1
|
Qiu Q, Tong X, Zhu M, Liu Z, Pang H, Li L, Feng Y, Hu X, Gong C. Changes in gene expression levels caused by H3K9me3/H3K9ac modifications are associated with BmCPV infection in Bombyx mori. Virulence 2025; 16:2510535. [PMID: 40418637 PMCID: PMC12118411 DOI: 10.1080/21505594.2025.2510535] [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: 08/04/2024] [Revised: 05/02/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Abstract
Changes in chromatin accessibility caused by histone modifications regulate gene transcription. However, little is known about associations between gene expression changes caused by histone modifications and viral infections. We investigate the midguts of silkworms infected with Bombyx mori cypovirus (BmCPV) at 48 h and 96 h post infection (CPV48 and CPV96), and corresponding midguts of uninfected silkworms (GUT48 and GUT96) using CUT&Tag-seq and RNA-seq. We report H3K9me3, H3K9ac, and gene expression profiles at the genome-wide level to change with BmCPV infection. Differential H3K9me3 peak-related genes were mainly enriched in MAPK, Wnt, and Hippo signalling pathways; Differential H3K9ac peaks-related genes were mainly enriched in the Hippo signalling, apoptosis, and citrate cycle pathways; and differentially expressed genes (DEGs) were mainly enriched in carbon metabolism, protein processing in endoplasmic reticulum, and glycolysis/gluconeogenesis pathways. Integration analysis between H3K9me3/H3K9ac peaks and gene expression revealed changes in gene expression profiles to be associated with alteration of H3K9me3/H3K9ac at promoters; gene expression correlates negatively with corresponding H3K9me3 signals in gene bodies, and positively with corresponding H3K9ac signals at the transcription start site. Intersection genes with log2foldchange of both CUT&Tag-seq peak and RNA-seq FPKM > 1 were screened and annotated. Genes shared by differential H3K9me3 peak-related genes and DEGs were enriched in insect hormone biosynthesis, MAPK signalling, and TGF-beta signalling pathways, and genes shared by differential H3K9ac peak-related genes and DEGs were enriched in glycolysis/gluconeogenesis, TGF-beta signalling, and mitophagy pathways. These results indicate that BmCPV regulates gene expression through H3K9me3/H3K9ac.
Collapse
Affiliation(s)
- Qunnan Qiu
- School of Life Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural Biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| | - Xinyu Tong
- School of Life Sciences, Soochow University, Suzhou, China
| | - Min Zhu
- School of Life Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural Biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| | - Zhe Liu
- School of Life Sciences, Soochow University, Suzhou, China
| | - Huilin Pang
- School of Life Sciences, Soochow University, Suzhou, China
| | - Liuyang Li
- School of Life Sciences, Soochow University, Suzhou, China
| | - Yongjie Feng
- School of Life Sciences, Soochow University, Suzhou, China
| | - Xiaolong Hu
- School of Life Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural Biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| | - Chengliang Gong
- School of Life Sciences, Soochow University, Suzhou, China
- Agricultural Biotechnology Research Institute, Agricultural Biotechnology and Ecological Research Institute, Soochow University, Suzhou, China
| |
Collapse
|
2
|
Carter R, Storey KB. Levels of post-translationally modified histones in ground squirrel livers are altered during deep torpor. Cryobiology 2025; 119:105256. [PMID: 40345109 DOI: 10.1016/j.cryobiol.2025.105256] [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: 01/24/2025] [Revised: 03/18/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025]
Abstract
Thirteen-lined ground squirrels (Ictidomys tridecemlineatus) are obligate hibernators capable of reducing their metabolic rates by up to 99 % during winter. Their ability to remain dormant without food for an extended period in cold conditions has made them compelling subjects for research. Developing a clearer understanding of mechanisms surrounding the pre-transcriptional control of hibernating tissues is crucial for cryobiological applications such as organ preservation. Thus, we investigated the differential expression of 24 modified histones (MH) in the livers of torpid and euthermic free-ranging ground squirrels by immunoblotting histone-enriched extracts (p < 0.05). We identified the torpor-responsive downregulation of multiple permissive MHs (H2BK5ac, H3K18ac, H3K23ac, H3K27ac, H3K4me2, H3K4me3, H4K20me1, H4R3me2s), including total H2B and H4, while the linker histone H1.0 was the only histone species that was upregulated. The present study provides valuable insights into the involvement of histone post-translational modifications in the epigenetic landscape of deeply torpid ground squirrel livers.
Collapse
Affiliation(s)
- Remy Carter
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada.
| | - Kenneth B Storey
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada.
| |
Collapse
|
3
|
Xu L, Wang Y, Li X, Hu Q, Adamkova V, Xu J, Harris CJ, Ausin I. H3K4me3 binding ALFIN-LIKE proteins recruit SWR1 for gene-body deposition of H2A.Z. Genome Biol 2025; 26:137. [PMID: 40399998 PMCID: PMC12096798 DOI: 10.1186/s13059-025-03605-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 05/06/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The H2A.Z histone variant is highly enriched over gene bodies, playing an essential role in several genome-templated processes, including transcriptional regulation and epigenetic patterning across eukaryotes. Deposition of H2A.Z is mediated by the SWR1 remodeling complex. How SWR1 is directed to gene bodies is largely unknown. RESULTS Here, we show that ALFIN-LIKE (AL) proteins are responsible for H2A.Z gene body patterning in Arabidopsis. AL proteins encode H3K4me3-binding PHD domains, and by ChIP-seq, we confirm preferential binding of AL5 to H3K4me3 over H3K4me1/2 in planta. We observe a global reduction in H2A.Z in al septuple mutants (al7m), especially over H3K4me3-enriched genic regions. While MBD9 recruits SWR1 to nucleosome-free regions, ALs act non-redundantly with MBD9 for deposition of H2A.Z. Notably, al7m mutants show severe developmental abnormalities and upregulation of H2A.Z gene body-enriched responsive genes. CONCLUSIONS Therefore, we propose a model whereby AL proteins direct gene body enrichment of H2A.Z by recruiting SWR1 to H3K4me3-containing responsive genes.
Collapse
Affiliation(s)
- Linhao Xu
- Department of Plant Sciences, University of Cambridge, Cambridge, CB2 3EA, UK
| | - Yafei Wang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Life Sciences and Institute of Future Agriculture, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xueying Li
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Life Sciences and Institute of Future Agriculture, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Qin Hu
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Life Sciences and Institute of Future Agriculture, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Vanda Adamkova
- Department of Plant Sciences, University of Cambridge, Cambridge, CB2 3EA, UK
| | - Junjie Xu
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Life Sciences and Institute of Future Agriculture, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - C Jake Harris
- Department of Plant Sciences, University of Cambridge, Cambridge, CB2 3EA, UK.
| | - Israel Ausin
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Life Sciences and Institute of Future Agriculture, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
4
|
Yang W, Wang XQ, Wei F, Yu J, Liu Y, Dou Y. Revealing long-range heterogeneous organization of nucleoproteins with 6mA footprinting by ipdTrimming. Genome Biol 2025; 26:136. [PMID: 40399934 PMCID: PMC12096735 DOI: 10.1186/s13059-025-03592-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/27/2025] [Indexed: 05/23/2025] Open
Abstract
Enabled by long-read sequencing technologies, particularly Single Molecule, Real-Time sequencing, N6-methyladenine (6mA) footprinting is a transformative methodology for revealing the heterogenous and dynamic distribution of nucleosomes and other DNA-binding proteins. Here, we present ipdTrimming, a novel 6mA-calling pipeline that outperforms existing tools in both computational efficiency and accuracy. Utilizing this optimized experimental and computational framework, we are able to map nucleosome positioning and transcription factor occupancy in nuclear DNA and establish high-resolution, long-range binding events in mitochondrial DNA. Our study highlights the potential of 6mA footprinting to capture coordinated nucleoprotein binding and to unravel epigenetic heterogeneity.
Collapse
Affiliation(s)
- Wentao Yang
- Department of Cancer Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Xue Qing Wang
- Department of Cancer Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Fan Wei
- Department of Cancer Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jingqi Yu
- Department of Cancer Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Yifan Liu
- Department of Cancer Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| | - Yali Dou
- Department of Cancer Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
5
|
Huang H, Baxter AE, Zhang Z, Good CR, Alexander KA, Chen Z, Garcia PAA, Samareh P, Collins SM, Glastad KM, Wang L, Donahue G, Manne S, Giles JR, Shi J, Berger SL, Wherry EJ. Deciphering the role of histone modifications in memory and exhausted CD8 T cells. Sci Rep 2025; 15:17359. [PMID: 40389726 PMCID: PMC12089470 DOI: 10.1038/s41598-025-99804-0] [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: 12/20/2024] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
Exhausted CD8 T cells (TEX) arising during chronic infections and cancer have reduced functional capacity and limited fate flexibility that prevents optimal disease control and response to immunotherapies. Compared to memory (TMEM) cells, TEX have a unique open chromatin landscape underlying a distinct gene expression program. How TEX transcriptional and epigenetic landscapes are regulated through histone post-translational modifications (hPTMs) remains unclear. Here, we profiled key activating (H3K27ac and H3K4me3) and repressive (H3K27me3 and H3K9me3) histone modifications in naive CD8 T cells (TN), TMEM and TEX. We identified H3K27ac-associated super-enhancers that distinguish TN, TMEM and TEX, along with key transcription factor networks predicted to regulate these different transcriptional landscapes. Promoters of some key genes were poised in TN, but activated in TMEM or TEX whereas other genes poised in TN were repressed in TMEM or TEX, indicating that both repression and activation of poised genes may enforce these distinct cell states. Moreover, narrow peaks of repressive H3K9me3 were associated with increased gene expression in TEX, suggesting an atypical role for this modification. These data indicate that beyond chromatin accessibility, hPTMs differentially regulate specific gene expression programs of TEX compared to TMEM through both activating and repressive pathways.
Collapse
Affiliation(s)
- Hua Huang
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Amy E Baxter
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Zhen Zhang
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, Anhui, China
| | - Charly R Good
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Katherine A Alexander
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Cold Spring Harbor Laboratories, Cold Spring Harbor, NY, 11724, USA
| | - Zeyu Chen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Cell Biology and Pathology, Harvard Medical School, Boston, MA, 02115, USA
| | - Paula A Agudelo Garcia
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Parisa Samareh
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sierra M Collins
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Karl M Glastad
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Biology, University of Rochester, Rochester, NY, 14620, USA
| | - Lu Wang
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Biochemistry and Structural Biology, University of Texas Health Sciences Center at San Antonio, San Antonio, TX, 78229, USA
| | - Gregory Donahue
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sasikanth Manne
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Josephine R Giles
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Junwei Shi
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Shelley L Berger
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Tallan A, Kucinski J, Sunkel B, Taslim C, LaHaye S, Liu Q, Qi J, Wang M, Kendall GC, Stanton BZ. Highly quantitative measurement of differential protein-genome binding with PerCell chromatin sequencing. CELL REPORTS METHODS 2025:101052. [PMID: 40393455 DOI: 10.1016/j.crmeth.2025.101052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/21/2025] [Accepted: 04/24/2025] [Indexed: 05/22/2025]
Abstract
Quantitative comparison of ChIP-seq profiling between experimental conditions or samples remains technically challenging for the epigenetics field. Here, we report a strategy combining the use of well-defined cellular spike-in ratios of orthologous species' chromatin and a bioinformatic analysis pipeline to facilitate highly quantitative comparisons of 2D chromatin sequencing across experimental conditions. We find that the PerCell methodology results in efficient and consistent levels of spike-in vs. experimental genomic reads. We demonstrate use of the method and pipeline to enable quantitative, internally normalized chromatin sequencing on zebrafish embryos and human cancer cells. Overall, we propose the PerCell method to enable cross-species comparative epigenomics and promote uniformity of data analyses and sharing across labs.
