1
|
Reddy IA, Han L, Sanchez-Roige S, Niarchou M, Ruderfer DM, Davis LK. Identification of Transdiagnostic Childhood Externalizing Pathology Within an Electronic Medical Records Database and Application to the Analysis of Rare Copy Number Variation. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33020. [PMID: 39744833 PMCID: PMC12048253 DOI: 10.1002/ajmg.b.33020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/18/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025]
Abstract
Externalizing traits and behaviors are broadly defined by impairments in self-regulation and impulse control that typically begin in childhood and adolescence. Externalizing behaviors, traits, and symptoms span a range of traditional psychiatric diagnostic categories. In this study, we sought to generate an algorithm that could reliably identify transdiagnostic childhood-onset externalizing cases and controls within a university hospital electronic health record (EHR) database. Within the Vanderbilt University Medical Center (VUMC) EHR, our algorithm identified cases with a clinician-validated positive predictive value of 90% and controls with a negative predictive value of 88%. In individuals of genetically defined European ancestry (CEU-clustered; Ncase = 487, Ncontrol = 5638), case status was significantly associated with psychiatric comorbidity and with elevated externalizing polygenic scores (OR: 1.20; 95% CI: 1.09-1.33; p = 1.14 × 10-3; based on published genome-wide association data). To test whether our cohort definitions could be applied to generate novel genetic insights, we examined rare (allele frequency < 0.5%) copy number variation. An association (OR: 9.70; CI: 3.24-29.0) was identified in the CEU-clustered cohort on chromosome 2 (chr2: 45,408,678-45,551,530; duplication), although the statistical strength of this association was modest (p = 0.052). We also examined the role of an externalizing burden score based on the number of externalizing diagnoses present in cases and found similar results to our case-control analysis. This analysis identified several other statistically significant CNV region associations. This study provides a framework for identifying childhood externalizing case-control cohorts within an EHR. Future work should validate this framework within other health systems. A broadly applicable algorithm, like this one, may allow for detection of rare outcomes or outcomes in populations historically excluded from genomic research through meta-analysis of data across health care systems.
Collapse
Affiliation(s)
- India A Reddy
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lide Han
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sandra Sanchez-Roige
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, California, USA
| | - Maria Niarchou
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Douglas M Ruderfer
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lea K Davis
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Division of Data-Driven and Digital Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
2
|
Li J, Xu L, Liang X, Li L, Farnir F, Huang X, Chen Q. A multi-tissue atlas of allelic-specific expression reveals the characteristics, mechanisms, and relationship with dominant effects in cattle. BMC Biol 2025; 23:148. [PMID: 40442638 PMCID: PMC12123890 DOI: 10.1186/s12915-025-02257-0] [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: 11/26/2024] [Accepted: 05/21/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Allele-specific expression (ASE) analysis is a crucial tool for validating expression quantitative trait loci (eQTLs), identifying causal variants associated with complex traits, and investigating the genetic mechanisms underlying heterosis. In this study, we characterized ASE variants across 35 tissues using 7532 publicly available RNA-seq datasets. Additionally, we explored the mechanisms driving ASE through integration with epigenomic data and examined the relationship between ASE and dominance effects on gene expression and milk-related traits in Holstein cattle. RESULTS ASE variants exhibited stronger tissue specificity and lower reproducibility compared to eQTLs. Interestingly, variants with opposite directional effects demonstrated greater resilience across diverse environments. Functional annotation revealed that ASE variants were enriched in both enhancer and promoter regions during transcription and implicated in post-transcriptional and translational processes, including mutations that affect mRNA splicing and trigger nonsense-mediated decay. Analysis of eQTLs, splicing QTLs (sQTLs), and validated QTLs associated with milk-related traits in Holstein cattle, coupled with enrichment analysis in QTL databases and effect size evaluation, indicated that ASE variants were more closely aligned with dominant effects than additive effects, particularly in reproductive and immune-related tissues/traits, which exhibited higher levels of heterosis. CONCLUSIONS Our findings not only enhance our understanding of the genetic mechanisms underlying heterosis and ASE formation but also provide a valuable resource of regulatory variants that can be leveraged to improve economic traits through molecular breeding or the strategic exploitation of heterosis.
Collapse
Affiliation(s)
- Jiaqi Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China
| | - Lei Xu
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China
| | - Xiaoyun Liang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China
| | - Letian Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China
| | - Frederic Farnir
- Faculte de Medecine Veterinaire, Universite de Liege, Quartier Vallee 2, Avenue de Cureghem 6 (B43), Liege, 4000, Belgium
| | - Xixia Huang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China.
| | - Qiuming Chen
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China.
| |
Collapse
|
3
|
Velazquez-Rivera E, Dey O, Kim NS, Cao W, Ye Q, Gao P, Thai A, Nguyen JK, Zhang H, Ting JT, Gopi M, Ren B, Holmes TC, Xu X. Specific targeting of brain endothelial cells using enhancer AAV vectors. Neuron 2025; 113:1562-1578.e6. [PMID: 40403707 DOI: 10.1016/j.neuron.2025.03.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/24/2025] [Accepted: 03/26/2025] [Indexed: 05/24/2025]
Abstract
Brain endothelial cells (BECs) in brain vasculature are critical structural and functional components of the blood brain barrier (BBB). Adeno-associated virus (AAV) capsids have previously been genetically engineered to confer specificity to endothelial cells, but these capsids show limited endothelial cell specificity that varies by delivery conditions. We developed a set of new BEC-enhancer AAV vectors that specifically target BECs based on the cis-regulatory elements identified from single-cell epigenetic datasets. Ex vivo and in vivo characterization of BEC-enhancer AAVs in wild-type, Ai9 reporter, and Alzheimer's disease model mouse brains show their utility for high transduction selectivity of the BECs with little off-target transduction in the liver. Our BEC-enhancer AAVs target the brain vasculature by systemic administration and can be minimally invasive in terms of the route of administration. They are useful new tools for delivering genetic payloads specifically to BECs for normal and diseased brain studies.
Collapse
Affiliation(s)
- Eric Velazquez-Rivera
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Oyshi Dey
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Nayoon S Kim
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Wenhao Cao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Qiao Ye
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Pan Gao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Andy Thai
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Jason K Nguyen
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Hai Zhang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Jonathan T Ting
- Human Cell Types Program, Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - M Gopi
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Center for Neural Circuit Mapping (CNCM), University of California, Irvine, Irvine, CA 92697, USA
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Center for Neural Circuit Mapping (CNCM), University of California, Irvine, Irvine, CA 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA 92697, USA; Center for Neural Circuit Mapping (CNCM), University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
4
|
Chen F, Zhang Y, Li W, Sedlazeck FJ, Shen L, Creighton CJ. Global DNA methylation differences involving germline structural variation impact gene expression in pediatric brain tumors. Nat Commun 2025; 16:4713. [PMID: 40399292 PMCID: PMC12095544 DOI: 10.1038/s41467-025-60110-y] [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: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
The extent of genetic variation and its influence on gene expression across multiple tissue and cellular contexts is still being characterized, with germline Structural Variants (SVs) being historically understudied. DNA methylation also represents a component of normal germline variation across individuals. Here, we combine germline SVs (by short-read sequencing) with tumor DNA methylation across 1292 pediatric brain tumor patients. For thousands of methylation probes for CpG Islands (CGIs) or enhancers, rare and common SV breakpoints upstream or downstream associate with differential methylation in tumors spanning various histologic types, a significant subset involving genes with SV-associated differential expression. Cancer predisposition genes involving SV-associated differential methylation and expression include MSH2, RSPA, and PALB2. SV breakpoints falling within CGIs or histone marks H3K36me3 or H3K9me3 associate with differential CGI methylation. Genes with SVs and CGI methylation associated with patient survival include POLD4. Our results capture a class of normal phenotypic variation having disease implications.
