functional characterization assay based annotation of regulatory activity across diverse human cell types. Genome Biol 2025; 26:123. [PMID: 40346707 PMCID: PMC12063466 DOI: 10.1186/s13059-025-03579-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
We introduce ChromActivity, a computational framework for predicting and annotating regulatory activity across the genome through integration of multiple epigenomic maps and various functional characterization datasets. ChromActivity generates genomewide predictions of regulatory activity associated with each functional characterization dataset across many cell types based on available epigenomic data. It then for each cell type produces ChromScoreHMM genome annotations based on the combinatorial and spatial patterns within these predictions and ChromScore tracks of overall predicted regulatory activity. ChromActivity provides a resource for analyzing and interpreting the human regulatory genome across diverse cell types.
Collapse
Affiliation(s)
- Tevfik Umut Dincer
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jason Ernst
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Computer Science Department, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
8
|
Butterfield GL, Reisman SJ, Iglesias N, Gersbach CA. Gene regulation technologies for gene and cell therapy. Mol Ther 2025; 33:2104-2122. [PMID: 40195118 DOI: 10.1016/j.ymthe.2025.04.004] [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: 03/17/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025] Open
Abstract
Gene therapy stands at the forefront of medical innovation, offering unique potential to treat the underlying causes of genetic disorders and broadly enable regenerative medicine. However, unregulated production of therapeutic genes can lead to decreased clinical utility due to various complications. Thus, many technologies for controlled gene expression are under development, including regulated transgenes, modulation of endogenous genes to leverage native biological regulation, mapping and repurposing of transcriptional regulatory networks, and engineered systems that dynamically react to cell state changes. Transformative therapies enabled by advances in tissue-specific promoters, inducible systems, and targeted delivery have already entered clinical testing and demonstrated significantly improved specificity and efficacy. This review highlights next-generation technologies under development to expand the reach of gene therapies by enabling precise modulation of gene expression. These technologies, including epigenome editing, antisense oligonucleotides, RNA editing, transcription factor-mediated reprogramming, and synthetic genetic circuits, have the potential to provide powerful control over cellular functions. Despite these remarkable achievements, challenges remain in optimizing delivery, minimizing off-target effects, and addressing regulatory hurdles. However, the ongoing integration of biological insights with engineering innovations promises to expand the potential for gene therapy, offering hope for treating not only rare genetic disorders but also complex multifactorial diseases.
Collapse
Affiliation(s)
- Gabriel L Butterfield
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Samuel J Reisman
- Department of Cell Biology, Duke University, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Nahid Iglesias
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Department of Cell Biology, Duke University, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
9
|
Frömel R, Rühle J, Bernal Martinez A, Szu-Tu C, Pacheco Pastor F, Martinez-Corral R, Velten L. Design principles of cell-state-specific enhancers in hematopoiesis. Cell 2025:S0092-8674(25)00449-0. [PMID: 40345201 DOI: 10.1016/j.cell.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 02/02/2025] [Accepted: 04/10/2025] [Indexed: 05/11/2025]
Abstract
During cellular differentiation, enhancers transform overlapping gradients of transcription factors (TFs) to highly specific gene expression patterns. However, the vast complexity of regulatory DNA impedes the identification of the underlying cis-regulatory rules. Here, we characterized 64,400 fully synthetic DNA sequences to bottom-up dissect design principles of cell-state-specific enhancers in the context of the differentiation of blood stem cells to seven myeloid lineages. Focusing on binding sites for 38 TFs and their pairwise interactions, we found that identical sites displayed both repressive and activating function as a consequence of cell state, site combinatorics, or simply predicted occupancy of a TF on an enhancer. Surprisingly, combinations of activating sites frequently neutralized one another or gained repressive function. These negative synergies convert quantitative imbalances in TF expression into binary activity patterns. We exploit this principle to automatically create enhancers with specificity to user-defined combinations of hematopoietic progenitor cell states from scratch.
Collapse
Affiliation(s)
- Robert Frömel
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Julia Rühle
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Aina Bernal Martinez
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Chelsea Szu-Tu
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Felix Pacheco Pastor
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Rosa Martinez-Corral
- CRG (Barcelona Collaboratorium for Modelling and Predictive Biology), Dr. Aiguader 88, Barcelona 08003, Spain
| | - Lars Velten
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
10
|
Jajodia A, Mishra A, Doni Jayavelu N, Lambert K, Moss N, Yang Z, Cerosaletti K, Buckner JH, Hawkins RD. Functional dissection of noncoding variants associated with rheumatoid arthritis. Ann Rheum Dis 2025:S0003-4967(25)00890-8. [PMID: 40318978 DOI: 10.1016/j.ard.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVES Noncoding variants are critical to our understanding of the genetic basis of diseases and disorders such as rheumatoid arthritis (RA). While genome-wide association studies have identified regions of the genome associated with disease, functional studies are still lagging that can identify potentially causative variants. METHODS In order to functionally fine-map RA-associated variants, we identified variants at enhancers marked in primary activated T helper cells and conducted massively parallel reporter assay in these cells. RESULTS We found that combinations of functional variant genotypes are often exclusive to patients with RA. We leveraged 3-dimensional genome architecture and expression quantitative trait loci data to identify target genes of enhancers exhibiting allelic differences in activity. We confirmed enhancer activity and target gene interactions by Clustered Regularly Interpaced Short Palindromic Repeats Cas9 (CRISPR-Cas9) deletion in primary T cells. CONCLUSIONS The identification of functional enhancer variants suggests possible causal variants, and their target genes reveal known and novel genes as likely drivers of RA, as well as a means for therapeutic intervention.
Collapse
Affiliation(s)
- Ajay Jajodia
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Arpit Mishra
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Naresh Doni Jayavelu
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Nicholas Moss
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Zongchen Yang
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Jane H Buckner
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - R David Hawkins
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA; Benaroya Research Institute at Virginia Mason, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
11
|
Alizada A, Martins A, Mouniée N, Rodriguez Suarez JV, Bertin B, Gueguen N, Mirouse V, Papameletiou AM, Rivera AJ, Lau NC, Akkouche A, Maupetit-Méhouas S, Hannon GJ, Czech Nicholson B, Brasset E. The transcription factor Traffic jam orchestrates the somatic piRNA pathway in Drosophila ovaries. Cell Rep 2025; 44:115453. [PMID: 40209715 DOI: 10.1016/j.celrep.2025.115453] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/26/2025] [Accepted: 02/28/2025] [Indexed: 04/12/2025] Open
Abstract
The PIWI-interacting RNA (piRNA) pathway is essential for transposable element (TE) silencing in animal gonads. While the transcriptional regulation of piRNA pathway components in germ cells has been documented in mice and flies, their control in somatic cells of Drosophila ovaries remains unresolved. Here, we demonstrate that Traffic jam (Tj), the Drosophila ortholog of large Maf transcription factors in mammals, is a master regulator of the somatic piRNA pathway. Tj binds to regulatory regions of somatic piRNA factors and the major piRNA cluster flamenco, which carries a Tj-bound enhancer downstream of its promoter. Depletion of Tj in somatic follicle cells causes downregulation of piRNA factors, loss of flamenco expression, and derepression of gypsy-family TEs. We propose that the arms race between the host and TEs led to the co-evolution of promoters in piRNA pathway genes as well as TE regulatory regions, which both rely on a shared transcription factor.
Collapse
Affiliation(s)
- Azad Alizada
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Aline Martins
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Faculté de Médecine, 63000 Clermont-Ferrand, France
| | - Nolwenn Mouniée
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Faculté de Médecine, 63000 Clermont-Ferrand, France
| | - Julia V Rodriguez Suarez
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Benjamin Bertin
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Faculté de Médecine, 63000 Clermont-Ferrand, France
| | - Nathalie Gueguen
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Faculté de Médecine, 63000 Clermont-Ferrand, France
| | - Vincent Mirouse
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Faculté de Médecine, 63000 Clermont-Ferrand, France
| | - Anna-Maria Papameletiou
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Austin J Rivera
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Nelson C Lau
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Abdou Akkouche
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Faculté de Médecine, 63000 Clermont-Ferrand, France
| | | | - Gregory J Hannon
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK.
| | - Benjamin Czech Nicholson
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK.
| | - Emilie Brasset
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Faculté de Médecine, 63000 Clermont-Ferrand, France.
| |
Collapse
|
12
|
Becker J, Domenger C, Choksi P, Krämer C, Baumgartl C, Maiakovska O, Kim JJ, Weinmann J, Huber G, Schmidt F, Thirion C, Müller OJ, Willenbring H, Grimm D. Identification of a robust promoter in mouse and human hepatocytes by in vivo biopanning of a barcoded AAV library. Mol Ther 2025:S1525-0016(25)00301-6. [PMID: 40263935 DOI: 10.1016/j.ymthe.2025.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/21/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
Recombinant adeno-associated viruses (AAVs) are leading vectors for in vivo human gene therapy. An integral vector element is promoters, which control transgene expression in either a ubiquitous or cell-type-selective manner. Identifying optimal capsid-promoter combinations is challenging, especially when considering on- versus off-target expression. Here, we report a pipeline for in vivo promoter biopanning in AAV building on our AAV capsid barcoding technology and illustrate its potential by screening 53 promoters in 16 murine tissues using an AAV9 vector. Surprisingly, the 2.2-kb human glial fibrillary acidic protein (GFAP) promoter was the top hit in the liver, where it outperformed robust benchmarks such as the human α-1-antitrypsin promoter or the clinically used liver-specific promoter 1 (LP1). Analysis of hepatic cell populations revealed preferred GFAP promoter activity in hepatocytes. Notably, the GFAP promoter also surpassed the LP1 and cytomegalovirus promoters in human hepatocytes engrafted in an immune-deficient mouse. These findings establish the GFAP promoter as an exciting alternative for research and clinical applications requiring efficient and specific transgene expression in hepatocytes. Our pipeline expands the arsenal of technologies for high-throughput in vivo screening of viral vector components and is compatible with capsid barcoding, facilitating the combinatorial interrogation of complex AAV libraries.