Collapse
Affiliation(s)
- Alexi Tallan
- Nationwide Children's Hospital, Center for Childhood Cancer Research, Columbus, OH, USA; Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH, USA
| | - Jack Kucinski
- Nationwide Children's Hospital, Center for Childhood Cancer Research, Columbus, OH, USA; Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH, USA
| | - Benjamin Sunkel
- Nationwide Children's Hospital, Center for Childhood Cancer Research, Columbus, OH, USA
| | - Cenny Taslim
- Nationwide Children's Hospital, Center for Childhood Cancer Research, Columbus, OH, USA
| | - Stephanie LaHaye
- Nationwide Children's Hospital, Center for Childhood Cancer Research, Columbus, OH, USA
| | - Qi Liu
- Cancer Biology Department, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jun Qi
- Cancer Biology Department, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Meng Wang
- Nationwide Children's Hospital, Center for Childhood Cancer Research, Columbus, OH, USA.
| | - Genevieve C Kendall
- Nationwide Children's Hospital, Center for Childhood Cancer Research, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| | - Benjamin Z Stanton
- Nationwide Children's Hospital, Center for Childhood Cancer Research, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Biological Chemistry & Pharmacology, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
7
|
Alavattam KG, Dickson BM, Hirano R, Dell R, Tsukiyama T. ChIP-seq Data Processing and Relative and Quantitative Signal Normalization for Saccharomyces cerevisiae. Bio Protoc 2025; 15:e5299. [PMID: 40364978 PMCID: PMC12067309 DOI: 10.21769/bioprotoc.5299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Chromatin immunoprecipitation with high-throughput sequencing (ChIP-seq) is a widely used technique for genome-wide analyses of protein-DNA interactions. This protocol provides a guide to ChIP-seq data processing in Saccharomyces cerevisiae, with a focus on signal normalization to address data biases and enable meaningful comparisons within and between samples. Designed for researchers with minimal bioinformatics experience, it includes practical overviews and refers to scripting examples for key tasks, such as configuring computational environments, trimming and aligning reads, processing alignments, and visualizing signals. This protocol employs the sans-spike-in method for quantitative ChIP-seq (siQ-ChIP) and normalized coverage for absolute and relative comparisons of ChIP-seq data, respectively. While spike-in normalization, which is semiquantitative, is addressed for context, siQ-ChIP and normalized coverage are recommended as mathematically rigorous and reliable alternatives. Key features • ChIP-seq data processing workflow for Linux and macOS integrating data acquisition, trimming, alignment, processing, and multiple forms of signal computation, with a focus on reproducibility. • ChIP-seq signal generation using siQ-ChIP to quantify absolute IP efficiency-providing a rigorous alternative to spike-in normalization-and normalized coverage for relative comparisons. • Broad applicability demonstrated with Saccharomyces cerevisiae (experimental) and Schizosaccharomyces pombe (spike-in) data but suitable for ChIP-seq in any species. • In-depth notes and troubleshooting guide users through setup challenges and key concepts in basic bioinformatics, data processing, and signal computation. Graphical overview Flowchart depicting ChIP-seq data processing steps covered in this protocol.
Collapse
Affiliation(s)
- Kris G. Alavattam
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Rina Hirano
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Rachel Dell
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Toshio Tsukiyama
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
8
|
Zhao Z, Dai X, Jiang G, Lin F. Absent, Small, or Homeotic 2-Like-Mediated H3K4 Methylation and Nephrogenesis. J Am Soc Nephrol 2025; 36:798-811. [PMID: 39774048 PMCID: PMC12059113 DOI: 10.1681/asn.0000000600] [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: 09/04/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Key Points Deficits in nephron numbers are associated with higher risk of adult-onset kidney disease seen in congenital anomalies of the kidney and urinary tract. Mouse model experiments suggested that absent, small, or homeotic 2-like was vital for kidney development by activating cell cycle genes through histone methylation. Our findings identified absent, small, or homeotic 2-like–regulated genes as a potential target for treating congenital anomalies of the kidney and urinary tract. Background Many congenital anomalies of the kidney and urinary tract involve deficits in the number of nephrons, which are associated with a higher risk of hypertension and CKD later in life. Prior work has implicated histone modifications in regulating kidney lineage–specific gene transcription and nephron endowment. Our earlier study suggested that absent, small, or homeotic 2-like (ASH2L), a core subunit of the H3K4 methyltransferase complex, plays a role in ureteric bud morphogenesis during mammalian kidney development. However, the potential involvement of ASH2L in nephron formation remains an open question. Methods To investigate the role of ASH2L in nephron development, we inactivated Ash2l specifically in nephron progenitor cells by crossing Six2 -e(Kozak-GFPCre-Wpre-polyA)1 mice with Ash2l fl/fl mice. We used RNA sequencing combined with Cleavage Under Targets and Tagmentation sequencing to screen for gene and epigenomic changes, which were further verified by rescue experiments conducted on ex vivo culture explants. Results Inactivating ASH2L in nephron progenitor cells disrupted H3K4 trimethylation establishment at promoters of genes controlling nephron progenitor cell stemness, differentiation, and cell cycle, inhibiting their progression through the cell cycle and differentiation into epithelial cell types needed to form nephrons. Inhibition of the TGF-β /suppressor of mothers against decapentaplegic signaling pathway partially rescued the dysplastic phenotype of the mutants. Conclusions ASH2L-mediated H3K4 methylation was identified as a novel epigenetic regulator of kidney development. Downregulation of ASH2L expression or H3K4 trimethylation may be linked to congenital anomalies of the kidney and urinary tract.
Collapse
Affiliation(s)
- Ziyi Zhao
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuantong Dai
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gengru Jiang
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Centre for Rare Disease, Shanghai, China
| | - Fujun Lin
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Centre for Rare Disease, Shanghai, China
| |
Collapse
|
9
|
Schmidleithner L, Stüve P, Feuerer M. Transposable elements as instructors of the immune system. Nat Rev Immunol 2025:10.1038/s41577-025-01172-3. [PMID: 40301669 DOI: 10.1038/s41577-025-01172-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2025] [Indexed: 05/01/2025]
Abstract
Transposable elements (TEs) are mobile repetitive nucleic acid sequences that have been incorporated into the genome through spontaneous integration, accounting for almost 50% of human DNA. Even though most TEs are no longer mobile today, studies have demonstrated that they have important roles in different biological processes, such as ageing, embryonic development, and cancer. TEs influence these processes through various mechanisms, including active transposition of TEs contributing to ongoing evolution, transposon transcription generating RNA or protein, and by influencing gene regulation as enhancers. However, how TEs interact with the immune system remains a largely unexplored field. In this Perspective, we describe how TEs might influence different aspects of the immune system, such as innate immune responses, T cell activation and differentiation, and tissue adaptation. Furthermore, TEs can serve as a source of neoantigens for T cells in antitumour immunity. We suggest that TE biology is an important emerging field of immunology and discuss the potential to harness the TE network therapeutically, for example, to improve immunotherapies for cancer and autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
| | - Philipp Stüve
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Markus Feuerer
- Leibniz Institute for Immunotherapy, Regensburg, Germany.
- Chair for Immunology, University Regensburg, Regensburg, Germany.
| |
Collapse
|
10
|
Vinayak V, Basir R, Golloshi R, Toth J, Sant'Anna L, Lakadamyali M, McCord RP, Shenoy VB. Polymer model integrates imaging and sequencing to reveal how nanoscale heterochromatin domains influence gene expression. Nat Commun 2025; 16:3816. [PMID: 40268925 PMCID: PMC12019571 DOI: 10.1038/s41467-025-59001-z] [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: 09/27/2024] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
Chromatin organization regulates gene expression, with nanoscale heterochromatin domains playing a fundamental role. Their size varies with microenvironmental stiffness and epigenetic interventions, but how these factors regulate their formation and influence transcription remains unclear. To address this, we developed a sequencing-informed copolymer model that simulates chromatin evolution through diffusion and active epigenetic reactions. Our model predicts the formation of nanoscale heterochromatin domains and quantifies how domain size scales with epigenetic reaction rates, showing that epigenetic and compaction changes primarily occur at domain boundaries. We validated these predictions via Hi-C and super-resolution imaging of hyperacetylated melanoma cells and identified differential expression of metastasis-related genes through RNA-seq. We validated our findings in hMSCs, where epigenetic reaction rates respond to microenvironmental stiffness. Conclusively, our simulations reveal that heterochromatin domain boundaries regulate gene expression and epigenetic memory. These findings demonstrate how external cues drive chromatin organization and transcriptional memory in development and disease.
Collapse
Affiliation(s)
- Vinayak Vinayak
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ramin Basir
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rosela Golloshi
- Departments of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Giovanis Institute for Translational Cell Biology, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Joshua Toth
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Lucas Sant'Anna
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Melike Lakadamyali
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Patton McCord
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, USA
| | - Vivek B Shenoy
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Sospedra-Arrufat I, Martínez-García PM, Gil-Gálvez A, Alcaina-Caro A, Tena JJ, Nusspaumer G, Lopez-Rios J. Epigenomic signatures of cis-regulatory elements in the developing mouse and pig forelimb. Sci Data 2025; 12:598. [PMID: 40210926 PMCID: PMC11986027 DOI: 10.1038/s41597-025-04946-5] [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: 10/09/2024] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
Cis-regulatory elements (CREs) orchestrate the spatiotemporal regulation of key transcriptional programs. These genomic regions-including promoters, enhancers, and insulators-play a crucial role during embryonic development, and their functional diversification is thought to contribute significantly to the morphological evolution of animal body plans. We performed chromatin immunoprecipitation for various histone modifications (H3K4me3, H3K27ac, and H3K4me1) from equivalent developmental stages of mouse (E11.5) and pig (day 23; D23) to identify active regulatory regions during forelimb development in both species. The overlap of these epigenomic signatures with the pattern of open chromatin allowed us to classify these putative regulatory regions into different chromatin states in mouse and pig limb primordia. Our profiling of the regulatory genome in mouse and pig limb buds offers a valuable resource in the evo-devo field for exploring mechanisms underlying the morphological evolution of the tetrapod limb.
Collapse
Affiliation(s)
- Ismael Sospedra-Arrufat
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, 41013, Seville, Spain
| | - Pedro Manuel Martínez-García
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, 41013, Seville, Spain
| | - Alejandro Gil-Gálvez
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, 41013, Seville, Spain
| | - Ana Alcaina-Caro
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, 41013, Seville, Spain
| | - Juan J Tena
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, 41013, Seville, Spain
| | - Gretel Nusspaumer
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, 41013, Seville, Spain
| | - Javier Lopez-Rios
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, 41013, Seville, Spain.
- Universidad Loyola Andalucía, School of Health Sciences, Seville Campus, 41704, Dos Hermanas, Seville, Spain.
| |
Collapse
|
12
|
Sigurðardóttir H, Eriksson S, Niazi A, Rhodin M, Albertsdóttir E, Kristjansson T, Lindgren G. Genetic influence of a STAU2 frameshift mutation and RELN regulatory elements on performance in Icelandic horses. Sci Rep 2025; 15:11641. [PMID: 40185812 PMCID: PMC11971302 DOI: 10.1038/s41598-025-95593-8] [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: 11/18/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Selection for performance in horse breeding benefits from precise genetic insights at a molecular level, but knowledge remains limited. This study used whole-genome sequences of 39 elite and non-elite Icelandic horses to identify candidate causal variants linked to previously identified haplotypes in the STAU2 and RELN genes affecting pace and other gaits. A frameshift variant in linkage disequilibrium with the previously identified haplotypes in the STAU2 gene (r2 = 0.85) was identified within a predicted STAU2 transcript. This variant alters the amino acid sequence and introduces a premature stop codon but does not appear harmful or disease-causing and is potentially unique to equine biology. A large portion of the RELN haplotype overlapped with an H3K27me3 modification mark, suggesting a regulatory role of this region. Despite the small sample size, the RELN haplotype's effects were validated for tölt, trot, and canter/gallop. Additionally, the RELN haplotype significantly influenced the age at which horses were presented for breeding field tests, indicating a potential role of the region in precocity and trainability. Functional experiments are needed to further investigate the regions' influences on biological processes and their potential impact on horse performance.
Collapse
Affiliation(s)
- Heiðrún Sigurðardóttir
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, Uppsala, SE-75007, Sweden.