Collapse
Affiliation(s)
- Fengju Chen
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Computer Science, Rice University, Houston, TX, 77005, USA
| | - Lanlan Shen
- USDA Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Xiong H, Shen P, Luo Q, Zhang L, Li B, Ding Z, Wang L. Elucidating the Genetic Underpinnings of Human Musculoskeletal System Aging Through Genomic Structural Equation Modeling. Clin Genet 2025. [PMID: 40369705 DOI: 10.1111/cge.14766] [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: 02/12/2025] [Revised: 04/11/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
The genetic architecture underlying traits related to Human Musculoskeletal System Aging (MSA) remains largely unexplored. In this study, we conducted a large-scale multivariate genome-wide association study (GWAS) of MSA utilizing Genomic Structural Equation Modeling (Genomic SEM). We estimated causal single nucleotide polymorphisms (SNPs) associated with independent variation and identified 14 genome-wide significant loci (mean.PP > 0.95). We employed multiple transcriptome-wide association methods to analyze tissue, cellular levels, and genomic elements, identifying loci with high relevance to MSA susceptibility, along with associated element information. Our research represents the first comprehensive delineation of the genetic architecture of Musculoskeletal System Aging through a GWAS of unmeasured phenotype.
Collapse
Affiliation(s)
- Hao Xiong
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Pan Shen
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Qinghua Luo
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Leichang Zhang
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Department of Anorectal Surgery, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Bo Li
- Medical Department, Sias University, Zhengzhou, China
| | - Zhaohui Ding
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Pulmonary Disease Department, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Lihua Wang
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Pulmonary Disease Department, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
6
|
Erdmann É, Agolli S, Fix S, Cottard F, Keyser C, Zvenigorosky V, Gonzalez A, Haili Z, Kieffer B, Céraline J. Human-specific genomic evolution of a regulatory network enables fine-tuning of N-cadherin gene expression. Cell Mol Life Sci 2025; 82:196. [PMID: 40343501 PMCID: PMC12064536 DOI: 10.1007/s00018-025-05725-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/04/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Androgen receptor (AR), a member of the nuclear receptor superfamily controls prostate epithelial cell plasticity by repressing a panel of genes involved in epithelial-mesenchymal transition (EMT), including the human CDH2 gene encoding N-cadherin. At the opposite, pathological AR variants such as AR-V7 associated with prostate tumor progression upregulate those EMT genes. Here, focusing on the human CDH2 gene, we show that this duality between AR and AR-V7 relies on a potential human accelerated region present in the intron 1. This fastest-evolving region of the human genome is actually a variable number tandem repeat (VNTR) comprising 24 repetitions of a DNA sequence that englobes binding sites for steroid hormone receptors, recombination signal binding protein for immunoglobulin kappa j region (RBPJ) an effector of the Notch pathway, and zinc finger e-box binding homeobox 1 (ZEB1). Genomic DNA sequencing, multiple sequence alignment, data mining, as well as protein-DNA interaction and gene expression analyses indicate that this VNTR constitutes a potential transcriptional hub for different transcription factors to control human CDH2 expression. Also, our data suggest that prostate tumor cells may unlock an up to now unknown molecular mechanism associated with a fine-tuned control of human CDH2 gene expression.
Collapse
Affiliation(s)
- Éva Erdmann
- CNRS UMR 7104, INSERM U1258, IGBMC, Université de Strasbourg, Illkirch, 67404, France
| | - Savera Agolli
- CNRS UMR 7104, INSERM U1258, IGBMC, Université de Strasbourg, Illkirch, 67404, France
| | - Simon Fix
- CNRS UMR 7104, INSERM U1258, IGBMC, Université de Strasbourg, Illkirch, 67404, France
| | - Félicie Cottard
- CNRS UMR 7242, ESBS, Université de Strasbourg, Illkirch, 67404, France
| | | | | | - Angéla Gonzalez
- Strasbourg Institute of Legal Medicine, Strasbourg, 67085, France
| | - Zakary Haili
- CNRS UMR 7104, INSERM U1258, IGBMC, Université de Strasbourg, Illkirch, 67404, France
| | - Bruno Kieffer
- CNRS UMR 7104, INSERM U1258, IGBMC, Université de Strasbourg, Illkirch, 67404, France
| | - Jocelyn Céraline
- CNRS UMR 7104, INSERM U1258, IGBMC, Université de Strasbourg, Illkirch, 67404, France.
- Hôpitaux Universitaires de Strasbourg, Strasbourg, 67091, France.
- Fédération de Médecine Translationnelle de Strasbourg, FMTS, Université de Strasbourg, Strasbourg, 67085, France.
- CNRS UMR 7104, INSERM U1258, IGBMC, 1, rue Laurent Fries, Illkirch, 67404, France.
| |
Collapse
|
7
|
Carrington JT, Wilson RHC, de La Vega E, Thiyagarajan S, Barker T, Catchpole L, Durrant A, Knitlhoffer V, Watkins C, Gharbi K, Nieduszynski CA. Most human DNA replication initiation is dispersed throughout the genome with only a minority within previously identified initiation zones. Genome Biol 2025; 26:122. [PMID: 40346587 PMCID: PMC12063229 DOI: 10.1186/s13059-025-03591-w] [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: 04/29/2024] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND The identification of sites of DNA replication initiation in mammalian cells has been challenging. Here, we present unbiased detection of replication initiation events in human cells using BrdU incorporation and single-molecule nanopore sequencing. RESULTS Increases in BrdU incorporation allow us to measure DNA replication dynamics, including identification of replication initiation, fork direction, and termination on individual nanopore sequencing reads. Importantly, initiation and termination events are identified on single molecules with high resolution, throughout S-phase, genome-wide, and at high coverage at specific loci using targeted enrichment. We find a significant enrichment of initiation sites within the broad initiation zones identified by population-level studies. However, these focused initiation sites only account for ~ 20% of all identified replication initiation events. Most initiation events are dispersed throughout the genome and are missed by cell population approaches. This indicates that most initiation occurs at sites that, individually, are rarely used. These dispersed initiation sites contrast with the focused sites identified by population studies, in that they do not show a strong relationship to transcription or a particular epigenetic signature. CONCLUSIONS We show here that single-molecule sequencing enables unbiased detection and characterization of DNA replication initiation events, including the numerous dispersed initiation events that replicate most of the human genome.
Collapse
Affiliation(s)
| | | | | | | | - Tom Barker
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK
| | - Leah Catchpole
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK
| | - Alex Durrant
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK
| | | | - Chris Watkins
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK
| | - Karim Gharbi
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK
| | - Conrad A Nieduszynski
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK.