Collapse
Affiliation(s)
- Jonas Becker
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Claire Domenger
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Pervinder Choksi
- Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Chiara Krämer
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Conradin Baumgartl
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Olena Maiakovska
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Jae-Jun Kim
- Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Jonas Weinmann
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Georg Huber
- Revvity Gene Delivery GmbH, 82166 Gräfelfing, Germany
| | - Florian Schmidt
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | | | - Oliver J Müller
- Department of Internal Medicine V, University Hospital Schleswig-Holstein and University of Kiel, 24105 Kiel, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Holger Willenbring
- Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Heidelberg, 69120 Heidelberg, Germany; Faculty of Engineering Sciences, University of Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
13
|
Seufert I, Vargas C, Wille SJ, Rippe K. Deregulated enhancer-promoter communication in cancer through altered nuclear architecture. Int J Cancer 2025. [PMID: 40219822 DOI: 10.1002/ijc.35424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025]
Abstract
Enhancers are critical regulators of gene expression. Structural variations in cancer genomes can lead to enhancer hijacking, where oncogenes are activated by mistargeted enhancer activity. Novel enhancer-promoter interactions may also arise through chromosomal rearrangements that create extrachromosomal DNA elements. Additionally, fusion proteins and other mutation-induced alterations in protein properties can lead to the aberrant assembly of proteins into large complexes on the size scale of 0.1-1 μm termed onco-condensates. Transcription factors and co-activators accumulate with cis-regulatory elements in these structures, driving oncogenic programs. Here, we review current evidence of how altered genome architecture and macromolecular assembly result in deregulated enhancer-promoter communication. We discuss emerging strategies to exploit these mechanisms for clinical applications.
Collapse
Affiliation(s)
- Isabelle Seufert
- German Cancer Research Center (DKFZ) Heidelberg, Division of Chromatin Networks, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), Heidelberg University, Heidelberg, Germany
| | - Claire Vargas
- German Cancer Research Center (DKFZ) Heidelberg, Division of Chromatin Networks, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), Heidelberg University, Heidelberg, Germany
| | - Sina Jasmin Wille
- German Cancer Research Center (DKFZ) Heidelberg, Division of Chromatin Networks, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), Heidelberg University, Heidelberg, Germany
| | - Karsten Rippe
- German Cancer Research Center (DKFZ) Heidelberg, Division of Chromatin Networks, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
14
|
Cuomo M, Costabile D, Della Monica R, Buonaiuto M, Trio F, De Riso G, Visconti R, Chiariotti L. A specific pluripotency-associated eRNA controls Nanog locus by shaping the epigenetic landscape and stabilizing enhancer-promoter interaction. Nucleic Acids Res 2025; 53:gkaf274. [PMID: 40219964 PMCID: PMC11992674 DOI: 10.1093/nar/gkaf274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Despite a plethora of studies exploring the transcriptional regulation of the Nanog gene, the role of the enhancer RNAs (eRNAs) derived from Nanog-interacting super-enhancers (SEs) remains under-investigated. In the present study, we examined the functional role of the eRNAs transcribed from the -5 kb Nanog SE in mouse embryonic stem cells (mESCs) and found that an eRNA, here defined as -5KNAR, was essential to maintain the Nanog locus in an epigenetically active configuration, thereby ensuring pluripotency. We found that the here identified -5KNAR functionally interacts with the RAD21 protein, suggesting a role in stabilizing a cohesin complex at the Nanog locus, ensuring the generation and maintenance of an enhancer-promoter loop. Silencing of -5KNAR caused a cascade of events, including the generation of a DNA methylation wave (likely spreading from a single methylated CpG site), substantial chromatin remodeling, and loss of the enhancer-promoter loop, inducing Nanog silencing and mESC differentiation. Under these conditions, exogenous re-expression of Nanog was unable to restore either the endogenous Nanog expression or the enhancer-promoter interaction, suggesting that, at hierarchical level, the expression of the -5KNAR plays a prominent role in maintaining the pluripotency in mESCs.
Collapse
Affiliation(s)
- Mariella Cuomo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Via S. Pansini 5, Naples 80131, Italy
- CEINGE Advanced Biotechnologies “Franco Salvatore”, Naples 80145, Italy
| | - Davide Costabile
- CEINGE Advanced Biotechnologies “Franco Salvatore”, Naples 80145, Italy
- SEMM-European School of Molecular Medicine, Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples 80131, Italy
| | - Rosa Della Monica
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Via S. Pansini 5, Naples 80131, Italy
- CEINGE Advanced Biotechnologies “Franco Salvatore”, Naples 80145, Italy
| | - Michela Buonaiuto
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Via S. Pansini 5, Naples 80131, Italy
- CEINGE Advanced Biotechnologies “Franco Salvatore”, Naples 80145, Italy
| | - Federica Trio
- CEINGE Advanced Biotechnologies “Franco Salvatore”, Naples 80145, Italy
| | - Giulia De Riso
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Via S. Pansini 5, Naples 80131, Italy
| | - Roberta Visconti
- CEINGE Advanced Biotechnologies “Franco Salvatore”, Naples 80145, Italy
- Institute for the Experimental Endocrinology and Oncology “G. Salvatore”, Italian National Council of Research, Via S. Pansini 5, 80131 Naples, Italy
| | - Lorenzo Chiariotti
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Via S. Pansini 5, Naples 80131, Italy
- CEINGE Advanced Biotechnologies “Franco Salvatore”, Naples 80145, Italy
| |
Collapse
|
15
|
Brown AR, Fox GA, Kaplow IM, Lawler AJ, Phan BN, Gadey L, Wirthlin ME, Ramamurthy E, May GE, Chen Z, Su Q, McManus CJ, van de Weerd R, Pfenning AR. An in vivo systemic massively parallel platform for deciphering animal tissue-specific regulatory function. Front Genet 2025; 16:1533900. [PMID: 40270544 PMCID: PMC12016043 DOI: 10.3389/fgene.2025.1533900] [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: 11/25/2024] [Accepted: 03/13/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction: Transcriptional regulation is an important process wherein non-protein coding enhancer sequences play a key role in determining cell type identity and phenotypic diversity. In neural tissue, these gene regulatory processes are crucial for coordinating a plethora of interconnected and regionally specialized cell types, ensuring their synchronized activity in generating behavior. Recognizing the intricate interplay of gene regulatory processes in the brain is imperative, as mounting evidence links neurodevelopment and neurological disorders to non-coding genome regions. While genome-wide association studies are swiftly identifying non-coding human disease-associated loci, decoding regulatory mechanisms is challenging due to causal variant ambiguity and their specific tissue impacts. Methods: Massively parallel reporter assays (MPRAs) are widely used in cell culture to study the non-coding enhancer regions, linking genome sequence differences to tissue-specific regulatory function. However, widespread use in animals encounters significant challenges, including insufficient viral library delivery and library quantification, irregular viral transduction rates, and injection site inflammation disrupting gene expression. Here, we introduce a systemic MPRA (sysMPRA) to address these challenges through systemic intravenous AAV viral delivery. Results: We demonstrate successful transduction of the MPRA library into diverse mouse tissues, efficiently identifying tissue specificity in candidate enhancers and aligning well with predictions from machine learning models. We highlight that sysMPRA effectively uncovers regulatory effects stemming from the disruption of MEF2C transcription factor binding sites, single-nucleotide polymorphisms, and the consequences of genetic variations associated with late-onset Alzheimer's disease. Conclusion: SysMPRA is an effective library delivering method that simultaneously determines the transcriptional functions of hundreds of enhancers in vivo across multiple tissues.
Collapse
Affiliation(s)
- Ashley R. Brown
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Grant A. Fox
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Irene M. Kaplow
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Alyssa J. Lawler
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - BaDoi N. Phan
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Lahari Gadey
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Morgan E. Wirthlin
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Easwaran Ramamurthy
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Gemma E. May
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Ziheng Chen
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Qiao Su
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Robert van de Weerd
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Andreas R. Pfenning
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
16
|
Dababneh SF, Babini H, Jiménez-Sábado V, Teves SS, Kim KH, Tibbits GF. Dissecting cardiovascular disease-associated noncoding genetic variants using human iPSC models. Stem Cell Reports 2025; 20:102467. [PMID: 40118058 PMCID: PMC12069897 DOI: 10.1016/j.stemcr.2025.102467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/23/2025] Open
Abstract
Advancements in genomics have revealed hundreds of loci associated with cardiovascular diseases, highlighting the role genetic variants play in disease pathogenesis. Notably, most variants lie within noncoding genomic regions that modulate transcription factor binding, chromatin accessibility, and thereby the expression levels and cell type specificity of gene transcripts. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as a powerful tool to delineate the pathogenicity of such variants and elucidate the underlying transcriptional mechanisms. Our review discusses the basics of noncoding variant-mediated pathogenesis, the methodologies utilized, and how hiPSC-based heart models can be leveraged to dissect the mechanisms of noncoding variants.
Collapse
Affiliation(s)
- Saif F Dababneh
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Hosna Babini
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Departments of Molecular Biology and Biochemistry / Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Verónica Jiménez-Sábado
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Departments of Molecular Biology and Biochemistry / Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Sheila S Teves
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Kyoung-Han Kim
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Glen F Tibbits
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Departments of Molecular Biology and Biochemistry / Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC V5A 1S6, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada.
| |
Collapse
|
17
|
Nayak N, Mehrotra S, Karamchandani AN, Santelia D, Mehrotra R. Recent advances in designing synthetic plant regulatory modules. FRONTIERS IN PLANT SCIENCE 2025; 16:1567659. [PMID: 40241826 PMCID: PMC11999978 DOI: 10.3389/fpls.2025.1567659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025]
Abstract
Introducing novel functions in plants through synthetic multigene circuits requires strict transcriptional regulation. Currently, the use of natural regulatory modules in synthetic circuits is hindered by our limited knowledge of complex plant regulatory mechanisms, the paucity of characterized promoters, and the possibility of crosstalk with endogenous circuits. Synthetic regulatory modules can overcome these limitations. This article introduces an integrative de novo approach for designing plant synthetic promoters by utilizing the available online tools and databases. The recent achievements in designing and validating synthetic plant promoters, enhancers, transcription factors, and the challenges of establishing synthetic circuits in plants are also discussed.