- Faculty of Agricultural Sciences, Agricultural University of Iceland, Hvanneyri, Borgarbyggð, IS-311, Iceland.
| | - Susanne Eriksson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, Uppsala, SE-75007, Sweden
| | - Adnan Niazi
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, Uppsala, SE-75007, Sweden
| | - Marie Rhodin
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, Uppsala, SE-75007, Sweden
| | | | - Thorvaldur Kristjansson
- Faculty of Agricultural Sciences, Agricultural University of Iceland, Hvanneyri, Borgarbyggð, IS-311, Iceland
- The Icelandic Agricultural Advisory Centre, Höfðabakka 9, Reykjavik, IS-110, Iceland
| | - Gabriella Lindgren
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, Uppsala, SE-75007, Sweden
- Center for Animal Breeding and Genetics, Department of Biosystems, KU Leuven, Kasteelpark Arenberg 30, Leuven, BE-3001, Belgium
| |
Collapse
|
13
|
Hoffmann J, Schütze TM, Kolodziejczyk A, Küster K, Kränkel A, Reinhardt S, Derihaci RP, Birdir C, Wimberger P, Koseki H, Albert M. Canonical and non-canonical PRC1 differentially contribute to regulation of neural stem cell fate. Life Sci Alliance 2025; 8:e202403006. [PMID: 39933923 PMCID: PMC11814486 DOI: 10.26508/lsa.202403006] [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: 08/21/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Neocortex development is characterized by sequential phases of neural progenitor cell (NPC) expansion, neurogenesis, and gliogenesis. Polycomb-mediated epigenetic mechanisms are known to play important roles in regulating the lineage potential of NPCs during development. The composition of Polycomb repressive complex 1 (PRC1) is highly diverse in mammals and was hypothesized to contribute to context-specific regulation of cell fate. Here, we have performed a side-by-side comparison of the role of canonical PRC1.2/1.4 and non-canonical PRC1.3/1.5, all of which are expressed in the developing neocortex, in NSC proliferation and differentiation. We found that the deletion of Pcgf2/4 in NSCs led to a strong reduction in proliferation and to altered lineage fate, both during the neurogenic and gliogenic phase, whereas Pcgf3/5 played a minor role. Mechanistically, genes encoding stem cell and neurogenic factors were bound by PRC1 and differentially expressed upon Pcgf2/4 deletion. Thus, rather than different PRC1 subcomplexes contributing to different phases of neural development, we found that canonical PRC1 played a more significant role in NSC regulation during proliferative, neurogenic, and gliogenic phases compared with non-canonical PRC1.
Collapse
Affiliation(s)
- Janine Hoffmann
- Center for Regenerative Therapies Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Theresa M Schütze
- Center for Regenerative Therapies Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Annika Kolodziejczyk
- Center for Regenerative Therapies Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Karolin Küster
- Center for Regenerative Therapies Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Annekathrin Kränkel
- DRESDEN-Concept Genome Center, Center for Molecular and Cellular Bioengineering, Technology Platform of the TUD Dresden University of Technology, Dresden, Germany
| | - Susanne Reinhardt
- DRESDEN-Concept Genome Center, Center for Molecular and Cellular Bioengineering, Technology Platform of the TUD Dresden University of Technology, Dresden, Germany
| | - Razvan P Derihaci
- Department of Gynecology and Obstetrics, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases, Dresden, Germany
| | - Cahit Birdir
- Department of Gynecology and Obstetrics, Technische Universität Dresden, Dresden, Germany
- Center for Feto/Neonatal Health, Technische Universität Dresden, Dresden, Germany
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases, Dresden, Germany
| | - Haruhiko Koseki
- Laboratory of Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mareike Albert
- Center for Regenerative Therapies Dresden, TUD Dresden University of Technology, Dresden, Germany
| |
Collapse
|
14
|
Mätlik K, Govek EE, Hatten ME. Histone bivalency in CNS development. Genes Dev 2025; 39:428-444. [PMID: 39880657 PMCID: PMC11960699 DOI: 10.1101/gad.352306.124] [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] [Indexed: 01/31/2025]
Abstract
Neuronal maturation is guided by changes in the chromatin landscape that control developmental gene expression programs. Histone bivalency, the co-occurrence of activating and repressive histone modifications, has emerged as an epigenetic feature of developmentally regulated genes during neuronal maturation. Although initially associated with early embryonic development, recent studies have shown that histone bivalency also exists in differentiated and mature neurons. In this review, we discuss methods to study bivalency in specific populations of neurons and summarize emerging studies on the function of bivalency in central nervous system neuronal maturation and in adult neurons.
Collapse
Affiliation(s)
- Kärt Mätlik
- Laboratory of Developmental Neurobiology, The Rockefeller University, New York, New York 10065, USA;
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn 12618, Estonia
| | - Eve-Ellen Govek
- Laboratory of Developmental Neurobiology, The Rockefeller University, New York, New York 10065, USA
| | - Mary E Hatten
- Laboratory of Developmental Neurobiology, The Rockefeller University, New York, New York 10065, USA;
| |
Collapse
|
15
|
Marinov GK, Ramalingam V, Greenleaf WJ, Kundaje A. An updated compendium and reevaluation of the evidence for nuclear transcription factor occupancy over the mitochondrial genome. PLoS One 2025; 20:e0318796. [PMID: 40163815 PMCID: PMC11957562 DOI: 10.1371/journal.pone.0318796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/20/2025] [Indexed: 04/02/2025] Open
Abstract
In most eukaryotes, mitochondrial organelles contain their own genome, usually circular, which is the remnant of the genome of the ancestral bacterial endosymbiont that gave rise to modern mitochondria. Mitochondrial genomes are dramatically reduced in their gene content due to the process of endosymbiotic gene transfer to the nucleus; as a result most mitochondrial proteins are encoded in the nucleus and imported into mitochondria. This includes the components of the dedicated mitochondrial transcription and replication systems and regulatory factors, which are entirely distinct from the information processing systems in the nucleus. However, since the 1990s several nuclear transcription factors have been reported to act in mitochondria, and previously we identified 8 human and 3 mouse transcription factors (TFs) with strong localized enrichment over the mitochondrial genome using ChIP-seq (Chromatin Immunoprecipitation) datasets from the second phase of the ENCODE (Encyclopedia of DNA Elements) Project Consortium. Here, we analyze the greatly expanded in the intervening decade ENCODE compendium of TF ChIP-seq datasets (a total of 6,153 ChIP experiments for 942 proteins, of which 763 are sequence-specific TFs) combined with interpretative deep learning models of TF occupancy to create a comprehensive compendium of nuclear TFs that show evidence of association with the mitochondrial genome. We find some evidence for chrM occupancy for 50 nuclear TFs and two other proteins, with bZIP TFs emerging as most likely to be playing a role in mitochondria. However, we also observe that in cases where the same TF has been assayed with multiple antibodies and ChIP protocols, evidence for its chrM occupancy is not always reproducible. In the light of these findings, we discuss the evidential criteria for establishing chrM occupancy and reevaluate the overall compendium of putative mitochondrial-acting nuclear TFs.
Collapse
Affiliation(s)
- Georgi K Marinov
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | | | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, California, United States of America
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, California, United States of America
- Department of Applied Physics, Stanford University, Stanford, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| |
Collapse
|
16
|
Schröder CM, Zissel L, Mersiowsky SL, Tekman M, Probst S, Schüle KM, Preissl S, Schilling O, Timmers HTM, Arnold SJ. EOMES establishes mesoderm and endoderm differentiation potential through SWI/SNF-mediated global enhancer remodeling. Dev Cell 2025; 60:735-748.e5. [PMID: 39662466 DOI: 10.1016/j.devcel.2024.11.014] [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: 10/27/2023] [Revised: 09/17/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024]
Abstract
Mammalian pluripotent cells first segregate into neuroectoderm (NE), or mesoderm and endoderm (ME), characterized by lineage-specific transcriptional programs and chromatin states. To date, the relationship between transcription factor activities and dynamic chromatin changes that guide cell specification remains ill-defined. In this study, we employ mouse embryonic stem cell differentiation toward ME lineages to reveal crucial roles of the Tbx factor Eomes to globally establish ME enhancer accessibility as the prerequisite for ME lineage competence and ME-specific gene expression. EOMES cooperates with the SWItch/sucrose non-fermentable (SWI/SNF) complex to drive chromatin rewiring that is essential to overcome default NE differentiation, which is favored by asymmetries in chromatin accessibility at pluripotent state. Following global ME enhancer remodeling, ME-specific gene transcription is controlled by additional signals such as Wnt and transforming growth factor β (TGF-β)/NODAL, as a second layer of gene expression regulation, which can be mechanistically separated from initial chromatin remodeling activities.
Collapse
Affiliation(s)
- Chiara M Schröder
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Lea Zissel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Sophie-Luise Mersiowsky
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Mehmet Tekman
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Simone Probst
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Katrin M Schüle
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Sebastian Preissl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Medical Centre, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - H Th Marc Timmers
- Department of Urology, Medical Centre, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; German Cancer Consortium (DKTK), partner site Freiburg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
17
|
Peng B, Wang Q, Zhang F, Shen H, Du P. Mouse totipotent blastomere-like cells model embryogenesis from zygotic genome activation to post implantation. Cell Stem Cell 2025; 32:391-408.e11. [PMID: 39826539 DOI: 10.1016/j.stem.2024.12.006] [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: 09/15/2023] [Revised: 09/30/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025]
Abstract
Embryo development begins with zygotic genome activation (ZGA), eventually generating blastocysts for implantation. However, in vitro systems modeling the pre-implantation development are still absent and challenging. Here, we used mouse totipotent blastomere-like cells (TBLCs) to develop spontaneous differentiation and blastoid formation systems, respectively. We found Wnt signaling enabled the rapid expansion of TBLCs and the optimization of their culture medium. We successfully developed a TBLC-spontaneous differentiation system in which mouse TBLCs (mTBLCs) firstly converted into two types of ZGA-like cells (ZLCs) distinguished by Zscan4 expression. Surprisingly, Zscan4-, but not Zscan4+, ZLCs further passed through intermediate 4-cell and then 8-cell/morula stages to produce epiblast, primitive endoderm, and trophectoderm lineages. Significantly, single TBLCs underwent expansion, compaction, and polarization to efficiently generate blastocyst-like structures and even post-implantation egg-cylinder-like structures. Conclusively, we established TBLC-based differentiation and embryo-like structure formation systems to model early embryonic development, offering criteria for evaluating and understanding totipotency.
Collapse
Affiliation(s)
- Bing Peng
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Qingyi Wang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Feixiang Zhang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Hui Shen
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Peng Du
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing Advanced Center of RNA Biology, Peking University, Beijing 100871, China.
| |
Collapse
|
18
|
Kumar B, Navarro C, Yung PYK, Lyu J, Salazar Mantero A, Katsori AM, Schwämmle H, Martin M, Elsässer SJ. Multiplexed chromatin immunoprecipitation sequencing for quantitative study of histone modifications and chromatin factors. Nat Protoc 2025; 20:779-809. [PMID: 39363107 DOI: 10.1038/s41596-024-01058-z] [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: 09/29/2023] [Accepted: 07/29/2024] [Indexed: 10/05/2024]
Abstract
ChIP-seq is a widely used technique for studying histone post-translational modifications and DNA-binding proteins. DNA fragments associated with a specific protein or histone modification epitope are captured by using antibodies, sequenced and mapped to a reference genome. Albeit versatile and popular, performing many parallel ChIP-seq experiments to compare different conditions, replicates and epitopes is laborious, is prone to experimental variation and does not allow quantitative comparisons unless adequate spike-in chromatin is included. We present a detailed protocol for performing and analyzing a multiplexed quantitative chromatin immunoprecipitation-sequencing experiment (MINUTE-ChIP), in which multiple samples are profiled against multiple epitopes in a single workflow. Multiplexing not only dramatically increases the throughput of ChIP-seq experiments (e.g., profiling 12 samples against multiple histone modifications or DNA-binding proteins in a single experiment), but also enables accurate quantitative comparisons. The protocol consists of four parts: sample preparation (i.e., lysis, chromatin fragmentation and barcoding of native or formaldehyde-fixed material), pooling and splitting of the barcoded chromatin into parallel immunoprecipitation reactions, preparation of next-generation sequencing libraries from input and immunoprecipitated DNA and data analysis using our dedicated analysis pipeline. This pipeline autonomously generates quantitatively scaled ChIP-seq tracks for downstream analysis and visualization, alongside necessary quality control indicators. The entire workflow requires basic knowledge in molecular biology and bioinformatics and can be completed in 1 week. MINUTE-ChIP empowers biologists to perform every ChIP-seq experiment with an appropriate number of replicates and control conditions, delivering more statistically robust, exquisitely quantitative and biologically meaningful results.
Collapse
Affiliation(s)
- Banushree Kumar
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Solna, Sweden
- Epigenica AB, A Working Lab, Solna, Sweden
| | - Carmen Navarro
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Solna, Sweden
- Epigenica AB, A Working Lab, Solna, Sweden
| | - Philip Yuk Kwong Yung
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Solna, Sweden
| | - Jing Lyu
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Solna, Sweden
| | - Angelo Salazar Mantero
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Solna, Sweden
| | - Anna-Maria Katsori
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Solna, Sweden
| | - Hannah Schwämmle
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Solna, Sweden
| | - Marcel Martin
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Simon J Elsässer
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden.