- University of East Anglia, Norwich, UK.
| |
Collapse
|
8
|
Wang P, Liu W, Wang J, Liu Y, Li P, Xu P, Cui W, Zhang R, Long Q, Hu Z, Fang C, Dong J, Zhang C, Chen Y, Wang C, Liu G, Xie H, Zhang Y, Xiao M, Chen S, Jiang H, Chen Y, Yang G, Zhang S, Meng Z, Wang X, Feng G, Li X, Zhou Y. scCompass: An Integrated Multi-Species scRNA-seq Database for AI-Ready. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500870. [PMID: 40317650 DOI: 10.1002/advs.202500870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/29/2025] [Indexed: 05/07/2025]
Abstract
Emerging single-cell sequencing technology has generated large amounts of data, allowing analysis of cellular dynamics and gene regulation at the single-cell resolution. Advances in artificial intelligence enhance life sciences research by delivering critical insights and optimizing data analysis processes. However, inconsistent data processing quality and standards remain to be a major challenge. Here scCompass is proposed, which provides a comprehensive resource designed to build large-scale, multi-species, and model-friendly single-cell data collection. By applying standardized data pre-processing, scCompass integrates and curates transcriptomic data from nearly 105 million single cells across 13 species. Using this extensive dataset, it is able to identify stable expression genes (SEGs) and organ-specific expression genes (OSGs) in humans and mice. Different scalable datasets are provided that can be easily adapted for AI model training and the pretrained checkpoints with state-of-the-art single-cell foundation models. In summary, scCompass is highly efficient and scalable database for AI-ready, which combined with user-friendly data sharing, visualization, and online analysis, greatly simplifies data access and exploitation for researchers in single-cell biology (http://www.bdbe.cn/kun).
Collapse
Affiliation(s)
- Pengfei Wang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Wenhao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Jiajia Wang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
| | - Yana Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Pengjiang Li
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Ping Xu
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Wentao Cui
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Ran Zhang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Qingqing Long
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Zhilong Hu
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Chen Fang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Jingxi Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Chunyang Zhang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
| | - Yan Chen
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
| | - Chengrui Wang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
| | - Guole Liu
- State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Hanyu Xie
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
| | - Yiyang Zhang
- CEMS, NCMIS, HCMS, MDIS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Meng Xiao
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
| | - Shubai Chen
- Beijing Key Laboratory of Mobile Computing and Pervasive Device, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Haiping Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Yiqiang Chen
- Beijing Key Laboratory of Mobile Computing and Pervasive Device, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Ge Yang
- State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Shihua Zhang
- CEMS, NCMIS, HCMS, MDIS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Zhen Meng
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Xuezhi Wang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Xin Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Yuanchun Zhou
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100083, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| |
Collapse
|
9
|
Tekola-Ayele F, Biedrzycki RJ, Habtewold TD, Wijesiriwardhana P, Burt A, Marsit CJ, Ouidir M, Wapner R. Sex-differentiated placental methylation and gene expression regulation has implications for neonatal traits and adult diseases. Nat Commun 2025; 16:4004. [PMID: 40312437 PMCID: PMC12045980 DOI: 10.1038/s41467-025-58128-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 03/10/2025] [Indexed: 05/03/2025] Open
Abstract
Sex differences in physiological and disease traits are pervasive and begin during early development, but the genetic architecture of these differences is largely unknown. Here, we leverage the human placenta, a transient organ during pregnancy critical to fetal development, to investigate the impact of sex in the regulatory landscape of placental autosomal methylome and transcriptome, and its relevance to health and disease. We find that placental methylation and its genetic regulation are extensively impacted by fetal sex, whereas sex differences in placental gene expression and its genetic regulation are limited. We identify molecular processes and regulatory targets that are enriched in a sex-specific manner, and find enrichment of imprinted genes in sex-differentiated placental methylation, including female-biased methylation within the well-known KCNQ1OT1/CDKN1C imprinting cluster of genes expressed in a parent-of-origin dependent manner. We establish that several sex-differentiated genetic effects on placental methylation and gene expression colocalize with birthweight and adult disease genetic associations, facilitating mechanistic insights on early life origins of health and disease outcomes shaped by sex.
Collapse
Affiliation(s)
- Fasil Tekola-Ayele
- Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| | - Richard J Biedrzycki
- Glotech, Inc., contractor for Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Tesfa Dejenie Habtewold
- Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Prabhavi Wijesiriwardhana
- Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Amber Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health of Emory University, Atlanta, GA, USA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health of Emory University, Atlanta, GA, USA
| | - Marion Ouidir
- Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- University of Grenoble Alpes, Inserm, Team of Environmental Epidemiology applied to Reproduction and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Ronald Wapner
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| |
Collapse
|
10
|
Puppione DL. Two rodent suborders have evolved missing amino acids in the lipid-binding region of apolipoprotein E. Lipids 2025; 60:143-153. [PMID: 39805706 PMCID: PMC12059251 DOI: 10.1002/lipd.12426] [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: 08/28/2024] [Revised: 10/29/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
The order Rodentia comprises nearly 45% of all extant taxa, currently organized into 31 living families, some 450 genera, and roughly 2010 species (Kelt & Patton, 2020). Considering that rodents began evolving at least 66 million years ago, it is not surprising that they have diversified into five distinct suborders. With the advent of molecular biology, this difference can often be seen at the molecular level as well. Previous studies have indicated that the apolipoprotein E (APOE) of guinea pigs, belonging to the suborder Hystricomorpha, have fewer amino acids than have been reported for other suborders of Rodentia. Searching the genomic database for hystricomorph APOE genes, it was found that hystricomorphs were missing residues both in the vicinity of the hinge region and in the lipid-binding region of the apolipoprotein. In the hinge region, missing residues varied between 5 and 3, and in the latter region, seven residues were missing. The search also revealed that castorimorphs, although lacking the smaller of the two deletions, were also missing the same seven residue deletion as found in APOE of the hystricomorphs.
Collapse
Affiliation(s)
- Don L. Puppione
- Molecular Biology Institute, University of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
11
|
Mori MP, Lozoya OA, Brooks AM, Bortner CD, Nadalutti CA, Ryback B, Rickard BP, Overchuk M, Rizvi I, Rogasevskaia T, Huang KT, Hasan P, Hajnóczky G, Santos JH. Mitochondrial membrane hyperpolarization modulates nuclear DNA methylation and gene expression through phospholipid remodeling. Nat Commun 2025; 16:4029. [PMID: 40301431 PMCID: PMC12041266 DOI: 10.1038/s41467-025-59427-5] [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: 06/06/2024] [Accepted: 04/23/2025] [Indexed: 05/01/2025] Open
Abstract
Maintenance of the mitochondrial inner membrane potential (ΔΨm) is critical for many aspects of mitochondrial function. While ΔΨm loss and its consequences are well studied, little is known about the effects of mitochondrial hyperpolarization. In this study, we used cells deleted of ATP5IF1 (IF1), a natural inhibitor of the hydrolytic activity of the ATP synthase, as a genetic model of increased resting ΔΨm. We found that the nuclear DNA hypermethylates when the ΔΨm is chronically high, regulating the transcription of mitochondrial, carbohydrate and lipid genes. These effects can be reversed by decreasing the ΔΨm and recapitulated in wild-type (WT) cells exposed to environmental chemicals that cause hyperpolarization. Surprisingly, phospholipid changes, but not redox or metabolic alterations, linked the ΔΨm to the epigenome. Sorted hyperpolarized WT and ovarian cancer cells naturally depleted of IF1 also showed phospholipid remodeling, indicating this as an adaptation to mitochondrial hyperpolarization. These data provide a new framework for how mitochondria can impact epigenetics and cellular biology to influence health outcomes, including through chemical exposures and in disease states.