Collapse
Affiliation(s)
- Namitha Nayak
- Department of Biological Sciences, Birla Institute of Technology and Sciences Pilani, Goa, India
| | - Sandhya Mehrotra
- Department of Biological Sciences, Birla Institute of Technology and Sciences Pilani, Goa, India
| | | | - Diana Santelia
- Institute of Integrative Biology, ETH Zürich Universitätstrasse, Zürich, Switzerland
| | - Rajesh Mehrotra
- Department of Biological Sciences, Birla Institute of Technology and Sciences Pilani, Goa, India
| |
Collapse
|
18
|
Chen W, Wang Z, Wang Y, Lin J, Chen S, Chen H, Ma X, Zou X, Li X, Qin Y, Xiong K, Ma X, Liao Q, Qiao Y, Li L. Enhancer RNA Transcriptome-Wide Association Study Reveals a Distinctive Class of Pan-Cancer Susceptibility eRNAs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411974. [PMID: 39950845 PMCID: PMC11967800 DOI: 10.1002/advs.202411974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/10/2025] [Indexed: 04/05/2025]
Abstract
Many cancer risk variants are located within enhancer regions and lack sufficient molecular interpretation. Here, we constructed the first comprehensive atlas of enhancer RNA (eRNA)-mediated genetic effects from 28 033 RNA sequencing samples across 11 606 individuals, identifying 21 073 eRNA quantitative trait loci (eRNA-QTLs) significantly associated with eRNA expression. Mechanistically, eRNA-QTLs frequently altered binding motifs of transcription factors. In addition, 28.48% of cancer risk variants are strongly colocalized with eRNA-QTLs. A pan-cancer eRNA-based transcriptome-wide association study is conducted across 23 major cancer types, identifying 626 significant cancer susceptibility eRNAs predicted to modulate cancer risk via eRNA, from which 54.90% of the eRNA target genes are overlooked by traditional gene expression studies, and most are essential for cancer cell proliferation. As proof of principle validation, the enhancer functionality of two newly identified susceptibility eRNAs, CCND1e and SNAPC1e, is confirmed through CRISPR inhibition and shRNA-mediated knockdown, resulting in a marked decrease in the expression of their respective target genes, consequently suppressing the proliferation of prostate cancer cells. The study underscores the essential role of eRNA in unveiling new cancer susceptibility genes and establishes a strong framework for enhancing our understanding of human cancer etiology.
Collapse
Affiliation(s)
- Wenyan Chen
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Zeyang Wang
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Yinuo Wang
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Jianxiang Lin
- Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
- Shanghai Institute of Precision MedicineShanghai200125China
| | - Shuxin Chen
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Hui Chen
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Xuelian Ma
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Xudong Zou
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Xing Li
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Yangmei Qin
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Kewei Xiong
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Xixian Ma
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| | - Qi Liao
- School of Public HealthHealth Science CenterNingbo UniversityNingbo315211China
| | - Yunbo Qiao
- Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
- Shanghai Institute of Precision MedicineShanghai200125China
| | - Lei Li
- Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhen518055China
| |
Collapse
|
19
|
Abbasova L, Urbanaviciute P, Hu D, Ismail JN, Schilder BM, Nott A, Skene NG, Marzi SJ. CUT&Tag recovers up to half of ENCODE ChIP-seq histone acetylation peaks. Nat Commun 2025; 16:2993. [PMID: 40148272 PMCID: PMC11950320 DOI: 10.1038/s41467-025-58137-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
DNA-protein interactions have traditionally been profiled via chromatin immunoprecipitation followed by next-generation sequencing (ChIP-seq). Cleavage Under Targets & Tagmentation (CUT&Tag) is a rapidly expanding technique that enables the profiling of such interactions in situ at high sensitivity. However, thorough evaluation and benchmarking against established ChIP-seq datasets are lacking. Here, we comprehensively benchmarked CUT&Tag for H3K27ac and H3K27me3 against published ChIP-seq profiles from ENCODE in K562 cells. Combining multiple new and published CUT&Tag datasets, there was an average recall of 54% known ENCODE peaks for both histone modifications. We tested peak callers MACS2 and SEACR and identified optimal peak calling parameters. Overall, peaks identified by CUT&Tag represent the strongest ENCODE peaks and show the same functional and biological enrichments as ChIP-seq peaks identified by ENCODE. Our workflow systematically evaluates the merits of methodological adjustments, providing a benchmarking framework for the experimental design and analysis of CUT&Tag studies.
Collapse
Affiliation(s)
- Leyla Abbasova
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Paulina Urbanaviciute
- UK Dementia Research Institute at King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Di Hu
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Joy N Ismail
- UK Dementia Research Institute at King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Brian M Schilder
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Alexi Nott
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Nathan G Skene
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Sarah J Marzi
- Department of Brain Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute at King's College London, London, UK.
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
20
|
Wang SK, Li J, Nair S, Korasaju R, Chen Y, Zhang Y, Kundaje A, Liu Y, Wang N, Chang HY. Single-cell multiome and enhancer connectome of human retinal pigment epithelium and choroid nominate pathogenic variants in age-related macular degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644670. [PMID: 40196652 PMCID: PMC11974679 DOI: 10.1101/2025.03.21.644670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss worldwide. Genome-wide association studies (GWAS) of AMD have identified dozens of risk loci that may house disease targets. However, variants at these loci are largely noncoding, making it difficult to assess their function and whether they are causal. Here, we present a single-cell gene expression and chromatin accessibility atlas of human retinal pigment epithelium (RPE) and choroid to systematically analyze both coding and noncoding variants implicated in AMD. We employ HiChIP and Activity-by-Contact modeling to map enhancers in these tissues and predict cell and gene targets of risk variants. We further perform allele-specific self-transcribing active regulatory region sequencing (STARR-seq) to functionally test variant activity in RPE cells, including in the context of complement activation. Our work nominates new pathogenic variants and mechanisms in AMD and offers a rich and accessible resource for studying diseases of the RPE and choroid.
Collapse
Affiliation(s)
- Sean K Wang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jiaying Li
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Surag Nair
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Reshma Korasaju
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Yun Chen
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yuanyuan Zhang
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuwen Liu
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kunpeng Institute of Modern Agriculture at Foshan, Chinese Academy of Agricultural Sciences, Foshan, China
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Henan Academy of Innovations in Medical Science, Henan, China
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Current address: Amgen Research, South San Francisco, CA, USA
- Lead contact
| |
Collapse
|
21
|
Li X, Schmitz RJ. Cis-regulatory dynamics in plant domestication. Trends Genet 2025:S0168-9525(25)00046-0. [PMID: 40140332 DOI: 10.1016/j.tig.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/12/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025]
Abstract
Cis-regulatory elements (CREs) are critical sequence determinants for spatiotemporal control of gene expression. Genetic variants within CREs have driven phenotypic transitions from wild to cultivated plants during domestication. This review summarizes our current understanding of genetic variants within CREs involved in plant domestication. We also propose avenues for studies to expand our understanding of both CRE biology and domestication processes, such as examining primary mechanisms that generate CRE genetic variants during plant domestication and investigating the roles of CREs in domestication syndrome. Additionally, we discuss existing challenges and highlight future opportunities for exploring CREs in plant domestication, emphasizing the potential of modifying CREs to contribute to crop improvement.
Collapse
Affiliation(s)
- Xiang Li
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Robert J Schmitz
- Department of Genetics, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
22
|
Sathian R, Dutta P, Ay F, Davuluri RV. Genomic Language Model for Predicting Enhancers and Their Allele-Specific Activity in the Human Genome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.644040. [PMID: 40166250 PMCID: PMC11957021 DOI: 10.1101/2025.03.18.644040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Predicting and deciphering the regulatory logic of enhancers is a challenging problem, due to the intricate sequence features and lack of consistent genetic or epigenetic signatures that can accurately discriminate enhancers from other genomic regions. Recent machine-learning based methods have spotlighted the importance of extracting nucleotide composition of enhancers but failed to learn the sequence context and perform suboptimally. Motivated by advances in genomic language models, we developed DNABERT-Enhancer, a novel enhancer prediction method, by applying DNABERT pre-trained language model on the human genome. We trained two different models, using large collection of enhancers curated from the ENCODE registry of candidate cis-Regulatory Elements. The best fine-tuned model achieved 88.05% accuracy with Matthews correlation coefficient of 76% on independent set aside data. Further, we present the analysis of the predicted enhancers for all chromosomes of the human genome by comparing with the enhancer regions reported in publicly available databases. Finally, we applied DNABERT-Enhancer along with other DNABERT based regulatory genomic region prediction models to predict candidate SNPs with allele-specific enhancer and transcription factor binding activity. The genome-wide enhancer annotations and candidate loss-of-function genetic variants predicted by DNABERT-Enhancer provide valuable resources for genome interpretation in functional and clinical genomics studies.