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
19
|
Resar LMS, Luo LZ. High Mobility Group A1 Chromatin Keys: Unlocking the Genome During MPN Progression. Int J Mol Sci 2025; 26:2125. [PMID: 40076747 PMCID: PMC11899949 DOI: 10.3390/ijms26052125] [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: 11/07/2024] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Patients with chronic, indolent myeloproliferative neoplasms (MPNs) are at risk for transformation to highly lethal leukemia, although targetable mechanisms driving progression remain elusive. We discovered that the High Mobility Group A1 (HMGA1) gene is up-regulated with MPN progression in patients and required for evolution into myelofibrosis (MF) or acute myeloid leukemia (AML) in preclinical models. HMGA1 encodes the HMGA1 epigenetic regulators that modulate the chromatin state during embryogenesis and tissue regeneration. While HMGA1 is silenced in most differentiated cells, it becomes aberrantly re-expressed in JAK2 mutant (JAK2-V617F) MPN, with the highest levels after transformation to secondary MF or AML. Here, we review recent work highlighting HMGA1 function in MPN progression. Though underlying mechanisms continue to emerge, increasing evidence suggests that HMGA1 functions as a "chromatin key" required to "unlock" regions of the genome involved in clonal expansion and progression in MPN. Together, these findings illuminate HMGA1 as a driver of MPN progression and a promising therapeutic target.
Collapse
Affiliation(s)
- Linda M. S. Resar
- Departments of Medicine (Hematology), Oncology, Pathology and Institute for Cellular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | | |
Collapse
|
20
|
Schüle KM, Probst S. Epigenetic control of cell identities from epiblast to gastrulation. FEBS J 2025. [PMID: 39985220 DOI: 10.1111/febs.70024] [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: 08/22/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 02/24/2025]
Abstract
Epigenetic modifications of chromatin are essential for the establishment of cell identities during embryogenesis. Between embryonic days 3.5-7.5 of murine development, major cell lineage decisions are made that discriminate extraembryonic and embryonic tissues, and the embryonic primary germ layers are formed, thereby laying down the basic body plan. In this review, we cover the contribution of dynamic chromatin modifications by DNA methylation, changes of chromatin accessibility, and histone modifications, that in combination with transcription factors control gene expression programs of different cell types. We highlight the differences in regulation of enhancer and promoter marks and discuss their requirement in cell lineage specification. Importantly, in many cases, lineage-specific targeting of epigenetic modifiers is carried out by pioneer or master transcription factors, that in sum mediate the chromatin landscape and thereby control the transcription of cell-type-specific gene programs and thus, cell identities.
Collapse
Affiliation(s)
- Katrin M Schüle
- Faculty of Medicine, Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Germany
- Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Germany
| | - Simone Probst
- Faculty of Medicine, Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Germany
| |
Collapse
|
21
|
Sipola J, Munzur AD, Kwan EM, Seo CCY, Hauk BJ, Parekh K, Liao YJ(R, Bernales CQ, Donnellan G, Bloise I, Fung E, Ng SWS, Wang G, Vandekerkhove G, Nykter M, Annala M, Maurice-Dror C, Chi KN, Herberts C, Wyatt AW, Takeda DY. Plasma Cell-Free DNA Chromatin Immunoprecipitation Profiling Depicts Phenotypic and Clinical Heterogeneity in Advanced Prostate Cancer. Cancer Res 2025; 85:791-807. [PMID: 39652574 PMCID: PMC11832346 DOI: 10.1158/0008-5472.can-24-2052] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 12/04/2024] [Indexed: 02/18/2025]
Abstract
Cell phenotype underlies prostate cancer presentation and treatment resistance and can be regulated by epigenomic features. However, the osteotropic tendency of prostate cancer limits access to metastatic tissue, meaning most prior insights into prostate cancer chromatin biology are from preclinical models that do not fully represent disease complexity. Noninvasive chromatin immunoprecipitation of histones in plasma cell-free DNA (cfDNA) in humans may enable the capture of disparate prostate cancer phenotypes. In this study, we analyzed activating promoter- and enhancer-associated H3K4me2 from cfDNA in metastatic prostate cancer enriched for divergent patterns of metastasis and diverse clinical presentation. H3K4me2 density across prostate cancer genes, accessible chromatin, and lineage-defining transcription factor-binding sites correlated strongly with ctDNA fraction-demonstrating capture of prostate cancer-specific biology and informing the development of a statistical framework to adjust for ctDNA fraction. Chromatin hallmarks mirrored synchronously measured clinicogenomic features: bone- versus liver-predominant disease, serum PSA, biopsy-confirmed histopathologic subtype, and RB1 deletions convergently indicated phenotype segregation along an axis of differential androgen receptor activity and neuroendocrine identity. Detection of lineage switching after sequential progression on systemic therapy in select patients indicates potential use for individualized resistance monitoring. Epigenomic footprints of metastasis-induced normal tissue destruction were evident in bulk cfDNA from two patients. Finally, a public epigenomic resource was generated using a distinct chromatin marker that has not been widely investigated in prostate cancer. These results provide insights into the adaptive molecular landscape of aggressive prostate cancer and endorse plasma cfDNA chromatin profiling as a biomarker source and biological discovery tool. Significance: Plasma cell-free chromatin immunoprecipitation sequencing enables phenotypic dissection of lethal prostate cancer and is a practical tool for biomarker discovery while overcoming prior limitations of access to relevant tissue and reliance on model systems.
Collapse
Affiliation(s)
- Joonatan Sipola
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere, Finland
| | - Asli D. Munzur
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Edmond M. Kwan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medicine, School of Clinical Sciences; Monash University; Melbourne, Victoria, Australia
| | - Clara C. Y. Seo
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Benjamin J. Hauk
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Karan Parekh
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Yi Jou (Ruby) Liao
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Cecily Q. Bernales
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Gráinne Donnellan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Ingrid Bloise
- Instituto Brasileiro de Controle ao Cancer, Sao Paulo, Brazil
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Emily Fung
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Sarah W. S. Ng
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Gang Wang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Gillian Vandekerkhove
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere, Finland
| | - Matti Annala
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere, Finland
| | | | - Kim N. Chi
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Cameron Herberts
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Alexander W. Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - David Y. Takeda
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
22
|
Huffines AK, Schneider DA. Differential impact of divalent metals on native elongating transcript sequencing (NET-seq) protocols for RNA polymerases I and II. PLoS One 2025; 20:e0315595. [PMID: 39946358 PMCID: PMC11824990 DOI: 10.1371/journal.pone.0315595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/28/2024] [Indexed: 02/16/2025] Open
Abstract
Throughout all domains of life, RNA polymerases (Pols) synthesize RNA from DNA templates, a process called transcription. During transcription, Pols require divalent metal cations for nucleotide addition and cleavage of the nascent RNA after misincorporation or polymerase stalling. Recently, several next-generation sequencing techniques have emerged to study transcription at single-nucleotide resolution in vivo. One such technique, native elongating transcript sequencing (NET-seq), allows for isolation of transcription elongation complexes associated with a specific Pol, defining polymerase occupancy on the DNA template. Originally developed to study RNA polymerase II (Pol II), NET-seq has been adapted for RNA polymerase I (Pol I) and bacterial RNA polymerase. We recently optimized Pol I NET-seq in Saccharomyces cerevisiae, however, we omitted nucleases and their metal cofactors, which are commonly used in Pol II NET-seq. Here, we investigated the effect of CaCl2 ± MNase and MnCl2 ± DNase I on Pol I occupancy. We found that exposure of Pol I to CaCl2 and MnCl2 during NET-seq caused a significant reduction in immunoprecipitation of nascent rRNA compared to the untreated control samples, with a more severe effect when incubated with MnCl2 vs. CaCl2. Surprisingly, in contrast to the Pol I results, we found that metal treatment during Pol II NET-seq did not have a significant effect on nascent transcript capture. Taken together, these observations reinforce the conclusion that transcription elongation complexes formed by Pols I and II have unique characteristics and emphasize the need to carefully consider experimental conditions deployed in all stages of nucleic acid library generation.
Collapse
Affiliation(s)
- Abigail K. Huffines
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - David A. Schneider
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, United States of America
| |
Collapse
|
23
|
DeSisto J, Balakrishnan I, Knox AJ, Link G, Venkataraman S, Vibhakar R, Green AL. PRMT5 Maintains Tumor Stem Cells to Promote Pediatric High-Grade Glioma Tumorigenesis. Mol Cancer Res 2025; 23:107-118. [PMID: 39422546 PMCID: PMC11799838 DOI: 10.1158/1541-7786.mcr-24-0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/02/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024]
Abstract
Pediatric high-grade gliomas (PHGG) are aggressive, undifferentiated central nervous system tumors with poor outcomes, for which no standard-of-care drug therapy currently exists. Through a knockdown (KD) screen for epigenetic regulators, we identified PRMT5 as essential for PHGG cell growth. We hypothesized that, similar to its effect in normal cells, PRMT5 promotes self-renewal of stem-like PHGG tumor-initiating cells essential for tumor growth. We conducted in vitro analyses, including limiting dilution studies of self-renewal, to determine the phenotypic effects of PRMT5 KD. We performed chromatin immunoprecipitation sequencing (ChIP-Seq) to identify PRMT5-mediated epigenetic changes and performed gene set enrichment analysis to identify pathways that PRMT5 regulates. Using an orthotopic xenograft model of PHGG, we tracked survival and histologic characteristics resulting from PRMT5 KD or administration of a PRMT5 inhibitor ± radiation therapy. In vitro, PRMT5 KD slowed cell-cycle progression, tumor growth and self-renewal, and altered chromatin occupancy at genes associated with differentiation, tumor formation, and growth. In vivo, PRMT5 KD increased survival and reduced tumor aggressiveness; however, pharmacologic inhibition of PRMT5 with or without radiation therapy did not improve survival. PRMT5 KD epigenetically reduced tumor-initiating cells' self-renewal, leading to increased survival in preclinical models. Pharmacologic inhibition of PRMT5 enzymatic activity may have failed in vivo due to insufficient reduction of PRMT5 activity by chemical inhibition, or this failure may suggest that nonenzymatic activities of PRMT5 are more relevant. Implications: PRMT5 maintains and promotes the growth of stem-like cells that initiate and drive tumorigenesis in PHGG.
Collapse
Affiliation(s)
- John DeSisto
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ilango Balakrishnan
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Aaron J. Knox
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Gabrielle Link
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sujatha Venkataraman
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Rajeev Vibhakar
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, Colorado
| | - Adam L. Green
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, Colorado
| |
Collapse
|
24
|
Bryan E, Valsakumar D, Idigo NJ, Warburton M, Webb KM, McLaughlin KA, Spanos C, Lenci S, Major V, Ambrosi C, Andrews S, Baubec T, Rappsilber J, Voigt P. Nucleosomal asymmetry shapes histone mark binding and promotes poising at bivalent domains. Mol Cell 2025; 85:471-489.e12. [PMID: 39731917 DOI: 10.1016/j.molcel.2024.12.002] [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: 05/24/2023] [Revised: 08/16/2024] [Accepted: 12/03/2024] [Indexed: 12/30/2024]
Abstract
Promoters of developmental genes in embryonic stem cells (ESCs) are marked by histone H3 lysine 4 trimethylation (H3K4me3) and H3K27me3 in an asymmetric nucleosomal conformation, with each sister histone H3 carrying only one of the two marks. These bivalent domains are thought to poise genes for timely activation upon differentiation. Here, we show that asymmetric bivalent nucleosomes recruit repressive H3K27me3 binders but fail to enrich activating H3K4me3 binders, thereby promoting a poised state. Strikingly, the bivalent mark combination further promotes recruitment of specific chromatin proteins that are not recruited by each mark individually, including the lysine acetyltransferase (KAT) complex KAT6B. Knockout of KAT6B blocks neuronal differentiation, demonstrating that KAT6B is critical for proper bivalent gene expression during ESC differentiation. These findings reveal how readout of the bivalent histone marks directly promotes a poised state at developmental genes while highlighting how nucleosomal asymmetry is critical for histone mark readout and function.