Collapse
Affiliation(s)
- Mateus Prates Mori
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Oswaldo A Lozoya
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Ashley M Brooks
- Biostatistics and Computational Biology Branch, Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Carl D Bortner
- Flow Cytometry Center, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Cristina A Nadalutti
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Birgitta Ryback
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Brittany P Rickard
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina (UNC), Chapel Hill, NC, USA
| | - Marta Overchuk
- Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, USA
| | - Imran Rizvi
- Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, USA
- Lineberger Comprehensive Cancer Center, UNC, Chapel Hill, NC, USA
| | | | - Kai Ting Huang
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Prottoy Hasan
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - György Hajnóczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Janine H Santos
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA.
| |
Collapse
|
12
|
Wang G, Li G, Song A, Zhao Y, Yu J, Wang Y, Dai W, Salas M, Qin H, Medrano L, Dow J, Li A, Armstrong B, Fueger PT, Yu H, Zhu Y, Shao M, Wu X, Jiang L, Campisi J, Yang X, Wang QA. Distinct adipose progenitor cells emerging with age drive active adipogenesis. Science 2025; 388:eadj0430. [PMID: 40273250 DOI: 10.1126/science.adj0430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 05/30/2024] [Accepted: 02/05/2025] [Indexed: 04/26/2025]
Abstract
Starting at middle age, adults often suffer from visceral adiposity and associated adverse metabolic disorders. Lineage tracing in mice revealed that adipose progenitor cells (APCs) in visceral fat undergo extensive adipogenesis during middle age. Thus, despite the low turnover rate of adipocytes in young adults, adipogenesis is unlocked during middle age. Transplantations quantitatively showed that APCs in middle-aged mice exhibited high adipogenic capacity cell-autonomously. Single-cell RNA sequencing identified a distinct APC population, the committed preadipocyte, age-enriched (CP-A), emerging at this age. CP-As demonstrated elevated proliferation and adipogenesis activity. Pharmacological and genetic manipulations indicated that leukemia inhibitory factor receptor signaling was indispensable for CP-A adipogenesis and visceral fat expansion. These findings uncover a fundamental mechanism of age-dependent adipose remodeling, offering critical insights into age-related metabolic diseases.
Collapse
Affiliation(s)
- Guan Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Gaoyan Li
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anying Song
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yutian Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiayu Yu
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yifan Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Wenting Dai
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Martha Salas
- Light Microscopy Core, City of Hope Medical Center, Duarte, CA, USA
| | - Hanjun Qin
- The Integrative Genomics Core, City of Hope Medical Center, Duarte, CA, USA
| | - Leonard Medrano
- Division of Developmental and Translational Diabetes and Endocrinology Research, City of Hope Medical Center, Duarte, CA, USA
| | - Joan Dow
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Metabolic Phenotyping Core, City of Hope Medical Center, Duarte, CA, USA
| | - Aimin Li
- Pathology Core of Shared Resources, City of Hope Medical Center, Duarte, CA, USA
| | - Brian Armstrong
- Light Microscopy Core, City of Hope Medical Center, Duarte, CA, USA
| | - Patrick T Fueger
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Metabolic Phenotyping Core, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Hua Yu
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yi Zhu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Mengle Shao
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Xiwei Wu
- The Integrative Genomics Core, City of Hope Medical Center, Duarte, CA, USA
| | - Lei Jiang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | | | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Qiong A Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| |
Collapse
|
13
|
Xie L, Cao B, Wen X, Zheng Y, Wang B, Zhou S, Zheng P. ReLume: Enhancing DNA storage data reconstruction with flow network and graph partitioning. Methods 2025; 240:101-112. [PMID: 40268154 DOI: 10.1016/j.ymeth.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/06/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
DNA storage is an ideal alternative to silicon-based storage, but focusing on data writing alone cannot address the inevitable errors and durability issues. Therefore, we propose ReLume, a DNA storage data reconstruction method based on flow networks and graph partitioning technology, which can accomplish the data reconstruction task of millions of reads on a laptop with 24 GB RAM. The results show that ReLume copes well with many types of errors, more than doubles sequence recovery rates, and reduces memory usage by about 60 %. ReLume is 10 times more durable than other representative methods, meaning that data can be read without loss after 100 years. Results from the wet lab DNA storage dataset show that ReLume's sequence recovery rates of 73 % and 93.2 %, respectively, significantly outperform existing methods. In summary, ReLume effectively overcomes the accuracy and hardware limitations and provides a feasible idea for the portability of DNA storage.
Collapse
Affiliation(s)
- Lei Xie
- Key Laboratory of Advanced Design and Intelligent Computing, Ministry of Education, School of Software Engineering, Dalian University, Dalian 116622, PR China
| | - Ben Cao
- School of Computer Science and Technology, Dalian University of Technology, 116024 Dalian, PR China
| | - Xiaoru Wen
- Key Laboratory of Advanced Design and Intelligent Computing, Ministry of Education, School of Software Engineering, Dalian University, Dalian 116622, PR China
| | - Yanfen Zheng
- School of Computer Science and Technology, Dalian University of Technology, 116024 Dalian, PR China
| | - Bin Wang
- Key Laboratory of Advanced Design and Intelligent Computing, Ministry of Education, School of Software Engineering, Dalian University, Dalian 116622, PR China.
| | - Shihua Zhou
- Key Laboratory of Advanced Design and Intelligent Computing, Ministry of Education, School of Software Engineering, Dalian University, Dalian 116622, PR China.
| | - Pan Zheng
- Department of Accounting and Information Systems, University of Canterbury, 8140 Christchurch, New Zealand
| |
Collapse
|
14
|
Mehta S, Wagner R, Do KT, Johnson JE, Yu F, Jubenville T, Richards K, Coleman S, Popescu FE, Nesvizhskii AI, Largaespada DA, Jagtap PD, Griffin TJ. A modular, Galaxy-based immunopeptidogenomic (iPepGen) analysis pipeline for discovery, verification, and prioritization of candidate cancer neoantigen peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647596. [PMID: 40291680 PMCID: PMC12026984 DOI: 10.1101/2025.04.07.647596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background Characterizing peptide antigens, processed from tumor-specific proteoforms, and bound to the major histocompatibility complex, is critical for immuno-oncology research. Next-generation sequencing predicts candidate neoantigen peptides derived from DNA mutations and/or RNA transcripts coding proteoform sequences that differ from the reference proteome. Mass spectrometry (MS)-based immunopeptidomics identifies predicted, MHC-bound neoantigen peptides and other tumor antigens. This "immunopeptidogenomic" approach requires multi-omic software integration, challenging researchers with limited bioinformatics expertise and resources. As a solution, we developed the immunopeptidogenomic (iPepGen) pipeline in the Galaxy ecosystem. iPepGen is composed of five core workflow modules, available via publicly accessible, scalable Galaxy instances, accompanied by training resources to empower community adoption. Findings Using representative multi-omic data from malignant peripheral nerve sheath tumors, we demonstrate the operation of iPepGen modules with these functions: 1) Predict neoantigen candidates from sequencing data and generate customized protein sequence databases, including reference and non-reference neoantigen candidate sequences; 2) Discover neoantigen peptide candidates by sequence database searching of tandem mass spectrometry (MS/MS) immunopeptidomics data; 3) Verify discovered peptide candidates through a secondary peptide-centric evaluation method against the MS/MS dataset; 4) Visualize and classify the nature of verified neoantigen peptides encoded by the genome and/or transcriptome; 5) Prioritize neoantigens for further exploration and empirical validation. Conclusions We demonstrate the effectiveness of the iPepGen pipeline for candidate neoantigen discovery and characterization. With tools, workflows, and training resources available in the open Galaxy ecosystem, iPepGen should provide cancer researchers with a flexible and accessible informatics resource tailored to accelerating immuno-oncology studies.