Collapse
|
23
|
Monsen Ø, Grønvold L, Datsomor A, Harvey T, Kijas J, Suh A, Hvidsten TR, Sandve SR. The role of transposon activity in shaping cis-regulatory element evolution after whole-genome duplication. Genome Res 2025; 35:475-488. [PMID: 39939177 PMCID: PMC11960703 DOI: 10.1101/gr.278931.124] [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: 01/02/2024] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Whole-genome duplications (WGDs) and transposable element (TE) activity can act synergistically in genome evolution. WGDs can increase TE activity directly through cellular stress or indirectly by relaxing selection against TE insertions in functionally redundant, duplicated regions. Because TEs can function as, or evolve into, TE-derived cis-regulatory elements (TE-CREs), bursts of TE activity following WGD are therefore likely to impact evolution of gene regulation. Yet, the role of TEs in genome regulatory evolution after WGDs is not well understood. Here we used Atlantic salmon as a model system to explore how TE activity after the salmonid WGD ∼100 MYA shaped CRE evolution. We identified 55,080 putative TE-CREs using chromatin accessibility data from the liver and brain. Retroelements were both the dominant source of TE-CREs and had higher regulatory activity in MPRA experiments compared with DNA elements. A minority of TE subfamilies (16%) accounted for 46% of TE-CREs, but these "CRE superspreaders" were mostly active prior to the WGD. Analysis of individual TE insertions, however, revealed enrichment of TE-CREs originating from WGD-associated TE activity, particularly for the DTT (Tc1-Mariner) DNA elements. Furthermore, coexpression analyses supported the presence of TE-driven gene regulatory network evolution, including DTT elements active at the time of WGD. In conclusion, our study supports a scenario in which TE activity has been important in genome regulatory evolution, either through relaxed selective constraints or through strong selection to recalibrate optimal gene expression phenotypes, during a transient period following genome doubling.
Collapse
Affiliation(s)
- Øystein Monsen
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, 1432 Ås, Norway
| | - Lars Grønvold
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, 1432 Ås, Norway
| | - Alex Datsomor
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, 1432 Ås, Norway
| | - Thomas Harvey
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, 1432 Ås, Norway
| | - James Kijas
- Aquaculture Programme, Commonwealth Scientific and Industrial Research Organisation, St. Lucia, Queensland 4067, Australia
| | - Alexander Suh
- School of Biological Sciences-Organisms and the Environment, University of East Anglia, NR4 7TU Norwich, United Kingdom
- Department of Organismal Biology-Systematic Biology (EBC), Uppsala University, SE-752 36 Uppsala, Sweden
| | - Torgeir R Hvidsten
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, 1432 Ås, Norway
| | - Simen Rød Sandve
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, 1432 Ås, Norway;
| |
Collapse
|
24
|
Paramo MI, Leung AKY, Shah SR, Zhang J, Tippens ND, Liang J, Yao L, Jin Y, Pan X, Ozer A, Lis JT, Yu H. Simultaneous measurement of intrinsic promoter and enhancer potential reveals principles of functional duality and regulatory reciprocity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643265. [PMID: 40161809 PMCID: PMC11952525 DOI: 10.1101/2025.03.14.643265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Growing evidence indicates that transcriptional regulatory elements can exert both promoter and enhancer activity; however, the relationship and determinants of this dual functionality remain poorly understood. We developed a massively parallel dual reporter assay that enables simultaneous assessment of the intrinsic promoter and enhancer potential exerted by the same sequence. Parallel quantification for thousands of elements reveals that canonical human promoters and enhancers can act as both promoters and enhancers under the same contexts, and that promoter activity may be necessary but not sufficient for enhancer function. We find that regulatory potential is intrinsic to element sequences, irrespective of downstream features typically associated with distinct element classes. Perturbations to element transcription factor binding motifs lead to disruptions in both activities, implicating a shared syntax for the two regulatory functions. Combinations of elements with different minimal promoters reveal reciprocal activity modulation between associated elements and a strong positive correlation between promoter and enhancer functions imply a bidirectional feedback loop used to maintain environments of high transcriptional activity. Finally, our results indicate that the magnitude and balance between promoter and enhancer functions are shaped by both intrinsic sequence properties and contextual regulatory influences, suggesting a degree of plasticity in regulatory action. Our approach provides a new lens for understanding fundamental principles of regulatory element biology.
Collapse
|
25
|
Qiu Y, Liu L, Yan J, Xiang X, Wang S, Luo Y, Deng K, Xu J, Jin M, Wu X, Liwei Cheng, Zhou Y, Xie W, Liu HJ, Fernie AR, Hu X, Yan J. Precise engineering of gene expression by editing plasticity. Genome Biol 2025; 26:51. [PMID: 40065399 PMCID: PMC11892124 DOI: 10.1186/s13059-025-03516-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Identifying transcriptional cis-regulatory elements (CREs) and understanding their role in gene expression are essential for the precise manipulation of gene expression and associated phenotypes. This knowledge is fundamental for advancing genetic engineering and improving crop traits. RESULTS We here demonstrate that CREs can be accurately predicted and utilized to precisely regulate gene expression beyond the range of natural variation. We firstly build two sequence-to-expression deep learning models to respectively identify distal and proximal CREs by combining them with interpretability methods in multiple crops. A large number of distal CREs are verified for enhancer activity in vitro using UMI-STARR-seq on 12,000 synthesized sequences. These comprehensively characterized CREs and their precisely predicted effects further contribute to the design of in silico editing schemes for precise engineering of gene expression. We introduce a novel concept of "editingplasticity" to evaluate the potential of promoter editing to alter expression of each gene. As a proof of concept, both exhaustive prediction and random knockout mutants are analyzed within the promoter region of ZmVTE4, a key gene affecting α-tocopherol content in maize. A high degree of agreement between predicted and observed expression is observed, extending the range of natural variation and thereby allowing the creation of an optimal phenotype. CONCLUSIONS Our study provides a robust computational framework that advances knowledge-guided gene editing for precise regulation of gene expression and crop improvement. By reliably predicting and validating CREs, we offer a tool for targeted genetic modifications, enhancing desirable traits in crops.
Collapse
Affiliation(s)
- Yang Qiu
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Lifen Liu
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jiali Yan
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Xianglei Xiang
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shouzhe Wang
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Yun Luo
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Kaixuan Deng
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jieting Xu
- WIMI Biotechnology Co., Ltd., Sanya, Hainan, 572000, China
| | - Minliang Jin
- WIMI Biotechnology Co., Ltd., Sanya, Hainan, 572000, China
| | - Xiaoyu Wu
- WIMI Biotechnology Co., Ltd., Sanya, Hainan, 572000, China
| | - Liwei Cheng
- WIMI Biotechnology Co., Ltd., Sanya, Hainan, 572000, China
| | - Ying Zhou
- Institute of Agricultural Sciences of Xishuangbanna Prefecture of Yunnan Province, Jinghong, Yunnan, 666100, China
- The Expert Workstation of Jianbing Yan in Yunnan Province, Jinghong, Yunnan, 666100, China
| | - Weibo Xie
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Hai-Jun Liu
- Yazhouwan National Laboratory, Sanya, 572024, China
| | - Alisdair R Fernie
- Department of Molecular Physiology, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, 14476, Germany
| | - Xuehai Hu
- Hubei Hongshan Laboratory, Wuhan, 430070, China.
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jianbing Yan
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, 430070, China.
- Hubei Hongshan Laboratory, Wuhan, 430070, China.
- Yazhouwan National Laboratory, Sanya, 572024, China.
| |
Collapse
|
26
|
Huang Y, Verstegen MJAM, Tjalsma SJD, Krijger PHL, Gupta K, Park M, Boettiger A, de Laat W. Two unrelated distal genes activated by a shared enhancer benefit from localizing inside the same small topological domain. Genes Dev 2025; 39:348-363. [PMID: 39870429 PMCID: PMC11874980 DOI: 10.1101/gad.352235.124] [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: 08/19/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025]
Abstract
Enhancers are tissue-specific regulatory DNA elements that can activate transcription of genes over distance. Their target genes most often are located in the same contact domain-chromosomal entities formed by cohesin DNA loop extrusion and typically flanked by CTCF-bound boundaries. Enhancers shared by multiple unrelated genes are underexplored but may be more common than anticipated. Here, we analyzed the interplay between an enhancer and two distal functionally unrelated genes residing at opposite domain boundaries. The enhancer strongly activated their expression and supported their frequent interactions. Cohesin structured the domain and supported their transcription, but the genes did not rely on each other's transcription or show gene competition. Deleting either domain boundary not only extended the contact domain but led to reduced contacts within the original domain and reduction in the expression of both genes. Conversely, by isolating either gene with the enhancer in shorter domains, through insertion of new CTCF boundaries, intradomain contact frequencies increased, and the gene isolated with the enhancer was upregulated. Collectively, this shows that an enhancer can independently activate unrelated distal genes and that long-range gene regulation benefits from operating in small contact domains.
Collapse
Affiliation(s)
- Yike Huang
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Marjon J A M Verstegen
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Sjoerd J D Tjalsma
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Peter H L Krijger
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Kavvya Gupta
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Minhee Park
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Alistair Boettiger
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Wouter de Laat
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands;
| |
Collapse
|
27
|
Yang J, Zhou F, Luo X, Fang Y, Wang X, Liu X, Xiao R, Jiang D, Tang Y, Yang G, You L, Zhao Y. Enhancer reprogramming: critical roles in cancer and promising therapeutic strategies. Cell Death Discov 2025; 11:84. [PMID: 40032852 DOI: 10.1038/s41420-025-02366-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 01/24/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
Transcriptional dysregulation is a hallmark of cancer initiation and progression, driven by genetic and epigenetic alterations. Enhancer reprogramming has emerged as a pivotal driver of carcinogenesis, with cancer cells often relying on aberrant transcriptional programs. The advent of high-throughput sequencing technologies has provided critical insights into enhancer reprogramming events and their role in malignancy. While targeting enhancers presents a promising therapeutic strategy, significant challenges remain. These include the off-target effects of enhancer-targeting technologies, the complexity and redundancy of enhancer networks, and the dynamic nature of enhancer reprogramming, which may contribute to therapeutic resistance. This review comprehensively encapsulates the structural attributes of enhancers, delineates the mechanisms underlying their dysregulation in malignant transformation, and evaluates the therapeutic opportunities and limitations associated with targeting enhancers in cancer.