Collapse
Affiliation(s)
- Elana Bryan
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Devisree Valsakumar
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK; Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK
| | - Nwamaka J Idigo
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Marie Warburton
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Kimberly M Webb
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Katy A McLaughlin
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Christos Spanos
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Simone Lenci
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Viktoria Major
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Christina Ambrosi
- Department of Molecular Mechanism of Disease, University of Zurich, 8057 Zurich, Switzerland
| | - Simon Andrews
- Bioinformatics Group, Babraham Institute, Cambridge CB22 3AT, UK
| | - Tuncay Baubec
- Department of Molecular Mechanism of Disease, University of Zurich, 8057 Zurich, Switzerland; Genome Biology and Epigenetics, Institute of Biodynamics and Biocomplexity, Department of Biology, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Juri Rappsilber
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK; Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany
| | - Philipp Voigt
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK; Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK.
| |
Collapse
|
25
|
Wan L, Zhang H, Liu J, He Q, Zhao J, Pan C, Zheng K, Tang Y. Lactylation and human disease. Expert Rev Mol Med 2025; 27:e10. [PMID: 39895568 PMCID: PMC11879378 DOI: 10.1017/erm.2025.3] [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: 06/28/2024] [Revised: 12/17/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Lactylation, a new epigenetic modification, is an important way in which lactate exerts physiological functions. There is a close relationship between increased lactylations caused by lactate and glycolysis, which can interact and play a role in disease through lactate as an intermediate mediator. Current research on lactylations has focused on histone lactylation, but non-histone lactylation also has greater research potential. Due to the ubiquity of lactate modifications in mammalian cells, an increasing number of studies have found that lactate modifications play important roles in tumour cell metabolism, gene transcription and immunity. METHODS A systematic literature search was carried out using search key terms and synonyms. Full-paper screening was performed based on specific inclusion and exclusion criteria. RESULTS Many literatures have reported that the lactylation of protein plays an important role in human diseases and is involved in the occurrence and development of human diseases. CONCLUSIONS This article summary the correlation between lactylation and glycolysis, histones and non-histone proteins; the relationship between lactonation modifications and tumour development; and the current existence of lactylation-related inhibitors, with a view to provide new basic research ideas and clinical therapeutic tools for lactylation-related diseases.
Collapse
Affiliation(s)
- Linlin Wan
- Department of Pathology, Suzhou Ninth Hospital Affiliated to Soochow University, Jiangsu, China
| | - HuiJuan Zhang
- Institute of Biomedical Engineering, Kunming medical university, Kunming, China
| | - Jialing Liu
- Department of Pathology, Suzhou Ninth Hospital Affiliated to Soochow University, Jiangsu, China
| | - Qian He
- Institute of Biomedical Engineering, Kunming medical university, Kunming, China
| | - Jiumei Zhao
- Laboratory medicine department, Chongqing Nanchuan District People’s Hospital, Chongqing, China
| | - Chenglong Pan
- Institute of Biomedical Engineering, Kunming medical university, Kunming, China
| | - Kepu Zheng
- Institute of Biomedical Engineering, Kunming medical university, Kunming, China
| | - Yu Tang
- Institute of Biomedical Engineering, Kunming medical university, Kunming, China
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
26
|
Richard Albert J, Urli T, Monteagudo-Sánchez A, Le Breton A, Sultanova A, David A, Scarpa M, Schulz M, Greenberg MVC. DNA methylation shapes the Polycomb landscape during the exit from naive pluripotency. Nat Struct Mol Biol 2025; 32:346-357. [PMID: 39448850 DOI: 10.1038/s41594-024-01405-4] [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: 09/26/2023] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
In mammals, 5-methylcytosine (5mC) and Polycomb repressive complex 2 (PRC2)-deposited histone 3 lysine 27 trimethylation (H3K27me3) are generally mutually exclusive at CpG-rich regions. As mouse embryonic stem cells exit the naive pluripotent state, there is massive gain of 5mC concomitantly with restriction of broad H3K27me3 to 5mC-free, CpG-rich regions. To formally assess how 5mC shapes the H3K27me3 landscape, we profiled the epigenome of naive and differentiated cells in the presence and absence of the DNA methylation machinery. Surprisingly, we found that 5mC accumulation is not required to restrict most H3K27me3 domains. Instead, this 5mC-independent H3K27me3 restriction is mediated by aberrant expression of the PRC2 antagonist Ezhip (encoding EZH inhibitory protein). At the subset of regions where 5mC appears to genuinely supplant H3K27me3, we identified 163 candidate genes that appeared to require 5mC deposition and/or H3K27me3 depletion for their activation in differentiated cells. Using site-directed epigenome editing to directly modulate 5mC levels, we demonstrated that 5mC deposition is sufficient to antagonize H3K27me3 deposition and confer gene activation at individual candidates. Altogether, we systematically measured the antagonistic interplay between 5mC and H3K27me3 in a system that recapitulates early embryonic dynamics. Our results suggest that H3K27me3 restraint depends on 5mC, both directly and indirectly. Our study also implies a noncanonical role of 5mC in gene activation, which may be important not only for normal development but also for cancer progression, as oncogenic cells frequently exhibit dynamic replacement of 5mC for H3K27me3 and vice versa.
Collapse
Affiliation(s)
| | - Teresa Urli
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Ana Monteagudo-Sánchez
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain
| | - Anna Le Breton
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Amina Sultanova
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
- Development and Disease Research Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Angélique David
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | | | - Mathieu Schulz
- Institut Curie, PSL Research University, INSERM U934, CNRS, UMR3215, Paris, France
- Department of Pathology and Cell Biology, Faculty of Medicine, University of Montreal Hospital Research Centre, Montréal, Québec, Canada
| | | |
Collapse
|
27
|
Katikaneni A, Lowe CB. Novelty versus innovation of gene regulatory elements in human evolution and disease. Curr Opin Genet Dev 2025; 90:102279. [PMID: 39591813 PMCID: PMC11769741 DOI: 10.1016/j.gde.2024.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024]
Abstract
It is not currently understood how much of human evolution is due to modifying existing functional elements in the genome versus forging novel elements from nonfunctional DNA. Many early experiments that aimed to assign genetic changes on the human lineage to their resulting phenotypic change have focused on mutations that modify existing elements. However, a number of recent studies have highlighted the potential ease and importance of forging novel gene regulatory elements from nonfunctional sequences on the human lineage. In this review, we distinguish gene regulatory element novelty from innovation. We propose definitions for these terms and emphasize their importance in studying the genetic basis of human uniqueness. We discuss why the forging of novel regulatory elements may have been less emphasized during the previous decades, and why novel regulatory elements are likely to play a significant role in both human adaptation and disease.
Collapse
Affiliation(s)
- Anushka Katikaneni
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; University Program in Genetics and Genomics, Duke University, Durham, NC 27708, USA
| | - Craig B Lowe
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; University Program in Genetics and Genomics, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
28
|
Zhang J, Li X, Zhao Q, Ji J, Cui H, Hou W, Wang X, Song E, Xiao S, Ling S, Gao S, Liu X, Wen D, Kong Q. Acetylation at lysine 27 on maternal H3.3 regulates minor zygotic genome activation. Cell Rep 2025; 44:115148. [PMID: 39932187 PMCID: PMC11892348 DOI: 10.1016/j.celrep.2024.115148] [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: 05/08/2024] [Revised: 09/23/2024] [Accepted: 12/12/2024] [Indexed: 03/12/2025] Open
Abstract
Zygotic genome activation (ZGA) initiates transcription in early embryogenesis and requires extensive chromatin remodeling, including rapid incorporation of the histone variant H3.3. The distinct sources of H3.3 from paternal and maternal alleles (paH3.3 and maH3.3) complicate tracking their individual contributions. Here, using an H3.3B-hemagglutinin (HA)-tagged mouse model, we profile the temporal dynamics of paH3.3 and maH3.3, revealing a unique pattern of maH3.3 enrichment at the promoter regions from zygotes to 2-cell embryos, highlighting the crucial role of maternally stored H3.3 mRNAs and proteins (mH3.3) in pre-implantation development. Knockdown of mH3.3 compromises cleavage and minor ZGA. Mechanistically, mH3.3 facilitates minor ZGA through H3.3S31ph-dependent H3K27ac deposition. Profiling of H3.3 landscape in parthenogenetic (PG) and androgenetic (AG) embryos highlights the role of mH3.3 in remodeling the paternal genome by establishing H3K27ac. These findings demonstrate that mH3.3-mediated parental chromatin reprogramming is essential for orchestrating minor ZGA.
Collapse
Affiliation(s)
- Jiaming Zhang
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Xuanwen Li
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Zhao
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingzhang Ji
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongdi Cui
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weibo Hou
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyu Wang
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Entong Song
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songling Xiao
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shukuan Ling
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shaorong Gao
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Xiaoyu Liu
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Duancheng Wen
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Qingran Kong
- Oujiang Laboratory, Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
29
|
Carrothers S, Trevisan R, Jayasundara N, Pelletier N, Weeks E, Meyer JN, Di Giulio R, Weinhouse C. An epigenetic memory at the CYP1A gene in cancer-resistant, pollution-adapted killifish. Sci Rep 2025; 15:3033. [PMID: 39856074 PMCID: PMC11759692 DOI: 10.1038/s41598-024-82740-w] [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: 08/28/2024] [Accepted: 12/09/2024] [Indexed: 01/27/2025] Open
Abstract
Human exposure to polycyclic aromatic hydrocarbons (PAH) is a significant public health problem that will worsen with a warming climate and increased large-scale wildfires. Here, we characterize an epigenetic memory at the cytochrome P450 1 A (CYP1A) gene in wild Fundulus heteroclitus that have adapted to chronic, extreme PAH pollution. In wild-type fish, CYP1A is highly induced by PAH. In PAH-tolerant fish, CYP1A induction is blunted. Since CYP1A metabolically activates PAH, this memory protects these fish from PAH-mediated cancer. However, PAH-tolerant fish reared in clean water recover CYP1A inducibility, indicating a non-genetic effect. We observed epigenetic control of this reversible memory of generational PAH stress in F1 PAH-tolerant embryos. We detected a bivalent domain in the CYP1A promoter enhancer comprising both activating and repressive histone post-translational modifications. Activating modifications, relative to repressive ones, showed greater increases in response to PAH in sensitive embryos, relative to tolerant, consistent with greater gene activation. PAH-tolerant adult fish showed persistent induction of CYP1A long after exposure cessation, which is consistent with defective CYP1A shutoff. These results indicate that PAH-tolerant fish have epigenetic protection against PAH-induced cancer in early life that degrades in response to continuous gene activation.
Collapse
Affiliation(s)
- Samantha Carrothers
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, 97239, Portland, OR, USA
| | - Rafael Trevisan
- Nicholas School of the Environment, Duke University, 27701, Durham, NC, USA
- Univ Brest, Ifremer, CNRS, IRD, UMR 6539, LEMAR, Plouzané, 29280, France
| | - Nishad Jayasundara
- Nicholas School of the Environment, Duke University, 27701, Durham, NC, USA
| | - Nicole Pelletier
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, 97239, Portland, OR, USA
| | - Emma Weeks
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, 97239, Portland, OR, USA
| | - Joel N Meyer
- Nicholas School of the Environment, Duke University, 27701, Durham, NC, USA
| | - Richard Di Giulio
- Nicholas School of the Environment, Duke University, 27701, Durham, NC, USA
| | - Caren Weinhouse
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, 97239, Portland, OR, USA.
| |
Collapse
|
30
|
Luo Y, Zhai H, Zhong X, Yang B, Xu Y, Liu T, Wang Q, Zhou Y, Mao Y, Liu Y, Tang Q, Lu Y, Wang Y, Xu J. Characterization and functional analysis of conserved non-coding sequences among poaceae: insights into gene regulation and phenotypic variation in maize. BMC Genomics 2025; 26:46. [PMID: 39833673 PMCID: PMC11745007 DOI: 10.1186/s12864-025-11221-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Conserved non-coding sequences (CNS) are islands of non-coding sequences conserved across species and play an important role in regulating the spatiotemporal expression of genes. Identification of CNS provides valuable information about potentially functional genomic elements, regulatory regions, and helps to gain insights into the genetic basis of crop agronomic traits. RESULTS Here, we comprehensively analyze CNS in maize, by comparing the genomes of maize inbred line B73 (Zea mays ssp. mays), its close wild relative Zea mays spp. mexicana, and other grasses in Poaceae, including sorghum (Sorghum bicolor), foxtail millet (Setaria italica) and two adlay (Coix lacryma) cultivars. There were 289,931 CNS found in two syntenic gene pairs, while 51,701 CNS were conserved within at least three species. To explore the regulatory characteristics of the CNS identified, the flanking regions of CNS were compared with the peaks called using both transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and chromatin immunoprecipitation with high-throughput sequencing (ChIP-Seq) data of histone modifications. It was found that CNS in maize were enriched in open chromatin regions compared with randomly selected non-coding regions of similar length. A significant enrichment of transcription factor binding sites was found within CNS sequences, including different transcription factors involved in abiotic stress response, such as OBP (OBF-BINDING PROTEIN) family and Adof1 (Encodes dof zinc finger protein). To investigate the epigenetic modification patterns in CNS, ChIP-Seq data for histone modifications H3K9ac, H3K4me3, H3K36me3, H3K9me3, and H3K27ac were further analyzed to depict the changes along CNS. Our findings revealed significantly elevated levels of transcription-promoting histone modifications in the CNS regions compared to randomly selected non-coding sequences with an equal number and similar length. Notably, CNS were also identified on both Vgt1 (Vegetative to generative transition 1) and ZmCCT10. In addition, CNS with potential functions were identified based on SNPs within CNS significantly associated with various agronomic traits in maize, which holds potential utility in molecular breeding for maize. CONCLUSIONS In summary, we identified and characterized CNS in maize through genomic comparative analysis, which provides valuable insights into their potential regulatory effects on gene expression and phenotypic variation.