Collapse
|
15
|
Kerr SM, Klaric L, Muckian MD, Johnston K, Drake C, Halachev M, Cowan E, Snadden L, Dean J, Zheng SL, Thami PK, Ware JS, Tzoneva G, Shuldiner AR, Miedzybrodzka Z, Wilson JF. Actionable genetic variants in 4,198 Scottish participants from the Orkney and Shetland founder populations and implementation of return of results. Am J Hum Genet 2025; 112:793-807. [PMID: 40088892 DOI: 10.1016/j.ajhg.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
The benefits of returning clinically actionable genetic results to participants in research cohorts are accruing, yet such a genome-first approach is challenging. Here, we describe the implementation of return of such results in two founder populations from Scotland. Between 2005 and 2015, we recruited >4,000 adults with grandparents from Orkney and Shetland into the Viking Genes research cohort. The return of genetic data was not offered at baseline, but in 2023, we sent invitations to participants for consent to return of actionable genetic findings. We generated exome sequence data from 4,198 participants and used the American College of Medical Genetics and Genomics (ACMG) v.3.2 list of 81 genes, ClinVar review, and pathogenicity status, plus manual curation, to develop a pipeline to identify potentially actionable variants. We identified 104 individuals (2.5%) with 108 actionable genotypes at 39 variants in 23 genes and validated these. Working with the NHS Clinical Genetics service, which provided genetic counseling and clinical verification of the research results, and after expert clinical review, we notified 64 consenting participants (or their next of kin) of their actionable genotypes. Ten actionable variants across seven genes (BRCA1, BRCA2, ATP7B, TTN, KCNH2, MUTYH, and GAA) have risen 50- to >3,000-fold in frequency through genetic drift in ancestral island localities. Viking Genes is one of the first UK research cohorts to return actionable findings, providing an ethical and logistical exemplar of return of results. The genetic structure in the Northern Isles of Scotland with multiple founder effects provides a unique opportunity for a tailored approach to disease prevention through genetic screening.
Collapse
Affiliation(s)
- Shona M Kerr
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Lucija Klaric
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Marisa D Muckian
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, UK; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Kiera Johnston
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, UK
| | - Camilla Drake
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Mihail Halachev
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Emma Cowan
- Department of Medical Genetics, Ashgrove House, NHS Grampian, Aberdeen AB25 2ZA, UK; Medical Genetics Group, University of Aberdeen, Polwarth Building, Aberdeen AB25 2ZD, UK
| | - Lesley Snadden
- Department of Medical Genetics, Ashgrove House, NHS Grampian, Aberdeen AB25 2ZA, UK; Medical Genetics Group, University of Aberdeen, Polwarth Building, Aberdeen AB25 2ZD, UK
| | - John Dean
- Medical Genetics Group, University of Aberdeen, Polwarth Building, Aberdeen AB25 2ZD, UK
| | - Sean L Zheng
- National Heart and Lung Institute, Imperial College London, London, UK; MRC Laboratory of Medical Sciences, Imperial College London, London, UK; Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Prisca K Thami
- National Heart and Lung Institute, Imperial College London, London, UK; MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK; MRC Laboratory of Medical Sciences, Imperial College London, London, UK; Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | | | | | - Zosia Miedzybrodzka
- Department of Medical Genetics, Ashgrove House, NHS Grampian, Aberdeen AB25 2ZA, UK; Medical Genetics Group, University of Aberdeen, Polwarth Building, Aberdeen AB25 2ZD, UK
| | - James F Wilson
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK; Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, UK; Centre for Genomic and Experimental Medicine, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
16
|
Novák J, Takács T, Tilajka Á, László L, Oravecz O, Farkas E, Than NG, Buday L, Balogh A, Vas V. The sweet and the bitter sides of galectin-1 in immunity: its role in immune cell functions, apoptosis, and immunotherapies for cancer with a focus on T cells. Semin Immunopathol 2025; 47:24. [PMID: 40178639 PMCID: PMC11968517 DOI: 10.1007/s00281-025-01047-8] [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: 07/11/2024] [Accepted: 02/07/2025] [Indexed: 04/05/2025]
Abstract
Galectin-1 (Gal-1), a member of the β-galactoside-binding soluble lectin family, is a double-edged sword in immunity. On one hand, it plays a crucial role in regulating diverse immune cell functions, including the apoptosis of activated T cells. These processes are key in resolving inflammation and preventing autoimmune diseases. On the other hand, Gal-1 has significant implications in cancer, where tumor cells and the tumor microenvironment (TME) (e.g., tumor-associated fibroblasts, myeloid-derived suppressor cells) secrete Gal-1 to evade immune surveillance and promote cancer cell growth. Within the TME, Gal-1 enhances the differentiation of tolerogenic dendritic cells, induces the apoptosis of effector T cells, and enhances the proliferation of regulatory T cells, collectively facilitating tumor immune escape. Therefore, targeting Gal-1 holds the potential to boost anti-tumor immunity and improve the efficacy of cancer immunotherapy. This review provides insights into the intricate role of Gal-1 in immune cell regulation, with an emphasis on T cells, and elucidates how tumors exploit Gal-1 for immune evasion and growth. Furthermore, we discuss the potential of Gal-1 as a therapeutic target to augment current immunotherapies across various cancer types.
Collapse
Affiliation(s)
- Julianna Novák
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Tamás Takács
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Álmos Tilajka
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Loretta László
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Orsolya Oravecz
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, 1117, Hungary
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Emese Farkas
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Károly Rácz Conservative Medicine Division, Doctoral College, Semmelweis University, Budapest, 1091, Hungary
| | - Nándor Gábor Than
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, 1088, Hungary
| | - László Buday
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Department of Molecular Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
| | - Virág Vas
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
| |
Collapse
|
17
|
Huang T, Radley A, Yanagida A, Ren Z, Carlisle F, Tahajjodi S, Kim D, O'Neill P, Clarke J, Lancaster MA, Heckhausen Z, Zhuo J, de Sousa JPA, Hajkova P, von Meyenn F, Imai H, Nakauchi H, Guo G, Smith A, Masaki H. Inhibition of PRC2 enables self-renewal of blastoid-competent naive pluripotent stem cells from chimpanzee. Cell Stem Cell 2025; 32:627-639.e8. [PMID: 40015279 DOI: 10.1016/j.stem.2025.02.002] [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: 03/11/2024] [Revised: 10/11/2024] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Naive pluripotent stem cells (PSCs) are counterparts of early epiblast in the mammalian embryo. Mouse and human naive PSCs differ in self-renewal requirements and extraembryonic lineage potency. Here, we investigated the generation of chimpanzee naive PSCs. Colonies generated by resetting or reprogramming failed to propagate. We discovered that self-renewal is enabled by inhibition of Polycomb repressive complex 2 (PRC2). Expanded cells show global transcriptome proximity to human naive PSCs and embryo pre-implantation epiblast, with shared expression of a subset of pluripotency transcription factors. Chimpanzee naive PSCs can transition to multilineage competence or can differentiate into trophectoderm and hypoblast, forming tri-lineage blastoids. They thus provide a higher primate comparative model for studying pluripotency and early embryogenesis. Genetic deletions confirm that PRC2 mediates growth arrest. Further, inhibition of PRC2 overcomes a roadblock to feeder-free propagation of human naive PSCs. Therefore, excess deposition of chromatin modification H3K27me3 is an unexpected barrier to naive PSC self-renewal.