Collapse
Affiliation(s)
- Jinshou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Feihan Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xiyuan Luo
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Yuan Fang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Decheng Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Yuemeng Tang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
28
|
Oberlin S, McManus MT. Decoding gene regulation with CRISPR perturbations. Nat Biotechnol 2025; 43:304-305. [PMID: 38760568 DOI: 10.1038/s41587-024-02222-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Affiliation(s)
- Stefan Oberlin
- Department of Microbiology and Immunology & UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Michael T McManus
- Department of Microbiology and Immunology & UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
29
|
Stock M, Losert C, Zambon M, Popp N, Lubatti G, Hörmanseder E, Heinig M, Scialdone A. Leveraging prior knowledge to infer gene regulatory networks from single-cell RNA-sequencing data. Mol Syst Biol 2025; 21:214-230. [PMID: 39939367 PMCID: PMC11876610 DOI: 10.1038/s44320-025-00088-3] [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: 10/10/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
Many studies have used single-cell RNA sequencing (scRNA-seq) to infer gene regulatory networks (GRNs), which are crucial for understanding complex cellular regulation. However, the inherent noise and sparsity of scRNA-seq data present significant challenges to accurate GRN inference. This review explores one promising approach that has been proposed to address these challenges: integrating prior knowledge into the inference process to enhance the reliability of the inferred networks. We categorize common types of prior knowledge, such as experimental data and curated databases, and discuss methods for representing priors, particularly through graph structures. In addition, we classify recent GRN inference algorithms based on their ability to incorporate these priors and assess their performance in different contexts. Finally, we propose a standardized benchmarking framework to evaluate algorithms more fairly, ensuring biologically meaningful comparisons. This review provides guidance for researchers selecting GRN inference methods and offers insights for developers looking to improve current approaches and foster innovation in the field.
Collapse
Affiliation(s)
- Marco Stock
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Corinna Losert
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Matteo Zambon
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany
| | - Niclas Popp
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany
| | - Gabriele Lubatti
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany
| | - Eva Hörmanseder
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
| | - Matthias Heinig
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- German Centre for Cardiovascular Research (DZHK), Munich Heart Association, Partner Site Munich, Berlin, Germany
| | - Antonio Scialdone
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany.
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany.
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany.
| |
Collapse
|
30
|
Krchlikova V, Lu Y, Sauter D. Viral influencers: deciphering the role of endogenous retroviral LTR12 repeats in cellular gene expression. J Virol 2025; 99:e0135124. [PMID: 39887236 PMCID: PMC11853044 DOI: 10.1128/jvi.01351-24] [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] [Indexed: 02/01/2025] Open
Abstract
The human genome is like a museum of ancient retroviral infections. It contains a large number of endogenous retroviruses (ERVs) that bear witness to past integration events. About 5,000 of them are so-called long terminal repeat 12 (LTR12) elements. Compared with 20,000 human genes, this is a remarkable number. Although LTR12 elements can act as promoters or enhancers of cellular genes, the function of most of these retroviral elements has remained unclear. In our mini-review, we show that different LTR12 elements share many similarities, including common transcription factor binding sites. Furthermore, we summarize novel insights into the epigenetic mechanisms governing their silencing and activation. Specific examples of genes and pathways that are regulated by LTR12 loci are used to illustrate the regulatory network built by these repetitive elements. A particular focus is on their role in the regulation of antiviral immune responses, tumor cell proliferation, and senescence. Finally, we describe how a targeted activation of this fascinating ERV family could be used for diagnostic or therapeutic purposes.
Collapse
Affiliation(s)
- Veronika Krchlikova
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Yueshuang Lu
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Daniel Sauter
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
31
|
Tonelli A, Cousin P, Jankowski A, Wang B, Dorier J, Barraud J, Zunjarrao S, Gambetta MC. Systematic screening of enhancer-blocking insulators in Drosophila identifies their DNA sequence determinants. Dev Cell 2025; 60:630-645.e9. [PMID: 39532105 DOI: 10.1016/j.devcel.2024.10.017] [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: 01/26/2024] [Revised: 06/21/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Long-range transcriptional activation of gene promoters by abundant enhancers in animal genomes calls for mechanisms to limit inappropriate regulation. DNA elements called insulators serve this purpose by shielding promoters from an enhancer when interposed. Unlike promoters and enhancers, insulators have not been systematically characterized due to lacking high-throughput screening assays, and questions regarding how insulators are distributed and encoded in the genome remain. Here, we establish "insulator-seq" as a plasmid-based massively parallel reporter assay in Drosophila cultured cells to perform a systematic insulator screen of selected genomic loci. Screening developmental gene loci showed that not all insulator protein binding sites effectively block enhancer-promoter communication. Deep insulator mutagenesis identified sequences flexibly positioned around the CTCF insulator protein binding motif that are critical for functionality. The ability to screen millions of DNA sequences without positional effect has enabled functional mapping of insulators and provided further insights into the determinants of insulators.
Collapse
Affiliation(s)
- Anastasiia Tonelli
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Pascal Cousin
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Aleksander Jankowski
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, 02-097 Warsaw, Poland
| | - Bihan Wang
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Julien Dorier
- Bioinformatics Competence Center, University of Lausanne, 1015 Lausanne, Switzerland; Bioinformatics Competence Center, Swiss Federal Institute of Technology Lausanne, 1015 Lausanne, Switzerland
| | - Jonas Barraud
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Sanyami Zunjarrao
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | | |
Collapse
|
32
|
Fu ZH, Cheng S, Li JW, Zhang N, Wu Y, Zhao GR. Synthetic tunable promoters for flexible control of multi-gene expression in mammalian cells. J Adv Res 2025:S2090-1232(25)00106-7. [PMID: 39938795 DOI: 10.1016/j.jare.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025] Open
Abstract
INTRODUCTION Synthetic biology revolutionizes our ability to decode and recode genetic systems. The capability to reconstruct and flexibly manipulate multi-gene systems is critical for understanding cellular behaviors and has significant applications in therapeutics. OBJECTIVES This study aims to construct a diverse library of synthetic tunable promoters (STPs) to enable flexible control of multi-gene expression in mammalian cells. METHODS We designed and constructed synthetic tunable promoters (STPs) that incorporate both a universal activation site (UAS) and a specific activation site (SAS), enabling multi-level expression control via the CRISPR activation (CRISPRa) system. To evaluate promoter activity, we utilized Massively Parallel Reporter Assays (MPRA) to assess the basal strengths of the STPs and their activation responses. Next, we constructed a three-gene reporter system to assess the capacity of the synthetic promoters for achieving multilevel control of single-gene expression within multi-gene systems. RESULTS The promoter library contains 24,960 unique non-redundant promoters with distinct sequence characteristics. MPRA revealed a wide range of promoter activities, showing different basal strengths and distinct activation levels when activated by the CRISPRa system. When regulated by targeting the SAS, the STPs exhibited orthogonality, allowing multilevel control of single-gene expression within multi-gene systems without cross-interference. Furthermore, the combinatorial activation of STPs in a multi-gene system enlarged the scope of expression levels achievable, providing fine-tuned control over gene expression. CONCLUSION We provide a diverse collection of synthetic tunable promoters, offering a valuable toolkit for the construction and manipulation of multi-gene systems in mammalian cells, with applications in gene therapy and biotechnology.
Collapse
Affiliation(s)
- Zong-Heng Fu
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, 300072, China
| | - Si Cheng
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, 300072, China
| | - Jia-Wei Li
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, 300072, China
| | - Nan Zhang
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, 300072, China
| | - Yi Wu
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, 300072, China.
| | - Guang-Rong Zhao
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
33
|
Wan J, van Ouwerkerk A, Mouren JC, Heredia C, Pradel L, Ballester B, Andrau JC, Spicuglia S. Comprehensive mapping of genetic variation at Epromoters reveals pleiotropic association with multiple disease traits. Nucleic Acids Res 2025; 53:gkae1270. [PMID: 39727170 PMCID: PMC11879118 DOI: 10.1093/nar/gkae1270] [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: 06/03/2024] [Revised: 10/28/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
There is growing evidence that a wide range of human diseases and physiological traits are influenced by genetic variation of cis-regulatory elements. We and others have shown that a subset of promoter elements, termed Epromoters, also function as enhancer regulators of distal genes. This opens a paradigm in the study of regulatory variants, as single nucleotide polymorphisms (SNPs) within Epromoters might influence the expression of several (distal) genes at the same time, which could disentangle the identification of disease-associated genes. Here, we built a comprehensive resource of human Epromoters using newly generated and publicly available high-throughput reporter assays. We showed that Epromoters display intrinsic and epigenetic features that distinguish them from typical promoters. By integrating Genome-Wide Association Studies (GWAS), expression Quantitative Trait Loci (eQTLs) and 3D chromatin interactions, we found that regulatory variants at Epromoters are concurrently associated with more disease and physiological traits, as compared with typical promoters. To dissect the regulatory impact of Epromoter variants, we evaluated their impact on regulatory activity by analyzing allelic-specific high-throughput reporter assays and provided reliable examples of pleiotropic Epromoters. In summary, our study represents a comprehensive resource of regulatory variants supporting the pleiotropic role of Epromoters.
Collapse
Affiliation(s)
- Jing Wan
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| | - Antoinette van Ouwerkerk
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| | | | - Carla Heredia
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, UMR 5535, Montpellier, France
| | - Lydie Pradel
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| | - Benoit Ballester
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
| | - Jean-Christophe Andrau
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, UMR 5535, Montpellier, France
| | - Salvatore Spicuglia
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| |
Collapse
|
34
|
Chesnokova E, Bal N, Alhalabi G, Balaban P. Regulatory Elements for Gene Therapy of Epilepsy. Cells 2025; 14:236. [PMID: 39937026 PMCID: PMC11816724 DOI: 10.3390/cells14030236] [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: 12/28/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
The problem of drug resistance in epilepsy means that in many cases, a surgical treatment may be advised. But this is only possible if there is an epileptic focus, and resective brain surgery may have adverse side effects. One of the promising alternatives is gene therapy, which allows the targeted expression of therapeutic genes in different brain regions, and even in specific cell types. In this review, we provide detailed explanations of some key terms related to genetic engineering, and describe various regulatory elements that have already been used in the development of different approaches to treating epilepsy using viral vectors. We compare a few universal promoters for their strength and duration of transgene expression, and in our description of cell-specific promoters, we focus on elements driving expression in glutamatergic neurons, GABAergic neurons and astrocytes. We also explore enhancers and some other cis-regulatory elements currently used in viral vectors for gene therapy, and consider future perspectives of state-of-the-art technologies for designing new, stronger and more specific regulatory elements. Gene therapy has multiple advantages and should become more common in the future, but there is still a lot to study and invent in this field.