Collapse
Affiliation(s)
- Yi Luo
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Hang Zhai
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Xiu Zhong
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Bo Yang
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Yang Xu
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Tianhong Liu
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- Sichuan Tianfu New Area Rural Revitalization Research Institute, Tianfu New Area, 610213, China
| | - Qi Wang
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Yang Zhou
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Yan Mao
- College of Chemistry and Life Sciences, Chengdu Normal University, Wenjiang, 611130, Sichuan, China
| | - Yaxi Liu
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- Triticeae Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Qi Tang
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Yanli Lu
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Yao Wang
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China
| | - Jie Xu
- Maize Research Institute, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China.
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Wenjiang, 611130, Sichuan, China.
| |
Collapse
|
31
|
Liang Y, Liu M, Liu B, Ziman B, Peng G, Mao Q, Wang X, Jiang L, Lin DC, Zheng Y. Comprehensive analysis of H3K27me3 LOCKs under different DNA methylation contexts reveal epigenetic redistribution in tumorigenesis. Epigenetics Chromatin 2025; 18:6. [PMID: 39833880 PMCID: PMC11748335 DOI: 10.1186/s13072-025-00570-0] [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: 10/24/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Histone modification H3K27me3 plays a critical role in normal development and is associated with various diseases, including cancer. This modification forms large chromatin domains, known as Large Organized Chromatin Lysine Domains (LOCKs), which span several hundred kilobases. RESULT In this study, we identify and categorize H3K27me3 LOCKs in 109 normal human samples, distinguishing between long and short LOCKs. Our findings reveal that long LOCKs are predominantly associated with developmental processes, while short LOCKs are enriched in poised promoters and are most associated with low gene expression. Further analysis of LOCKs in different DNA methylation contexts shows that long LOCKs are primarily located in partially methylated domains (PMDs), particularly in short-PMDs, where they are most likely responsible for the low expressions of oncogenes. We observe that in cancer cell lines, including those from esophageal and breast cancer, long LOCKs shift from short-PMDs to intermediate-PMDs and long-PMDs. Notably, a significant subset of tumor-associated long LOCKs in intermediate- and long-PMDs exhibit reduced H3K9me3 levels, suggesting that H3K27me3 compensates for the loss of H3K9me3 in tumors. Additionally, we find that genes upregulated in tumors following the loss of short LOCKs are typically poised promoter genes in normal cells, and their transcription is regulated by the ETS1 transcription factor. CONCLUSION These results provide new insights into the role of H3K27me3 LOCKs in cancer and underscore their potential impact on epigenetic regulation and disease mechanisms.
Collapse
Affiliation(s)
- Yuan Liang
- Clinical Big Data Research Center, Scientific Research Center, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People's Republic of China
| | - Mengni Liu
- Clinical Big Data Research Center, Scientific Research Center, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People's Republic of China
| | - Bingyuan Liu
- Clinical Big Data Research Center, Scientific Research Center, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People's Republic of China
| | - Benjamin Ziman
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, 2250 Alcazar Street - CSA 207D, Los Angeles, CA, 90033, USA
| | - Guanjie Peng
- Clinical Big Data Research Center, Scientific Research Center, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People's Republic of China
| | - Qiong Mao
- Clinical Big Data Research Center, Scientific Research Center, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People's Republic of China
| | - Xingzhe Wang
- Clinical Big Data Research Center, Scientific Research Center, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People's Republic of China
| | - Lizhen Jiang
- Clinical Big Data Research Center, Scientific Research Center, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People's Republic of China
| | - De-Chen Lin
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, 2250 Alcazar Street - CSA 207D, Los Angeles, CA, 90033, USA
| | - Yueyuan Zheng
- Clinical Big Data Research Center, Scientific Research Center, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, People's Republic of China.
| |
Collapse
|
32
|
De Paepe B, De Mey M. Biological Switches: Past and Future Milestones of Transcription Factor-Based Biosensors. ACS Synth Biol 2025; 14:72-86. [PMID: 39709556 PMCID: PMC11745168 DOI: 10.1021/acssynbio.4c00689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024]
Abstract
Since the description of the lac operon in 1961 by Jacob and Monod, transcriptional regulation in prokaryotes has been studied extensively and has led to the development of transcription factor-based biosensors. Due to the broad variety of detectable small molecules and their various applications across biotechnology, biosensor research and development have increased exponentially over the past decades. Throughout this period, key milestones in fundamental knowledge, synthetic biology, analytical tools, and computational learning have led to an immense expansion of the biosensor repertoire and its application portfolio. Over the years, biosensor engineering became a more multidisciplinary discipline, combining high-throughput analytical tools, DNA randomization strategies, forward engineering, and advanced protein engineering workflows. Despite these advances, many obstacles remain to fully unlock the potential of biosensor technology. This review analyzes the timeline of key milestones on fundamental research (1960s to 2000s) and engineering strategies (2000s onward), on both the DNA and protein level of biosensors. Moreover, insights into the future perspectives, remaining hurdles, and unexplored opportunities of this promising field are discussed.
Collapse
Affiliation(s)
- Brecht De Paepe
- Centre
for Synthetic Biology, Ghent University, Ghent 9000, Belgium
| | - Marjan De Mey
- Centre
for Synthetic Biology, Ghent University, Ghent 9000, Belgium
| |
Collapse
|
33
|
Kim MS, Lee R, Lee DH, Song H, Ha T, Kim JK, Kang BY, Agger K, Helin K, Shin D, Kang Y, Park C. ETV2/ER71 regulates hematovascular lineage generation and vascularization through an H3K9 demethylase, KDM4A. iScience 2025; 28:111538. [PMID: 39811655 PMCID: PMC11732216 DOI: 10.1016/j.isci.2024.111538] [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: 01/11/2023] [Revised: 12/15/2023] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
ETV2/ER71, an ETS (E-twenty six) transcription factor, is critical for hematopoiesis and vascular development. However, research about the molecular mechanisms behind ETV2-mediated gene transcription is limited. Herein, we demonstrate that ETV2 and KDM4A, an H3K9 demethylase, regulate hematopoietic and endothelial genes. Etv2 -/- mouse embryonic stem cells (mESCs), which fail to generate hematopoietic and endothelial cells, exhibit enhanced H3K9me3 levels in hematopoietic and endothelial genes. ETV2 interacts with KDM4A, and the ETV2-mediated transcriptional activation of hematopoietic and endothelial genes depends on KDM4A histone demethylase activity. The ETV2 and KDM4A complex binds to the transcription regulatory regions of genes directly regulated by ETV2. Mice lacking Kdm4a and Etv2 in endothelial cells (Cdh5Cre:Kdm:Etv2 f/f mice) display a more severe perfusion recovery and neovascularization defect, compared with Cdh5Cre:Kdm4a f/f mice, Cdh5Cre:Etv2 f/f mice, and controls. Collectively, we demonstrate that ETV2 interacts with KDM4A, and that this interaction is critical for hematovascular lineage generation and vascular regeneration.
Collapse
Affiliation(s)
- Min Seong Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Raham Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Dong Hun Lee
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biological Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Heesang Song
- Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - Taekyung Ha
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Joo Kyung Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Bum-Yong Kang
- Department of Medicine, Emory University School of Medicine, and Atlanta VA HCS, Atlanta, GA, USA
| | - Karl Agger
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Kristian Helin
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Donghyun Shin
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Yunhee Kang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Changwon Park
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| |
Collapse
|
34
|
Segert JA, Bulyk ML. Histone H4 lysine 20 monomethylation is not a mark of transcriptional silencers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.632211. [PMID: 39868205 PMCID: PMC11761030 DOI: 10.1101/2025.01.09.632211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Transcriptional silencers are cis-regulatory elements that downregulate the expression of target genes. Although thousands of silencers have been identified experimentally, a predictive chromatin signature of silencers has not been found. H4K20me1 previously was reported to be highly enriched among human silencers, but our reanalysis of those data using an appropriate background revealed that the enrichment is only marginal. We generated H4K20me1 ChIP-seq profiles in Drosophila S2 cells, which similarly showed that H4K20me1 does not mark Drosophila silencers and instead is associated with active transcription. Silencers remain a poorly annotated, difficult to predict class of cis-regulatory elements whose specific chromatin features remain to be identified.
Collapse
Affiliation(s)
- Julian A Segert
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Biological and Biomedical Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Martha L Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Biological and Biomedical Sciences, Harvard University, Cambridge, MA 02138, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
35
|
Mariner BL, McCoy BM, Greenier A, Brassington L, Slikas E, Adjangba C, Marye A, Harrison BR, Bamberger T, Algavi Y, Muller E, Harris A, Rout E, Avery A, Borenstein E, Promislow D, Snyder-Mackler N. DNA methylation of transposons pattern aging differences across a diverse cohort of dogs from the Dog Aging Project. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.08.617286. [PMID: 39416178 PMCID: PMC11482827 DOI: 10.1101/2024.10.08.617286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Within a species, larger individuals often have shorter lives and higher rates of age-related disease. Despite this well-known link, we still know little about underlying age-related epigenetic differences, which could help us better understand inter-individual variation in aging and the etiology, onset, and progression of age-associated disease. Dogs exhibit this negative correlation between size, health, and longevity and thus represent an excellent system in which to test the underlying mechanisms. Here, we quantified genome-wide DNA methylation in a cohort of 864 dogs in the Dog Aging Project. Age strongly patterned the dog epigenome, with the majority (66% of age-associated loci) of regions associating age-related loss of methylation. These age effects were non-randomly distributed in the genome and differed depending on genomic context. We found the LINE1 (long interspersed elements) class of TEs (transposable elements) were the most frequently hypomethylated with age (FDR < 0.05, 40% of all LINE1 regions). This LINE1 pattern differed in magnitude across breeds of different sizes- the largest dogs lost 0.26% more LINE1 methylation per year than the smallest dogs. This suggests that epigenetic regulation of TEs, particularly LINE1s, may contribute to accelerated age and disease phenotypes within a species. Since our study focused on the methylome of immune cells, we looked at LINE1 methylation changes in golden retrievers, a breed highly susceptible to hematopoietic cancers, and found they have accelerated age-related LINE1 hypomethylation compared to other breeds. We also found many of the LINE1s hypomethylated with age are located on the X chromosome and are, when considering X chromosome inactivation, counter-intuitively more methylated in males. These results have revealed the demethylation of LINE1 transposons as a potential driver of intra-species, demographic-dependent aging variation.