Collapse
Affiliation(s)
- Tao Huang
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Arthur Radley
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Ayaka Yanagida
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo 113-8657, Japan; Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Zhili Ren
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | | | | | - Dongwan Kim
- Stem Cell Therapy Division, Institute of Integrated Research, Institute of Science, Tokyo 113-8510, Japan
| | - Paul O'Neill
- University of Exeter Sequencing Facility, University of Exeter, Exeter EX4 4QD, UK
| | - James Clarke
- Wellcome-MRC Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Zoe Heckhausen
- MRC Laboratory of Medical Sciences (LMS), Du Cane Rd, London W12 0HS, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, W12 0NN, UK
| | - Jingran Zhuo
- Department of Health Sciences and Technology, ETH Zurich, 8603 Schwerzenbach, Switzerland
| | | | - Petra Hajkova
- MRC Laboratory of Medical Sciences (LMS), Du Cane Rd, London W12 0HS, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, W12 0NN, UK
| | - Ferdinand von Meyenn
- Department of Health Sciences and Technology, ETH Zurich, 8603 Schwerzenbach, Switzerland
| | - Hiroo Imai
- Department of Cellular and Molecular Biology, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; Stem Cell Therapy Division, Institute of Integrated Research, Institute of Science, Tokyo 113-8510, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ge Guo
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Austin Smith
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK.
| | - Hideki Masaki
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; Stem Cell Therapy Division, Institute of Integrated Research, Institute of Science, Tokyo 113-8510, Japan.
| |
Collapse
|
18
|
Layne TM, Rothstein JH, Song X, Andersen SW, Benn EKT, Sieh W, Klein RJ. Vitamin D-related genetic variants and prostate cancer risk in Black men. Cancer Epidemiol 2025; 95:102742. [PMID: 39823710 DOI: 10.1016/j.canep.2025.102742] [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: 06/25/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
BACKGROUND The relationship between vitamin D and prostate cancer has primarily been characterized among White men. Black men, however, have higher prostate cancer incidence and mortality rates, chronically low circulating vitamin D levels, and ancestry-specific genetic variants in vitamin D-related genes. Here, we examine critical genes in the vitamin D pathway and prostate cancer risk in Black men. METHODS We assessed a total of 73 candidate variants in genes (namely GC, CYP27A1, CYP27B1, CYP24A1, VDR, and RXRA) including functional variants previously associated with prostate cancer and circulating 25(OHD) in White men. Associations with prostate cancer risk were examined using genome-wide association study data for approximately 10,000 prostate cancer cases and 10,000 controls among Black men and over 85,000 cases and 91,000 controls among White men for comparison. A statistical significance threshold of 0.000685 was used to account for the 73 variants tested. RESULTS None of the variants examined were significantly associated with prostate cancer risk among Black men after multiple comparison adjustment. Suggestive associations (P < 0.05) for four variants were found in Black men, including two in RXRA (rs41400444 OR=1.09, 95 % CI: 1.01-1.17, P = 0.024 and rs10881574 OR = 0.93, 0.87-1.00, P = 0.046) and two in VDR (rs2853563 OR = 1.07, 1.01-1.13, P = 0.017 and rs1156882 OR = 1.06, 1.00-1.12, P = 0.045). Two variants in VDR were also positively associated with risk in White men (rs11568820 OR = 1.04, 1.02-1.06, P = 0.00024 and rs4516035 OR = 1.03, 1.01-1.04, P = 0.00055). CONCLUSION We observed suggestive associations between genetic variants in RXRA and VDR and prostate cancer risk in Black men. Future research exploring the relationship of vitamin D with cancer risk in Black men will need larger sample sizes to identify ancestry-specific variants relevant to risk in this population.
Collapse
Affiliation(s)
- Tracy M Layne
- Center for Scientific Diversity and Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Joseph H Rothstein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xiaoyu Song
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
| | - Shaneda Warren Andersen
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Emma K T Benn
- Center for Scientific Diversity and Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Weiva Sieh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
19
|
Liu Y, Dong Y, Jiang Y, Han S, Liu X, Xu X, Zhu A, Zhao Z, Gao Y, Zou Y, Zhang C, Bian Y, Zhang Y, Liu J, Zhao S, Zhao H, Chen ZJ. Caloric restriction prevents inheritance of polycystic ovary syndrome through oocyte-mediated DNA methylation reprogramming. Cell Metab 2025; 37:920-935.e6. [PMID: 39986273 DOI: 10.1016/j.cmet.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/17/2024] [Accepted: 01/15/2025] [Indexed: 02/24/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent metabolic and reproductive endocrine disorder with strong heritability. However, the independent role of oocytes in mediating this heritability remains unclear. Utilizing in vitro fertilization-embryo transfer and surrogacy, we demonstrated that oocytes from androgen-exposed mice (F1) transmitted PCOS-like traits to F2 and F3 generations. Notably, caloric restriction (CR) in F1 or F2 effectively prevented this transmission by restoring disrupted DNA methylation in oocyte genes related to insulin secretion and AMPK signaling pathways. Further detection in adult tissues of offspring revealed dysregulated DNA methylation and expression of those genes (e.g., Adcy3, Gnas, and Srebf1) were reversed by maternal CR. Moreover, similar benefits of CR were observed in aberrant embryonic methylome of women with PCOS. These findings elucidate the essential role of CR in preventing PCOS transmission via methylation reprogramming, emphasizing the importance of preconception metabolic management for women with PCOS.
Collapse
Affiliation(s)
- Yue Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China; Center for Reproductive Medicine, Gusu School, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital, Nanjing 212028, Jiangsu, China
| | - Yi Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yonghui Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Shan Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Xin Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Xin Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Aiqing Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Zihe Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yuan Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yang Zou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Chuanxin Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yuehong Bian
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Yuqing Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China
| | - Jiang Liu
- Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Shigang Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China; Shandong Technology Innovation Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan 250012, Shandong, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan 250012, Shandong, China.
| | - Han Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China; Center for Reproductive Medicine, Gusu School, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital, Nanjing 212028, Jiangsu, China; Shandong Technology Innovation Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan 250012, Shandong, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan 250012, Shandong, China.