Collapse
Affiliation(s)
- Ekaterina Chesnokova
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia; (E.C.); (P.B.)
| | - Natalia Bal
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia; (E.C.); (P.B.)
| | - Ghofran Alhalabi
- Laboratory of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia;
| | - Pavel Balaban
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia; (E.C.); (P.B.)
| |
Collapse
|
35
|
Caporale N, Leonardi O, Villa CE, Vitriolo A, Boeckx C, Testa G. Tile by tile: capturing the evolutionary mosaic of human conditions. Curr Opin Genet Dev 2025; 90:102297. [PMID: 39705881 DOI: 10.1016/j.gde.2024.102297] [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: 07/03/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024]
Abstract
The collection of Homo sapiens anatomical hallmarks hypothesized to support the 'human condition' did not appear at one specific time and place, but gradually, creating a reticulate evolutionary trajectory. The recent reconstruction of migration patterns and gene flows across different hominin species and populations draws a mosaic that we contend can be illuminated by genomic comparisons and specific experiments. Here, we first review key discoveries that could allow this experimental endeavor by describing recent advances in a variety of fields, stressing the importance of charting the current human neurodiversity as an interpretive substrate for evolutionary changes. Then, we identify key cellular and molecular observables. Finally, given the vast amount of possible variants, we focus the discussion on technologies that could allow their interrogation in a way that is compatible with the staggering amount of contemporary genomic and phenotypic characterization.
Collapse
Affiliation(s)
- Nicolò Caporale
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy. https://twitter.com/@NicoloCaporale
| | - Oliviero Leonardi
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy. https://twitter.com/@OlivieroLeonar2
| | - Carlo Emanuele Villa
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy. https://twitter.com/@CarloEmanueleV1
| | - Alessandro Vitriolo
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy. https://twitter.com/@AVitriolScience
| | - Cedric Boeckx
- University of Barcelona, 08007 Barcelona, Spain; University of Barcelona Institute of Complex Systems, 08007 Barcelona, Spain; University of Barcelona Institute of Neurosciences, 08007 Barcelona, Spain; Catalan Institute for Research and Advanced Studies (ICREA), 08007 Barcelona, Spain.
| | - Giuseppe Testa
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy.
| |
Collapse
|
36
|
Zolin IA, Georgieva SG, Nikolenko JV. Evolutionarily Conserved DHX9/MLE Helicase Is Involved in the Regulation of Its Own mRNA Expression Level in Drosophila melanogaster. DOKL BIOCHEM BIOPHYS 2025; 520:1-5. [PMID: 39899250 DOI: 10.1134/s1607672924601239] [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: 09/20/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 02/04/2025]
Abstract
The MLE helicase of D. melanogaster, like its ortholog DHX9 in mammals, is involved in a wide range of processes related to the regulation of gene expression. In the present study, we investigated the impact of the mle[9] mutation on its own mRNA expression level. It was shown that in addition to the previously described deletion in the catalytic domain of the protein, which impairs its helicase activity, the mle[9] mutation contains an additional small deletion in the C-terminal domain. In the mle[9] mutation background, there was a threefold increase in the expression of the main transcript of the mle gene encoding the full-length protein. Binding of MLE to chromatin at the coding region and promoters of the mle gene and nearby enhancers was analyzed. To exclude the influence of dosage compensation, experiments were performed on females. The data obtained indicate the role of MLE in specific regulation of its own mRNA expression level in vivo at the adult stage.
Collapse
Affiliation(s)
- I A Zolin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - S G Georgieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - J V Nikolenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
37
|
Chignon A, Lettre G. Using omics data and genome editing methods to decipher GWAS loci associated with coronary artery disease. Atherosclerosis 2025; 401:118621. [PMID: 39909615 DOI: 10.1016/j.atherosclerosis.2024.118621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 02/07/2025]
Abstract
Coronary artery disease (CAD) is due to atherosclerosis, a pathophysiological process that involves several cell-types and results in the accumulation of lipid-rich plaque that disrupt the normal blood flow through the coronary arteries to the heart. Genome-wide association studies have identified 1000s of genetic variants robustly associated with CAD or its traditional risk factors (e.g. blood pressure, blood lipids, type 2 diabetes, smoking). However, gaining biological insights from these genetic discoveries remain challenging because of linkage disequilibrium and the difficulty to interpret the functions of non-coding regulatory elements in the human genome. In this review, we present different statistical methods (e.g. Mendelian randomization) and molecular datasets (e.g. expression or protein quantitative trait loci) that have helped connect CAD-associated variants with genes, biological pathways, and cell-types or tissues. We emphasize that these various strategies make predictions, which need to be validated in orthologous systems. We discuss specific examples where the integration of omics data with GWAS results has prioritized causal CAD variants and genes. Finally, we review how targeted and genome-wide genome editing experiments using the CRISPR/Cas9 toolbox have been used to characterize new CAD genes in human cells. Researchers now have the statistical and bioinformatic methods, the molecular datasets, and the experimental tools to dissect comprehensively the loci that contribute to CAD risk in humans.
Collapse
Affiliation(s)
- Arnaud Chignon
- Montreal Heart Institute, Montreal, Quebec, Canada; Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada
| | - Guillaume Lettre
- Montreal Heart Institute, Montreal, Quebec, Canada; Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
38
|
Alizada A, Martins A, Mouniée N, Rodriguez Suarez JV, Bertin B, Gueguen N, Mirouse V, Papameletiou AM, Rivera AJ, Lau NC, Akkouche A, Maupetit-Mehouas S, Hannon GJ, Nicholson BC, Brasset E. The transcription factor Traffic jam orchestrates the somatic piRNA pathway in Drosophila ovaries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.10.612307. [PMID: 39314383 PMCID: PMC11419008 DOI: 10.1101/2024.09.10.612307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The PIWI-interacting RNA (piRNA) pathway is essential for transposable element (TE) silencing in animal gonads. While the transcriptional regulation of piRNA pathway components in germ cells has been documented in mice and flies, their control in somatic cells of Drosophila ovaries remains unresolved. Here, we demonstrate that Traffic jam (Tj), the Drosophila orthologue of large Maf transcription factors in mammals, is a master regulator of the somatic piRNA pathway. Tj binds to regulatory regions of somatic piRNA factors and the major piRNA cluster flamenco , which carries a Tj-bound enhancer downstream of its promoter. Depletion of Tj in somatic follicle cells causes downregulation of piRNA factors, loss of flam expression and de-repression of gypsy -family TEs. We propose that the arms race between the host and TEs led to the co-evolution of promoters in piRNA pathway genes as well as TE regulatory regions that both rely on a shared transcription factor. Highlights - Traffic jam (Tj) acts as a master regulator of the somatic piRNA pathway in Drosophila . - Tj regulates a network of piRNA pathway genes, mirroring the gene-regulatory mechanism of A-MYB in the mouse testis and Ovo in fly ovaries. - Cis -regulatory elements with Tj motifs are present at the promoters of somatic piRNA pathway genes. - The expression of the flamenco piRNA cluster is directly controlled by Tj.
Collapse
|
39
|
Thomas HF, Feng S, Haslhofer F, Huber M, García Gallardo M, Loubiere V, Vanina D, Pitasi M, Stark A, Buecker C. Enhancer cooperativity can compensate for loss of activity over large genomic distances. Mol Cell 2025; 85:362-375.e9. [PMID: 39626663 DOI: 10.1016/j.molcel.2024.11.008] [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: 01/02/2024] [Revised: 10/01/2024] [Accepted: 11/07/2024] [Indexed: 01/19/2025]
Abstract
Enhancers are short DNA sequences that activate their target promoter from a distance; however, increasing the genomic distance between the enhancer and the promoter decreases expression levels. Many genes are controlled by combinations of multiple enhancers, yet the interaction and cooperation of individual enhancer elements are not well understood. Here, we developed a synthetic platform in mouse embryonic stem cells that allows building complex regulatory landscapes from the bottom up. We tested the system by integrating individual enhancers at different distances and confirmed that the strength of an enhancer contributes to how strongly it is affected by increased genomic distance. Furthermore, synergy between two enhancer elements depends on the distance at which the two elements are integrated: introducing a weak enhancer between a strong enhancer and the promoter strongly increases reporter gene expression, allowing enhancers to activate from increased genomic distances.
Collapse
Affiliation(s)
- Henry F Thomas
- Max Perutz Laboratories, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology, and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; Vienna BioCenter PhD Program, a Doctoral School of the University of Vienna and the Medical University of Vienna, 1030 Vienna, Austria.
| | - Songjie Feng
- Max Perutz Laboratories, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology, and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; Vienna BioCenter PhD Program, a Doctoral School of the University of Vienna and the Medical University of Vienna, 1030 Vienna, Austria
| | - Felix Haslhofer
- Max Perutz Laboratories, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology, and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Marie Huber
- Max Perutz Laboratories, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology, and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - María García Gallardo
- Max Perutz Laboratories, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology, and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; Vienna BioCenter PhD Program, a Doctoral School of the University of Vienna and the Medical University of Vienna, 1030 Vienna, Austria
| | - Vincent Loubiere
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Daria Vanina
- Max Perutz Laboratories, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology, and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Mattia Pitasi
- Max Perutz Laboratories, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology, and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; Vienna BioCenter PhD Program, a Doctoral School of the University of Vienna and the Medical University of Vienna, 1030 Vienna, Austria
| | - Alexander Stark
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria; Medical University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Christa Buecker
- Max Perutz Laboratories, Vienna BioCenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology, and Genetics, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria.
| |
Collapse
|
40
|
Hunker AC, Mich JK, Taskin N, Torkelson A, Pham T, Bertagnolli D, Chakka AB, Chakrabarty R, Donadio NP, Ferrer R, Gasperini M, Goldy J, Guzman JB, Jin K, Khem S, Kutsal R, Lalanne JB, Martinez RA, Newman D, Pena N, Shapovalova NV, Weed N, Zhou T, Yao S, Shendure J, Smith KA, Lein ES, Tasic B, Levi BP, Ting JT. Technical and biological sources of noise confound multiplexed enhancer AAV screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633018. [PMID: 39868122 PMCID: PMC11760716 DOI: 10.1101/2025.01.14.633018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Cis -acting regulatory enhancer elements are valuable tools for gaining cell type-specific genetic access. Leveraging large chromatin accessibility atlases, putative enhancer sequences can be identified and deployed in adeno-associated virus (AAV) delivery platforms. However, a significant bottleneck in enhancer AAV discovery is charting their detailed expression patterns in vivo , a process that currently requires gold-standard one-by-one testing. Here we present a barcoded multiplex strategy for screening enhancer AAVs at cell type resolution using single cell RNA sequencing and taxonomy mapping. We executed a proof-of-concept study using a small pool of validated enhancer AAVs expressing in a variety of neuronal and non-neuronal cell types across the mouse brain. Unexpectedly, we encountered substantial technical and biological noise including chimeric packaging products, necessitating development of novel techniques to accurately deconvolve enhancer expression patterns. These results underscore the need for improved methods to mitigate noise and highlight the complexity of enhancer AAV biology in vivo .