Collapse
|
36
|
Pampari A, Shcherbina A, Kvon EZ, Kosicki M, Nair S, Kundu S, Kathiria AS, Risca VI, Kuningas K, Alasoo K, Greenleaf WJ, Pennacchio LA, Kundaje A. ChromBPNet: bias factorized, base-resolution deep learning models of chromatin accessibility reveal cis-regulatory sequence syntax, transcription factor footprints and regulatory variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.25.630221. [PMID: 39829783 PMCID: PMC11741299 DOI: 10.1101/2024.12.25.630221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Despite extensive mapping of cis-regulatory elements (cREs) across cellular contexts with chromatin accessibility assays, the sequence syntax and genetic variants that regulate transcription factor (TF) binding and chromatin accessibility at context-specific cREs remain elusive. We introduce ChromBPNet, a deep learning DNA sequence model of base-resolution accessibility profiles that detects, learns and deconvolves assay-specific enzyme biases from regulatory sequence determinants of accessibility, enabling robust discovery of compact TF motif lexicons, cooperative motif syntax and precision footprints across assays and sequencing depths. Extensive benchmarks show that ChromBPNet, despite its lightweight design, is competitive with much larger contemporary models at predicting variant effects on chromatin accessibility, pioneer TF binding and reporter activity across assays, cell contexts and ancestry, while providing interpretation of disrupted regulatory syntax. ChromBPNet also helps prioritize and interpret regulatory variants that influence complex traits and rare diseases, thereby providing a powerful lens to decode regulatory DNA and genetic variation.
Collapse
Affiliation(s)
- Anusri Pampari
- Department of Computer Science, Stanford University, Stanford CA, 94305
| | - Anna Shcherbina
- Department of Biomedical Data Sciences, Stanford University, Stanford CA, 94305
| | - Evgeny Z. Kvon
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Michael Kosicki
- Environmental Genomics & System Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Surag Nair
- Department of Computer Science, Stanford University, Stanford CA, 94305
| | - Soumya Kundu
- Department of Computer Science, Stanford University, Stanford CA, 94305
| | | | | | | | - Kaur Alasoo
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - William James Greenleaf
- Department of Genetics, Stanford University, Stanford CA, 94305
- Department of Applied Physics, Stanford University, Stanford, California 94305, USA
| | - Len A. Pennacchio
- Environmental Genomics & System Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford CA, 94305
- Department of Genetics, Stanford University, Stanford CA, 94305
| |
Collapse
|
37
|
Liu Y, Liu Y, Zhu Y, Hu D, Nie H, Xie Y, Sun R, He J, Zhang H, Lu F. KDM2A and KDM2B protect a subset of CpG islands from DNA methylation. J Genet Genomics 2025; 52:39-50. [PMID: 39522683 DOI: 10.1016/j.jgg.2024.10.012] [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: 07/11/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
In the mammalian genome, most CpGs are methylated. However, CpGs within the CpG islands (CGIs) are largely unmethylated, which are important for gene expression regulation. The mechanism underlying the low methylation levels at CGIs remains largely elusive. KDM2 proteins (KDM2A and KDM2B) are H3K36me2 demethylases known to bind specifically at CGIs. Here, we report that depletion of each or both KDM2 proteins, or mutation of all their JmjC domains that harbor the H3K36me2 demethylation activity, leads to an increase in DNA methylation at selective CGIs. The Kdm2a/2b double knockout shows a stronger increase in DNA methylation compared with the single mutant of Kdm2a or Kdm2b, indicating that KDM2A and KDM2B redundantly regulate DNA methylation at CGIs. In addition, the increase of CGI DNA methylation upon mutations of KDM2 proteins is associated with the chromatin environment. Our findings reveal that KDM2A and KDM2B function redundantly in regulating DNA methylation at a subset of CGIs in an H3K36me2 demethylation-dependent manner.
Collapse
Affiliation(s)
- Yuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunji Zhu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Hu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hu Nie
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yali Xie
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rongrong Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin He
- Department of Biochemistry & Molecular Biology, College of Natural Science, Michigan State University, East Lansing, MI 48824, USA
| | - Honglian Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Falong Lu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
38
|
Martínez VL, Fraga MF. Chromatin Preparation from Frozen Tissues for Chromatin Immunoprecipitation (ChIP) Assays. Methods Mol Biol 2025; 2930:203-217. [PMID: 40402456 DOI: 10.1007/978-1-0716-4558-1_14] [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] [Indexed: 05/23/2025]
Abstract
Chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-seq) is a crucial method for examining transcription factor binding and histone modifications across the entire genome. This is a key step in deciphering the complex mechanisms that control cancer immunosurveillance. Here, we provide a comprehensive protocol covering all the steps needed to obtain DNA for ChIP-seq library preparation, starting from tissue dissection, followed by fixation, chromatin preparation, immunoprecipitation, and finally DNA purification. The protocol is optimized for frozen mice tissues, but can be easily adapted for use with any model organism. The resulting immunoprecipitated chromatin is suitable for library preparation and sequencing on an Illumina platform.
Collapse
Affiliation(s)
- Virginia López Martínez
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Mario Fernández Fraga
- Cancer Epigenetics and Nanomedicine Laboratory, Centro de Investigación en Nanomateriales y Nanotecnología-Consejo Superior de Investigaciones Científicas (CINN-CSIC), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA-FINBA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain.
| |
Collapse
|
39
|
Hölzenspies JJ, Sengupta D, Bickmore WA, Brickman JM, Illingworth RS. PRC2 promotes canalisation during endodermal differentiation. PLoS Genet 2025; 21:e1011584. [PMID: 39883738 PMCID: PMC11813121 DOI: 10.1371/journal.pgen.1011584] [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: 08/27/2024] [Revised: 02/11/2025] [Accepted: 01/20/2025] [Indexed: 02/01/2025] Open
Abstract
The genetic circuitry that encodes the developmental programme of mammals is regulated by transcription factors and chromatin modifiers. During early gestation, the three embryonic germ layers are established in a process termed gastrulation. The impact of deleterious mutations in chromatin modifiers such as the polycomb proteins manifests during gastrulation, leading to early developmental failure and lethality in mouse models. Embryonic stem cells have provided key insights into the molecular function of polycomb proteins, but it is impossible to fully appreciate the role of these epigenetic factors in development, or how development is perturbed due to their deficiency, in the steady-state. To address this, we have employed a tractable embryonic stem cell differentiation system to model primitive streak formation and early gastrulation. Using this approach, we find that loss of the repressive polycomb mark H3K27me3 is delayed relative to transcriptional activation, indicating a subordinate rather than instructive role in gene repression. Despite this, chemical inhibition of polycomb enhanced endodermal differentiation efficiency, but did so at the cost of lineage fidelity. These findings highlight the importance of the polycomb system in stabilising the developmental transcriptional response and, in so doing, in shoring up cellular specification.
Collapse
Affiliation(s)
- Jurriaan Jochem Hölzenspies
- Novo Nordisk Foundation Center for Stem Cell Medicine—reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dipta Sengupta
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Wendy Anne Bickmore
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Joshua Mark Brickman
- Novo Nordisk Foundation Center for Stem Cell Medicine—reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert Scott Illingworth
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
40
|
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
|
41
|
Wu L, Liu X, Jiang Q, Li M, Liang M, Wang S, Wang R, Su L, Ni T, Dong N, Zhu L, Guan F, Zhu J, Zhang W, Wu M, Chen Y, Chen T, Wang B. Methamphetamine-induced impairment of memory and fleeting neuroinflammation: Profiling mRNA changes in mouse hippocampus following short-term and long-term exposure. Neuropharmacology 2024; 261:110175. [PMID: 39357738 DOI: 10.1016/j.neuropharm.2024.110175] [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: 02/27/2024] [Revised: 08/20/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Methamphetamine (METH) has been implicated in inducing memory impairment, but the precise mechanisms underlying this effect remain unclear. Current research often limits itself to singular models or focuses on individual gene or protein functions, which hampers a comprehensive understanding of the underlying mechanisms. In this study, we established three METH mouse exposure models, extracted hippocampal nuclei, and utilized RNA sequencing to analyze changes in mRNA expression profiles. Our results indicate that METH significantly impairs the learning and memory capabilities of mice. Additionally, we observed that METH-induced inflammatory responses occur in the early phase and do not further exacerbate with repeated injections. However, RNA sequencing revealed the persistent enrichment of inflammatory pathway molecules, which correlated with worsened behaviors. This suggests that although METH-induced neuroinflammation plays a critical role in learning and memory impairment, the continued enrichment of inflammatory pathway molecules is associated with behavioral outcomes. These findings provide crucial evidence for the potential application of immune intervention in METH-related disorders.
Collapse
Affiliation(s)
- Laiqiang Wu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Xiaorui Liu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Qingchen Jiang
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ming Li
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Min Liang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Shuai Wang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Rui Wang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Linlan Su
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Tong Ni
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Nan Dong
- School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Li Zhu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Fanglin Guan
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Jie Zhu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Wen Zhang
- Department of Pathology, Northwest Women's and Children's Hospital, Xi'an, China
| | - Min Wu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Yanjiong Chen
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Teng Chen
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China.
| | - Biao Wang
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.
| |
Collapse
|
42
|
Fu R, Dou Z, Li N, Fan X, Amin S, Zhang J, Wang Y, Li Z, Li Z, Yang P. Avenanthramide A potentiates Bim-mediated antineoplastic properties of 5-fluorouracil via targeting KDM4C/ MIR17HG/GSK-3 β negative feedback loop in colorectal cancer. Acta Pharm Sin B 2024; 14:5321-5340. [PMID: 39807336 PMCID: PMC11725033 DOI: 10.1016/j.apsb.2024.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/24/2024] [Accepted: 07/10/2024] [Indexed: 01/16/2025] Open
Abstract
Chemoresistance to 5-fluorouracil (5-FU) is a significant challenge in treating colorectal cancer (CRC). Novel combined regimens to thwart chemoresistance are therefore urgently needed. Herein, we demonstrated that the combination of Avenanthramide A (AVN A) and 5-FU has significant therapeutic advantages against CRC. Mechanistically, AVN A directly binds to the S198 site of the histone lysine demethylase KDM4C to promote its degradation, which subsequently fosters H3K9me3 occupancy on the MIR17HG promoter to block its transcription and derepress Bim expression. AVN A enhanced the therapeutic efficacy of 5-FU via impairing the KDM4C/MIR17HG/GSK-3β negative feedback loop. Importantly, the clinical correlation of the KDM4C/MIR17HG/Bim signaling axis with 5-FU response was validated in the refractory CRC patients. We provide evidence for the enhanced effectiveness of 5-FU when combined with AVN A in chemoresistant xenografts, CRC organoids, and Apc Min/+ mouse model. Additionally, AVN A mitigated the systemic adverse effects of 5-FU. Overall, our findings demonstrate that combinatorial therapy with AVN A and 5-FU represents an appealing opportunity and highlights KDM4C/MIR17HG/GSK-3β negative feedback loop which confers therapeutically exploitable vulnerability to chemo-refractory CRC patients.
Collapse
Affiliation(s)
- Rong Fu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zhangfeng Dou
- Department of Gastroenterology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Ning Li
- Department of Gastroenterology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xueyuan Fan
- School of Life Science, Shanxi University, Taiyuan 030006, China
| | - Sajid Amin
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Jinqi Zhang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Yuqing Wang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zongwei Li
- School of Life Science, Anhui Medical University, Hefei 230032, China
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Peng Yang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
43
|
Foong YH, Caldwell B, Thorvaldsen JL, Krapp C, Mesaros CA, Zhou W, Kohli RM, Bartolomei MS. TET1 displays catalytic and non-catalytic functions in the adult mouse cortex. Epigenetics 2024; 19:2374979. [PMID: 38970823 PMCID: PMC11229741 DOI: 10.1080/15592294.2024.2374979] [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: 01/23/2024] [Accepted: 06/26/2024] [Indexed: 07/08/2024] Open
Abstract
TET1/2/3 dioxygenases iteratively demethylate 5-methylcytosine, beginning with the formation of 5-hydroxymethylcytosine (5hmC). The post-mitotic brain maintains higher levels of 5hmC than most peripheral tissues, and TET1 ablation studies have underscored the critical role of TET1 in brain physiology. However, deletion of Tet1 precludes the disentangling of the catalytic and non-catalytic functions of TET1. Here, we dissect these functions of TET1 by comparing adult cortex of Tet1 wildtype (Tet1 WT), a novel Tet1 catalytically dead mutant (Tet1 HxD), and Tet1 knockout (Tet1 KO) mice. Using DNA methylation array, we uncover that Tet1 HxD and KO mutations perturb the methylation status of distinct subsets of CpG sites. Gene ontology (GO) analysis on specific differential 5hmC regions indicates that TET1's catalytic activity is linked to neuronal-specific functions. RNA-Seq further shows that Tet1 mutations predominantly impact the genes that are associated with alternative splicing. Lastly, we performed High-performance Liquid Chromatography Mass-Spectrometry lipidomics on WT and mutant cortices and uncover accumulation of lysophospholipids lysophosphatidylethanolamine and lysophosphatidylcholine in Tet1 HxD cortex. In summary, we show that Tet1 HxD does not completely phenocopy Tet1 KO, providing evidence that TET1 modulates distinct cortical functions through its catalytic and non-catalytic roles.