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, The Second Hospital, Institute of Women, Children and Reproductive Health, Shandong University, Jinan 250012, Shandong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan 250012, Shandong, China; Shandong Technology Innovation Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan 250012, Shandong, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan 250012, Shandong, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan 250012, Shandong, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200025, China; Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
20
|
Martin-Gonzalez E, Perez-Garcia J, Herrera-Luis E, Martin-Almeida M, Kebede-Merid S, Hernandez-Pacheco N, Lorenzo-Diaz F, González-Pérez R, Sardón O, Hernández-Pérez JM, Poza-Guedes P, Sánchez-Machín I, Mederos-Luis E, Corcuera P, López-Fernández L, Román-Bernal B, Toncheva AA, Harner S, Wolff C, Brandstetter S, Abdel-Aziz MI, Hashimoto S, Vijverberg SJH, Kraneveld AD, Potočnik U, Kabesch M, Maitland-van der Zee AH, Villar J, Melén E, Pino-Yanes M. Epigenome-Wide Association Study of Asthma Exacerbations in Europeans. Allergy 2025; 80:1086-1099. [PMID: 39907155 DOI: 10.1111/all.16490] [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: 07/10/2024] [Revised: 12/03/2024] [Accepted: 01/01/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Asthma exacerbations (AEs) represent the major contributor to the global asthma burden. Although genetic and environmental factors have been associated with AEs, the role of epigenetics remains uncovered. OBJECTIVE This study aimed to identify whole blood DNA methylation (DNAm) markers associated with AEs in Europeans. METHODS DNAm was assessed in 406 blood samples from Spanish individuals using the Infinium MethylationEPIC microarray (Illumina). An epigenome-wide association study was conducted to test the association of DNAm with AEs at differentially methylated positions, regions, and epigenetic modules. CpGs suggestively associated with AEs (false discovery rate [FDR] < 0.1) were followed up for replication in 222 European individuals, and the genome-wide significance (p < 9 × 10-8) was declared after meta-analyzing the discovery and replication samples. Additional assessment was performed using nasal tissue DNAm data from 155 Spanish individuals. The effects of genetic variation on DNAm were assessed through cis-methylation quantitative trait loci (meQTL) analysis. Enrichment analyses of previous EWAS signals were conducted. RESULTS Four CpGs were associated with AEs, and two were replicated and reached genomic significance in the meta-analysis (annotated to ZBTB16 and BAIAP2). Of those, CpG cg25345365 (ZBTB16) was cross-tissue validated in nasal epithelium (p= 0.003) and associated with five independent meQTLs (FDR < 0.05). Additionally, four differentially methylated regions and one module were significantly associated with AEs. Enrichment analyses revealed an overrepresentation of prior epigenetic associations with prenatal and environmental exposures, immune-mediated diseases, and mortality. CONCLUSIONS DNAm in whole blood and nasal samples may contribute to AEs in Europeans, capturing genetic and environmental risk factors.
Collapse
Affiliation(s)
- Elena Martin-Gonzalez
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
| | - Javier Perez-Garcia
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
| | - Esther Herrera-Luis
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mario Martin-Almeida
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
| | - Simon Kebede-Merid
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Natalia Hernandez-Pacheco
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Fabian Lorenzo-Diaz
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna (ULL), La Laguna, Spain
| | - Ruperto González-Pérez
- Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
- Severe Asthma Unit, Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
| | - Olaia Sardón
- Division of Pediatric Respiratory Medicine, Hospital Universitario Donostia, San Sebastián, Spain
- Department of Pediatrics, University of the Basque Country (UPV/EHU), San Sebastián, Spain
| | - José M Hernández-Pérez
- Department of Respiratory Medicine, Hospital Universitario de N.S de Candelaria, Santa Cruz de Tenerife, Spain
- Respiratory Medicine, Hospital Universitario de La Palma, Santa Cruz de Tenerife, Spain
| | - Paloma Poza-Guedes
- Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
- Severe Asthma Unit, Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
| | | | - Elena Mederos-Luis
- Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
| | - Paula Corcuera
- Division of Pediatric Respiratory Medicine, Hospital Universitario Donostia, San Sebastián, Spain
| | - Leyre López-Fernández
- Division of Pediatric Respiratory Medicine, Hospital Universitario Donostia, San Sebastián, Spain
| | | | - Antoaneta A Toncheva
- University Children's Hospital Regensburg (KUNO), Hospital St. Hedwig of the Order of St. John, University of Regensburg, Regensburg, Germany
| | - Susanne Harner
- University Children's Hospital Regensburg (KUNO), Hospital St. Hedwig of the Order of St. John, University of Regensburg, Regensburg, Germany
| | - Christine Wolff
- University Children's Hospital Regensburg (KUNO), Hospital St. Hedwig of the Order of St. John, University of Regensburg, Regensburg, Germany
| | - Susanne Brandstetter
- University Children's Hospital Regensburg (KUNO), Hospital St. Hedwig of the Order of St. John, University of Regensburg, Regensburg, Germany
| | - Mahmoud Ibrahim Abdel-Aziz
- Pulmonary Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Simone Hashimoto
- Pulmonary Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands
- Pediatric Pulmonology, Emma's Childrens Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Susanne J H Vijverberg
- Pulmonary Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Uroš Potočnik
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
- Department for Science and Research, University Medical Centre Maribor, Maribor, Slovenia
| | - Michael Kabesch
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
- Research and Development Campus Regensburg (WECARE) at the Hospital St. Hedwig of the Order of St. John, Regensburg, Germany
| | - Anke H Maitland-van der Zee
- Pulmonary Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands
- Pediatric Pulmonology, Emma's Childrens Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jesús Villar
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Dr. Negrín, Fundación Canaria Instituto de Investigación Sanitaria de Canarias, Las Palmas de Gran Canaria, Spain
- Faculty of Health Sciences, Universidad del Atlántico Medio, Las Palmas, Spain
- Li Ka Shing Knowledge Institute at St Michael's Hospital, Toronto, Canada
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), San Cristóbal de La Laguna, Spain
| |
Collapse
|
21
|
Patsakis M, Provatas K, Baltoumas FA, Chantzi N, Mouratidis I, Pavlopoulos GA, Georgakopoulos-Soares I. MAFin: motif detection in multiple alignment files. Bioinformatics 2025; 41:btaf125. [PMID: 40106711 PMCID: PMC11978385 DOI: 10.1093/bioinformatics/btaf125] [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: 10/11/2024] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Abstract
MOTIVATION Whole Genome and Proteome Alignments, represented by the multiple alignment file format, have become a standard approach in comparative genomics and proteomics. These often require identifying conserved motifs, which is crucial for understanding functional and evolutionary relationships. However, current approaches lack a direct method for motif detection within MAF files. We present MAFin, a novel tool that enables efficient motif detection and conservation analysis in MAF files to address this gap, streamlining genomic and proteomic research. RESULTS We developed MAFin, the first motif detection tool for Multiple Alignment Format files. MAFin enables the multithreaded search of conserved motifs using three approaches: (i) using user-specified k-mers to search the sequences. (ii) with regular expressions, in which case one or more patterns are searched, and (iii) with predefined Position Weight Matrices. Once the motif has been found, MAFin detects the motif instances and calculates the conservation across the aligned sequences. MAFin also calculates a conservation percentage, which provides information about the conservation levels of each motif across the aligned sequences, based on the number of matches relative to the length of the motif. A set of statistics enables the interpretation of each motif's conservation level, and the detected motifs are exported in JSON and CSV files for downstream analyses. AVAILABILITY AND IMPLEMENTATION MAFin is offered as a Python package under the GPL license as a multi-platform application and is available at: https://github.com/Georgakopoulos-Soares-lab/MAFin.