Collapse
|
41
|
Zhu X, Huang L, Wang C, Li G, Deng B, Kong D, Wang X, Chang R, Gu Y, Wen Q, Kong S, Liu Y, Zhang Y. Uncovering the whole genome silencers of human cells via Ss-STARR-seq. Nat Commun 2025; 16:723. [PMID: 39820458 PMCID: PMC11739512 DOI: 10.1038/s41467-025-55852-8] [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: 01/29/2024] [Accepted: 12/31/2024] [Indexed: 01/19/2025] Open
Abstract
Silencers, the yin to enhancers' yang, play a pivotal role in fine-tuning gene expression throughout the genome. However, despite their recognized importance, comprehensive identification of these regulatory elements in the genome is still in its early stages. We developed a method called Ss-STARR-seq to directly determine the activity of silencers in the whole genome. In this study, we applied Ss-STARR-seq to human cell lines K562, LNCaP, and 293 T, and identified 134,171, 137,753, and 125,307 silencers on a genome-wide scale, respectively, these silencers function in various cells in a cell-specific manner. Silencers exhibited a substantial enrichment of transcriptional-inhibitory motifs, including REST, and demonstrated overlap with the binding sites of repressor transcription factors within the endogenous environment. Interestingly, H3K27me3 did not reflect silencer activity but facilitated the silencer's inhibitory role on gene expression. Additionally, the silencer did not have any significant histone markers at the genome-wide level. Our findings unveil that aspect-silencers not only transition into enhancers throughout diverse cell lines but also achieve functional conversion with insulators. Regarding to biological effects, knockout experiments underscored the functional redundancy and specificity of silencers in regulating gene expression and cell proliferation. In summary, this study pioneers the elucidation of the genome-wide silencer landscape in human cells, delineates their global regulatory features, and identifies specific silencers influencing cancer cell proliferation.
Collapse
Affiliation(s)
- Xiusheng Zhu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Lei Huang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Chao Wang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Guoli Li
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Biao Deng
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Dashuai Kong
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xiaoxiao Wang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Rongrong Chang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yi Gu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Qiuhan Wen
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Siyuan Kong
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yuwen Liu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Yubo Zhang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| |
Collapse
|
42
|
Dong SS, Duan YY, Zhu RJ, Jia YY, Chen JX, Huang XT, Tang SH, Yu K, Shi W, Chen XF, Jiang F, Hao RH, Liu Y, Liu Z, Guo Y, Yang TL. Systematic functional characterization of non-coding regulatory SNPs associated with central obesity. Am J Hum Genet 2025; 112:116-134. [PMID: 39753113 PMCID: PMC11739881 DOI: 10.1016/j.ajhg.2024.11.005] [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: 08/05/2024] [Revised: 11/03/2024] [Accepted: 11/13/2024] [Indexed: 01/20/2025] Open
Abstract
Central obesity is associated with higher risk of developing a wide range of diseases independent of overall obesity. Genome-wide association studies (GWASs) have identified more than 300 susceptibility loci associated with central obesity. However, the functional understanding of these loci is limited by the fact that most loci are in non-coding regions. To address this issue, our study first prioritized 2,034 single-nucleotide polymorphisms (SNPs) based on fine-mapping and epigenomic annotation analysis. Subsequently, we employed self-transcribing active regulatory region sequencing (STARR-seq) to systematically evaluate the enhancer activity of these prioritized SNPs. The resulting data analysis identified 141 SNPs with allelic enhancer activity. Further analysis of allelic transcription factor (TF) binding prioritized 20 key TFs mediating the central-obesity-relevant genetic regulatory network. Finally, as an example, we illustrate the molecular mechanisms of how rs8079062 acts as an allele-specific enhancer to regulate the expression of its targeted RNF157. We also evaluated the role of RNF157 in the adipogenic differentiation process. In conclusion, our results provide an important resource for understanding the genetic regulatory mechanisms underlying central obesity.
Collapse
Affiliation(s)
- Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Yuan-Yuan Duan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Ren-Jie Zhu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Ying-Ying Jia
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Jia-Xin Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Xiao-Ting Huang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Shi-Hao Tang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Ke Yu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Wei Shi
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Xiao-Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Feng Jiang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Ruo-Han Hao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Zhongbo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| |
Collapse
|
43
|
Shireen H, Batool F, Khatoon H, Parveen N, Sehar NU, Hussain I, Ali S, Abbasi AA. Predicting genome-wide tissue-specific enhancers via combinatorial transcription factor genomic occupancy analysis. FEBS Lett 2025; 599:100-119. [PMID: 39367524 DOI: 10.1002/1873-3468.15030] [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: 07/14/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 10/06/2024]
Abstract
Enhancers are non-coding cis-regulatory elements crucial for transcriptional regulation. Mutations in enhancers can disrupt gene regulation, leading to disease phenotypes. Identifying enhancers and their tissue-specific activity is challenging due to their lack of stereotyped sequences. This study presents a sequence-based computational model that uses combinatorial transcription factor (TF) genomic occupancy to predict tissue-specific enhancers. Trained on diverse datasets, including ENCODE and Vista enhancer browser data, the model predicted 25 000 forebrain-specific cis-regulatory modules (CRMs) in the human genome. Validation using biochemical features, disease-associated SNPs, and in vivo zebrafish analysis confirmed its effectiveness. This model aids in predicting enhancers lacking well-characterized chromatin features, complementing experimental approaches in tissue-specific enhancer discovery.
Collapse
Affiliation(s)
- Huma Shireen
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fatima Batool
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hizran Khatoon
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Nazia Parveen
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Noor Us Sehar
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Irfan Hussain
- Centre for Regenerative Medicine and Stem Cells Research, Agha Khan University hospital, Karachi, Pakistan
| | - Shahid Ali
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL, USA
| | - Amir Ali Abbasi
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
44
|
Wang M, Yang X, Wu Q. High-resolution dissection of human cell type-specific enhancers in cis and trans activities. Genomics 2025; 117:110985. [PMID: 39755338 DOI: 10.1016/j.ygeno.2025.110985] [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: 07/17/2024] [Revised: 09/26/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
The spatiotemporal-specific gene expression is regulated by cell type-specific regulatory elements. Here we selected the H3K4me1-associated DNA sequences as candidate enhancers in two different human cell lines and performed ChIP-STARR-seq to quantify the cell-type-specific enhancer activities with high-resolution. We investigated how the activity landscape of enhancers would change when transferred from native cells (cis activity) to another cell lines (trans activity). We obtained enhancers cis activity maps and trans activity maps in two different cell lines. The cis and trans activity maps enabled us to identify cell type-specific active enhancers, with enrichment of motifs of differentially expressed TFs. Comparisons between the cis and trans activity maps revealed general consistent regulatory property with different levels of activity in two cell lines, suggesting sequence intrinsic regulatory properties remain similar in different types of cells. This study provides a new perspective on sequence intrinsic enhancer activities in different types of cells.
Collapse
Affiliation(s)
- Meng Wang
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, PR China.
| | - Xiaoxu Yang
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, PR China
| | - Qixi Wu
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, PR China.
| |
Collapse
|
45
|
Chang TY, Waxman DJ. HDI-STARR-seq: Condition-specific enhancer discovery in mouse liver in vivo. BMC Genomics 2024; 25:1240. [PMID: 39716078 DOI: 10.1186/s12864-024-11162-9] [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: 06/10/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND STARR-seq and other massively-parallel reporter assays are widely used to discover functional enhancers in transfected cell models, which can be confounded by plasmid vector-induced type-I interferon immune responses and lack the multicellular environment and endogenous chromatin state of complex mammalian tissues. RESULTS We describe HDI-STARR-seq, which combines STARR-seq plasmid library delivery to the liver, by hydrodynamic tail vein injection (HDI), with reporter RNA transcriptional initiation driven by a minimal Albumin promoter, which we show is essential for mouse liver STARR-seq enhancer activity assayed 7 days after HDI. Importantly, little or no vector-induced innate type-I interferon responses were observed. Comparisons of HDI-STARR-seq activity between male and female mouse livers and in livers from males treated with an activating ligand of the transcription factor (TF) CAR (Nr1i3) identified many condition-dependent enhancers linked to condition-specific gene expression. Further, thousands of active liver enhancers were identified using a high complexity STARR-seq library comprised of ~ 50,000 genomic regions released by DNase-I digestion of mouse liver nuclei. When compared to stringently inactive library sequences, the active enhancer sequences identified were highly enriched for liver open chromatin regions with activating histone marks (H3K27ac, H3K4me1, H3K4me3), were significantly closer to gene transcriptional start sites, and were significantly depleted of repressive (H3K27me3, H3K9me3) and transcribed region histone marks (H3K36me3). CONCLUSION HDI-STARR-seq offers substantial improvements over current methodologies for large scale, functional profiling of enhancers, including condition-dependent enhancers, in liver tissue in vivo, and can be adapted to characterize enhancer activities in a variety of species and tissues by selecting suitable tissue- and species-specific promoter sequences.