Collapse
Affiliation(s)
- Yee Hoon Foong
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
| | - Blake Caldwell
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Joanne L. Thorvaldsen
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
| | - Christopher Krapp
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
| | - Clementina A. Mesaros
- Translational Biomarkers Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wanding Zhou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Children’s Hospital of Philadelphia (CHOP), University of Pennsylvania, Philadelphia, PA, USA
- Penn Epigenetics Institute, Smilow Center for Translational Rsearch, Philadelphia, PA, USA
| | - Rahul M. Kohli
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Epigenetics Institute, Smilow Center for Translational Rsearch, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marisa S. Bartolomei
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
- Penn Epigenetics Institute, Smilow Center for Translational Rsearch, Philadelphia, PA, USA
| |
Collapse
|
44
|
Perez AA, Goronzy IN, Blanco MR, Yeh BT, Guo JK, Lopes CS, Ettlin O, Burr A, Guttman M. ChIP-DIP maps binding of hundreds of proteins to DNA simultaneously and identifies diverse gene regulatory elements. Nat Genet 2024; 56:2827-2841. [PMID: 39587360 DOI: 10.1038/s41588-024-02000-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 10/21/2024] [Indexed: 11/27/2024]
Abstract
Gene expression is controlled by dynamic localization of thousands of regulatory proteins to precise genomic regions. Understanding this cell type-specific process has been a longstanding goal yet remains challenging because DNA-protein mapping methods generally study one protein at a time. Here, to address this, we developed chromatin immunoprecipitation done in parallel (ChIP-DIP) to generate genome-wide maps of hundreds of diverse regulatory proteins in a single experiment. ChIP-DIP produces highly accurate maps within large pools (>160 proteins) for all classes of DNA-associated proteins, including modified histones, chromatin regulators and transcription factors and across multiple conditions simultaneously. First, we used ChIP-DIP to measure temporal chromatin dynamics in primary dendritic cells following LPS stimulation. Next, we explored quantitative combinations of histone modifications that define distinct classes of regulatory elements and characterized their functional activity in human and mouse cell lines. Overall, ChIP-DIP generates context-specific protein localization maps at consortium scale within any molecular biology laboratory and experimental system.
Collapse
Affiliation(s)
- Andrew A Perez
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Isabel N Goronzy
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Mario R Blanco
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Benjamin T Yeh
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jimmy K Guo
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Carolina S Lopes
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Olivia Ettlin
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Alex Burr
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Mitchell Guttman
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
45
|
Maslan A, Altemose N, Marcus J, Mishra R, Brennan LD, Sundararajan K, Karpen G, Straight AF, Streets A. Mapping protein-DNA interactions with DiMeLo-seq. Nat Protoc 2024; 19:3697-3720. [PMID: 39237830 PMCID: PMC11674881 DOI: 10.1038/s41596-024-01032-9] [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: 07/03/2022] [Accepted: 06/04/2024] [Indexed: 09/07/2024]
Abstract
We recently developed directed methylation with long-read sequencing (DiMeLo-seq) to map protein-DNA interactions genome wide. DiMeLo-seq is capable of mapping multiple interaction sites on single DNA molecules, profiling protein binding in the context of endogenous DNA methylation, identifying haplotype-specific protein-DNA interactions and mapping protein-DNA interactions in repetitive regions of the genome that are difficult to study with short-read methods. With DiMeLo-seq, adenines in the vicinity of a protein of interest are methylated in situ by tethering the Hia5 methyltransferase to an antibody using protein A. Protein-DNA interactions are then detected by direct readout of adenine methylation with long-read, single-molecule DNA sequencing platforms such as Nanopore sequencing. Here we present a detailed protocol and practical guidance for performing DiMeLo-seq. This protocol can be run on nuclei from fresh, lightly fixed or frozen cells. The protocol requires 1-2 d for performing in situ targeted methylation, 1-5 d for library preparation depending on desired fragment length and 1-3 d for Nanopore sequencing depending on desired sequencing depth. The protocol requires basic molecular biology skills and equipment, as well as access to a Nanopore sequencer. We also provide a Python package, dimelo, for analysis of DiMeLo-seq data.
Collapse
Affiliation(s)
- Annie Maslan
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Nicolas Altemose
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jeremy Marcus
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Reet Mishra
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Lucy D Brennan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | | | - Gary Karpen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of BioEngineering and BioMedical Sciences, Lawrence Berkeley National Laboratory, Berkeley, USA
| | - Aaron F Straight
- Department of Biochemistry, Stanford University, Stanford, CA, USA
| | - Aaron Streets
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA.
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, USA.
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
46
|
Liao C, Walters BW, DiStasio M, Lesch BJ. Human-specific epigenomic states in spermatogenesis. Comput Struct Biotechnol J 2024; 23:577-588. [PMID: 38274996 PMCID: PMC10809009 DOI: 10.1016/j.csbj.2023.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/23/2023] [Accepted: 12/23/2023] [Indexed: 01/27/2024] Open
Abstract
Infertility is becoming increasingly common, affecting one in six people globally. Half of these cases can be attributed to male factors, many driven by abnormalities in the process of sperm development. Emerging evidence from genome-wide association studies, genetic screening of patient cohorts, and animal models highlights an important genetic contribution to spermatogenic defects, but comprehensive identification and characterization of the genes critical for male fertility remain lacking. High divergence of gene regulation in spermatogenic cells across species poses challenges for delineating the genetic pathways required for human spermatogenesis using common model organisms. In this study, we leveraged post-translational histone modification and gene transcription data for 15,491 genes in four mammalian species (human, rhesus macaque, mouse, and opossum), to identify human-specific patterns of gene regulation during spermatogenesis. We combined H3K27me3 ChIP-seq, H3K4me3 ChIP-seq, and RNA-seq data to define epigenetic states for each gene at two stages of spermatogenesis, pachytene spermatocytes and round spermatids, in each species. We identified 239 genes that are uniquely active, poised, or dynamically regulated in human spermatogenic cells distinct from the other three species. While some of these genes have been implicated in reproductive functions, many more have not yet been associated with human infertility and may be candidates for further molecular and epidemiologic studies. Our analysis offers an example of the opportunities provided by evolutionary and epigenomic data for broadly screening candidate genes implicated in reproduction, which might lead to discoveries of novel genetic targets for diagnosis and management of male infertility and male contraception.
Collapse
Affiliation(s)
- Caiyun Liao
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | | | - Marcello DiStasio
- Department of Pathology, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
- Department of Opthamology & Visual Sciences, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | - Bluma J. Lesch
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
- Department of Genetics, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
- Yale Cancer Center, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
47
|
Cohen LRZ, Meshorer E. The many faces of H3.3 in regulating chromatin in embryonic stem cells and beyond. Trends Cell Biol 2024; 34:1044-1055. [PMID: 38614918 DOI: 10.1016/j.tcb.2024.03.003] [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: 01/22/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/15/2024]
Abstract
H3.3 is a highly conserved nonreplicative histone variant. H3.3 is enriched in promoters and enhancers of active genes, but it is also found within suppressed heterochromatin, mostly around telomeres. Accordingly, H3.3 is associated with seemingly contradicting functions: It is involved in development, differentiation, reprogramming, and cell fate, as well as in heterochromatin formation and maintenance, and the silencing of developmental genes. The emerging view is that different cellular contexts and histone modifications can promote opposing functions for H3.3. Here, we aim to provide an update with a focus on H3.3 functions in early mammalian development, considering the context of embryonic stem cell maintenance and differentiation, to finally conclude with emerging roles in cancer development and cell fate transition and maintenance.
Collapse
Affiliation(s)
- Lea R Z Cohen
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eran Meshorer
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
48
|
Lu S, Zheng Z, Zhu C. Histone methyltransferase WHSC1 cooperate with YBX1 promote glioblastoma progression via regulating PLK1 expression. Cell Signal 2024; 124:111471. [PMID: 39406278 DOI: 10.1016/j.cellsig.2024.111471] [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: 07/22/2024] [Revised: 10/03/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
Wolf-Hirschhorn syndrome candidate gene 1 (WHSC1), a histone methyltransferase, has been implicated in various tumor development processes by regulating target gene expression. However, the role of WHSC1 in glioblastoma remains unexplored. This study investigates the impact of WHSC1 in glioblastoma and its association with prognosis. Our findings reveal that WHSC1 is overexpressed in glioblastoma and correlates with poor patient outcomes. Functional assays demonstrate that the reduction of WHSC1 significantly impairs cell proliferation and tumorigenicity. Mechanistically, WHSC1 modulates PLK1 expression by binding to its promoter region, leading to the activation of the PLK1-AKT pathway, and regulating H3K36 dimethylation levels. Furthermore, YBX1 can cooperate with WHSC1 to activate PLK1 transcription. These results shed light on the potential significance of WHSC1 in glioblastoma and offer a promising avenue for future therapeutic approaches targeting this molecule in glioblastoma treatment.
Collapse
Affiliation(s)
- Shuaijun Lu
- The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Zhibo Zheng
- The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Changling Zhu
- The First Affiliated Hospital of Ningbo University, Ningbo 315020, China.
| |
Collapse
|
49
|
Llewellyn J, Baratam R, Culig L, Beerman I. Cellular stress and epigenetic regulation in adult stem cells. Life Sci Alliance 2024; 7:e202302083. [PMID: 39348938 PMCID: PMC11443024 DOI: 10.26508/lsa.202302083] [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: 04/07/2023] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024] Open
Abstract
Stem cells are a unique class of cells that possess the ability to differentiate and self-renew, enabling them to repair and replenish tissues. To protect and maintain the potential of stem cells, the cells and the environment surrounding these cells (stem cell niche) are highly responsive and tightly regulated. However, various stresses can affect the stem cells and their niches. These stresses are both systemic and cellular and can arise from intrinsic or extrinsic factors which would have strong implications on overall aging and certain disease states. Therefore, understanding the breadth of drivers, namely epigenetic alterations, involved in cellular stress is important for the development of interventions aimed at maintaining healthy stem cells and tissue homeostasis. In this review, we summarize published findings of epigenetic responses to replicative, oxidative, mechanical, and inflammatory stress on various types of adult stem cells.
Collapse
Affiliation(s)
- Joey Llewellyn
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Rithvik Baratam
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Luka Culig
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| |
Collapse
|
50
|
VanBelzen J, Sakelaris B, Brickner DG, Marcou N, Riecke H, Mangan NM, Brickner JH. Chromatin endogenous cleavage provides a global view of yeast RNA polymerase II transcription kinetics. eLife 2024; 13:RP100764. [PMID: 39607887 PMCID: PMC11604220 DOI: 10.7554/elife.100764] [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: 11/30/2024] Open
Abstract
Chromatin immunoprecipitation (ChIP-seq) is the most common approach to observe global binding of proteins to DNA in vivo. The occupancy of transcription factors (TFs) from ChIP-seq agrees well with an alternative method, chromatin endogenous cleavage (ChEC-seq2). However, ChIP-seq and ChEC-seq2 reveal strikingly different patterns of enrichment of yeast RNA polymerase II (RNAPII). We hypothesized that this reflects distinct populations of RNAPII, some of which are captured by ChIP-seq and some of which are captured by ChEC-seq2. RNAPII association with enhancers and promoters - predicted from biochemical studies - is detected well by ChEC-seq2 but not by ChIP-seq. Enhancer/promoter-bound RNAPII correlates with transcription levels and matches predicted occupancy based on published rates of enhancer recruitment, preinitiation assembly, initiation, elongation, and termination. The occupancy from ChEC-seq2 allowed us to develop a stochastic model for global kinetics of RNAPII transcription which captured both the ChEC-seq2 data and changes upon chemical-genetic perturbations to transcription. Finally, RNAPII ChEC-seq2 and kinetic modeling suggests that a mutation in the Gcn4 transcription factor that blocks interaction with the NPC destabilizes promoter-associated RNAPII without altering its recruitment to the enhancer.
Collapse
Affiliation(s)
- Jake VanBelzen
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Bennet Sakelaris
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States
| | - Donna G Brickner
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Nikita Marcou
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Hermann Riecke
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States
| | - Niall M Mangan
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States
| | - Jason H Brickner
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| |
Collapse
|