Collapse
Affiliation(s)
- Ting-Ya Chang
- Departments of Biology and Biomedical Engineering, and Bioinformatics Program, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA
| | - David J Waxman
- Departments of Biology and Biomedical Engineering, and Bioinformatics Program, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA.
| |
Collapse
|
46
|
Falo-Sanjuan J, Diaz-Tirado Y, Turner MA, Rourke O, Davis J, Medrano C, Haines J, McKenna J, Karshenas A, Eisen MB, Garcia HG. Targeted mutagenesis of specific genomic DNA sequences in animals for the in vivo generation of variant libraries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598328. [PMID: 38915503 PMCID: PMC11195090 DOI: 10.1101/2024.06.10.598328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Understanding how the number, placement and affinity of transcription factor binding sites dictates gene regulatory programs remains a major unsolved challenge in biology, particularly in the context of multicellular organisms. To uncover these rules, it is first necessary to find the binding sites within a regulatory region with high precision, and then to systematically modulate this binding site arrangement while simultaneously measuring the effect of this modulation on output gene expression. Massively parallel reporter assays (MPRAs), where the gene expression stemming from 10,000s of in vitro-generated regulatory sequences is measured, have made this feat possible in high-throughput in single cells in culture. However, because of lack of technologies to incorporate DNA libraries, MPRAs are limited in whole organisms. To enable MPRAs in multicellular organisms, we generated tools to create a high degree of mutagenesis in specific genomic loci in vivo using base editing. Targeting GFP integrated in the genome of Drosophila cell culture and whole animals as a case study, we show that the base editor AIDevoCDA1 stemming from sea lamprey fused to nCas9 is highly mutagenic. Surprisingly, longer gRNAs increase mutation efficiency and expand the mutating window, which can allow the introduction of mutations in previously untargetable sequences. Finally, we demonstrate arrays of >20 gRNAs that can efficiently introduce mutations along a 200bp sequence, making it a promising tool to test enhancer function in vivo in a high throughput manner.
Collapse
Affiliation(s)
- Julia Falo-Sanjuan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Yuliana Diaz-Tirado
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Meghan A. Turner
- Biophysics Graduate Group, University of California, Berkeley, CA, USA
| | - Olivia Rourke
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Julian Davis
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Claudia Medrano
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Jenna Haines
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Joey McKenna
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Arman Karshenas
- Biophysics Graduate Group, University of California, Berkeley, CA, USA
| | - Michael B. Eisen
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Department of Integrative Biology, University of California, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| | - Hernan G. Garcia
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Department of Physics, University of California, Berkeley, CA, USA
- Institute for Quantitative Biosciences-QB3, University of California, Berkeley, CA, USA
- Chan Zuckerberg Biohub – San Francisco, San Francisco, CA, USA
- Biophysics Graduate Group, University of California, Berkeley, CA, USA
| |
Collapse
|
47
|
Baniulyte G, McCann AA, Woodstock DL, Sammons MA. Crosstalk between paralogs and isoforms influences p63-dependent regulatory element activity. Nucleic Acids Res 2024; 52:13812-13831. [PMID: 39565223 DOI: 10.1093/nar/gkae1143] [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: 05/17/2024] [Revised: 10/04/2024] [Accepted: 11/01/2024] [Indexed: 11/21/2024] Open
Abstract
The p53 family of transcription factors (p53, p63 and p73) regulate diverse organismal processes including tumor suppression, maintenance of genome integrity and the development of skin and limbs. Crosstalk between transcription factors with highly similar DNA binding profiles, like those in the p53 family, can dramatically alter gene regulation. While p53 is primarily associated with transcriptional activation, p63 mediates both activation and repression. The specific mechanisms controlling p63-dependent gene regulatory activity are not well understood. Here, we use massively parallel reporter assays (MPRA) to investigate how local DNA sequence context influences p63-dependent transcriptional activity. Most regulatory elements with a p63 response element motif (p63RE) activate transcription, although binding of the p63 paralog, p53, drives a substantial proportion of that activity. p63RE sequence content and co-enrichment with other known activating and repressing transcription factors, including lineage-specific factors, correlates with differential p63RE-mediated activities. p63 isoforms dramatically alter transcriptional behavior, primarily shifting inactive regulatory elements towards high p63-dependent activity. Our analysis provides novel insight into how local sequence and cellular context influences p63-dependent behaviors and highlights the key, yet still understudied, role of transcription factor paralogs and isoforms in controlling gene regulatory element activity.
Collapse
Affiliation(s)
- Gabriele Baniulyte
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY 12222, USA
| | - Abby A McCann
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY 12222, USA
| | - Dana L Woodstock
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, 1400 Washington Ave, Albany, NY 12222, USA
| |
Collapse
|
48
|
Klagkou E, Valakos D, Foutadakis S, Polyzos A, Papadopoulou A, Vatsellas G, Thanos D. An Unbiased Approach to Identifying Cellular Reprogramming-Inducible Enhancers. Int J Mol Sci 2024; 25:13128. [PMID: 39684837 DOI: 10.3390/ijms252313128] [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: 10/27/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Cellular reprogramming of somatic cells towards induced pluripotency is a multistep stochastic process mediated by the transcription factors Oct4, Sox2, Klf4 and c-Myc (OSKM), which orchestrate global epigenetic and transcriptional changes. We performed a large-scale analysis of integrated ChIP-seq, ATAC-seq and RNA-seq data and revealed the spatiotemporal highly dynamic pattern of OSKM DNA binding during reprogramming. We found that OSKM show distinct temporal patterns of binding to different classes of pluripotency-related enhancers. Genes involved in reprogramming are regulated by the coordinated activity of multiple enhancers, which are sequentially bound by OSKM for strict transcriptional control. Based on these findings, we developed an unbiased approach to identify Reprogramming-Inducible Enhancers (RIEs), constructed enhancer-traps and isolated cells undergoing reprogramming in real time. We used a representative RIE taken from the Upp1 gene fused to Gfp and isolated cells at different time-points during reprogramming and found that they have unique developmental capacities as they are reprogrammed with high efficiency due to their distinct molecular signatures. In conclusion, our experiments have led to the development of an unbiased method to identify and isolate reprogrammable cells in real time by exploiting the functional dynamics of OSKM, which can be used as efficient reprogramming biomarkers.
Collapse
Affiliation(s)
- Eleftheria Klagkou
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, Zografou, 15772 Athens, Greece
| | - Dimitrios Valakos
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, Zografou, 15772 Athens, Greece
| | - Spyros Foutadakis
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Hellenic Institute for the Study of Sepsis (HISS), 11528 Athens, Greece
| | - Alexander Polyzos
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill, Cornell Medicine, New York, NY 10065, USA
| | - Angeliki Papadopoulou
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland
- Department of Biology, School of Sciences and Engineering, University of Crete, 70013 Irakleio, Greece
| | - Giannis Vatsellas
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
| | - Dimitris Thanos
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
| |
Collapse
|
49
|
Hofbauer L, Pleyer LM, Reiter F, Schleiffer A, Vlasova A, Serebreni L, Huang A, Stark A. A genome-wide screen identifies silencers with distinct chromatin properties and mechanisms of repression. Mol Cell 2024; 84:4503-4521.e14. [PMID: 39571581 DOI: 10.1016/j.molcel.2024.10.041] [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: 02/12/2024] [Revised: 08/20/2024] [Accepted: 10/29/2024] [Indexed: 12/08/2024]
Abstract
Differential gene transcription enables development and homeostasis in all animals and is regulated by two major classes of distal cis-regulatory DNA elements (CREs): enhancers and silencers. Although enhancers have been thoroughly characterized, the properties and mechanisms of silencers remain largely unknown. By an unbiased genome-wide functional screen in Drosophila melanogaster S2 cells, we discover a class of silencers that bind one of three transcription factors (TFs) and are generally not included in chromatin-defined CRE catalogs as they mostly lack detectable DNA accessibility. The silencer-binding TF CG11247, which we term Saft, safeguards cell fate decisions in vivo and functions via a highly conserved domain we term zinc-finger-associated C-terminal (ZAC) and the corepressor G9a, independently of G9a's H3K9-methyltransferase activity. Overall, our identification of silencers with unexpected properties and mechanisms has important implications for the understanding and future study of repressive CREs, as well as the functional annotation of animal genomes.
Collapse
Affiliation(s)
- Lorena Hofbauer
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Lisa-Marie Pleyer
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Franziska Reiter
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Alexander Schleiffer
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Anna Vlasova
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Leonid Serebreni
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Annie Huang
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Alexander Stark
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria; Medical University of Vienna, Vienna BioCenter (VBC), Vienna, Austria.
| |
Collapse
|
50
|
Moss ND, Lollis D, Silver DL. How our brains are built: emerging approaches to understand human-specific features. Curr Opin Genet Dev 2024; 89:102278. [PMID: 39549607 DOI: 10.1016/j.gde.2024.102278] [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: 06/28/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/18/2024]
Abstract
Understanding what makes us uniquely human is a long-standing question permeating fields from genomics, neuroscience, and developmental biology to medicine. The discovery of human-specific genomic sequences has enabled a new understanding of the molecular features of human brain evolution. Advances in sequencing, computational, and in vitro screening approaches collectively reveal new roles of uniquely human sequences in regulating gene expression. Here, we review the landscape of human-specific loci and describe how emerging technologies are being used to understand their molecular functions and impact on brain development. We describe current challenges in the field and the potential of integrating new hypotheses and approaches to propel our understanding of the human brain.
Collapse
Affiliation(s)
- Nicole D Moss
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Davoneshia Lollis
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA. https://twitter.com/@_mlollis
| | - Debra L Silver
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences and Duke Regeneration Center, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|