Collapse
Affiliation(s)
- Michail Patsakis
- Institute for Personalized Medicine, Department of Molecular and Precision Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, United States
| | - Kimonas Provatas
- Institute for Personalized Medicine, Department of Molecular and Precision Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, United States
- Division of Basic Sciences, University of Crete Medical School, Heraklion 71110, Greece
| | - Fotis A Baltoumas
- Institute for Fundamental Biomedical Research, BSRC “Alexander Fleming”, Vari 16672, Greece
| | - Nikol Chantzi
- Institute for Personalized Medicine, Department of Molecular and Precision Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, United States
| | - Ioannis Mouratidis
- Institute for Personalized Medicine, Department of Molecular and Precision Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, United States
| | - Georgios A Pavlopoulos
- Institute for Fundamental Biomedical Research, BSRC “Alexander Fleming”, Vari 16672, Greece
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Molecular and Precision Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, United States
| |
Collapse
|
22
|
Schaefer NK, Pavlovic BJ, Pollen AA. CellBouncer, A Unified Toolkit for Single-Cell Demultiplexing and Ambient RNA Analysis, Reveals Hominid Mitochondrial Incompatibilities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.23.644821. [PMID: 40166335 PMCID: PMC11957168 DOI: 10.1101/2025.03.23.644821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Pooled processing, in which cells from multiple sources are cultured or captured together, is an increasingly popular strategy for droplet-based single cell sequencing studies. This design allows efficient scaling of experiments, isolation of cell-intrinsic differences, and mitigation of batch effects. We present CellBouncer, a computational toolkit for demultiplexing and analyzing single-cell sequencing data from pooled experiments. We demonstrate that CellBouncer can separate and quantify multi-species and multi-individual cell mixtures, identify unknown mitochondrial haplotypes in cells, assign treatments from lipid-conjugated barcodes or CRISPR sgRNAs, and infer pool composition, outperforming existing methods. We also introduce methods to quantify ambient RNA contamination per cell, infer individual donors' contributions to the ambient RNA pool, and determine a consensus doublet rate harmonized across data types. Applying these tools to tetraploid composite cells, we identify a competitive advantage of human over chimpanzee mitochondria across 10 cell fusion lines and provide evidence for inter-mitochondrial incompatibility and mito-nuclear incompatibility between species.
Collapse
Affiliation(s)
- Nathan K Schaefer
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Bryan J Pavlovic
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Alex A Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
23
|
Minkin I, Salzberg SL. Conservation assessment of human splice site annotation based on a 470-genome alignment. Nucleic Acids Res 2025; 53:gkaf184. [PMID: 40119728 PMCID: PMC11928937 DOI: 10.1093/nar/gkaf184] [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: 05/16/2024] [Accepted: 02/24/2025] [Indexed: 03/24/2025] Open
Abstract
Despite many improvements over the years, the annotation of the human genome remains imperfect. The use of evolutionarily conserved sequences provides a strategy for selecting a high-confidence subset of the annotation. Using the latest whole-genome alignment, we found that splice sites from protein-coding genes in the high-quality MANE annotation are consistently conserved across >350 species. We also studied splice sites from the RefSeq, GENCODE, and CHESS databases not present in MANE. In addition, we analyzed the completeness of the alignment with respect to the human genome annotations and described a method that would allow us to fix up to 60% of the missing alignments of the protein-coding exons. We trained a logistic regression classifier to distinguish between the conservation exhibited by sites from MANE versus sites chosen randomly from neutrally evolving sequences. We found that splice sites classified by our model as well-supported have lower single nucleotide polymorphism rates and better transcriptomic evidence. We then computed a subset of transcripts using only "well-supported" splice sites or ones from MANE. This subset is enriched in high-confidence transcripts of the major gene catalogs that appear to be under purifying selection and are more likely to be correct and functionally relevant.
Collapse
Affiliation(s)
- Ilia Minkin
- Department of Biomedical Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, United States
- Center for Computational Biology, Johns Hopkins University, 3100 Wyman Park Drive, Baltimore, MD 21211, United States
| | - Steven L Salzberg
- Department of Biomedical Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, United States
- Center for Computational Biology, Johns Hopkins University, 3100 Wyman Park Drive, Baltimore, MD 21211, United States
- Department of Computer Science, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, United States
- Department of Biostatistics, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, United States
| |
Collapse
|
24
|
Auxillos J, Stigliani A, Vaagensø C, Garland W, Niazi A, Valen E, Jensen T, Sandelin A. True length of diverse capped RNA sequencing (TLDR-seq): 5'-3'-end sequencing of capped RNAs regardless of 3'-end status. Nucleic Acids Res 2025; 53:gkaf240. [PMID: 40183637 PMCID: PMC11969664 DOI: 10.1093/nar/gkaf240] [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: 10/22/2024] [Revised: 02/20/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Analysis of transcript function is greatly aided by knowledge of the full-length RNA sequence. New long-read sequencing enabled by Oxford Nanopore and PacBio devices have the potential to provide full-length transcript information; however, standard methods still lack the ability to capture true RNA 5' ends and select for polyadenylated (pA+) transcripts only. Here, we present a method that, by utilizing cap trapping and 3'-end adapter ligation, sequences transcripts between their exact 5' and 3' ends regardless of polyadenylation status and without the need for ribosomal RNA depletion, with the ability to characterize polyadenylation length of RNAs, if any. The method shows high reproducibility, can faithfully detect 5' ends, 3' ends and splice junctions, and produces gene-expression estimates that are highly correlated to those of short-read sequencing techniques. We also demonstrate that the method can detect and sequence full-length nonadenylated (pA-) RNAs, including long noncoding RNAs, promoter upstream transcripts, and enhancer RNAs, and present cases where pA+ and pA- RNAs show preferences for different but closely located transcription start sites. Our method is therefore useful for the characterization of diverse capped RNA species and analysis of relationships between transcription initiation, termination, and RNA processing.
Collapse
Affiliation(s)
- Jamie Auxillos
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, DK2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, DK2200 Copenhagen, Denmark
| | - Arnaud Stigliani
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, DK2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, DK2200 Copenhagen, Denmark
| | - Christian Skov Vaagensø
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, DK2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, DK2200 Copenhagen, Denmark
| | - William Garland
- Department of Molecular Biology and Genetics, Aarhus University, DK8000 Aarhus, Denmark
| | - Adnan Muhammed Niazi
- Computational Biology Unit, Department of Informatics, University of Bergen, N-5008 Bergen, Norway
| | - Eivind Valen
- Computational Biology Unit, Department of Informatics, University of Bergen, N-5008 Bergen, Norway
- Department of Biosciences, University of Oslo, N-0371 Oslo, Norway
| | - Torben Heick Jensen
- Department of Molecular Biology and Genetics, Aarhus University, DK8000 Aarhus, Denmark
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, DK2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, DK2200 Copenhagen, Denmark
| |
Collapse
|
25
|
Wilson PD, Yu X, Handelmann CR, Buck MJ. Nucleosome binding by TP53, TP63, and TP73 is determined by the composition, accessibility, and helical orientation of their binding sites. Genome Res 2025; 35:404-416. [PMID: 39929723 PMCID: PMC11960462 DOI: 10.1101/gr.279541.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 02/03/2025] [Indexed: 02/19/2025]
Abstract
The TP53 family of transcription factors plays key roles in driving development and combating cancer by regulating gene expression. TP53, TP63, and TP73-the three members of the TP53 family-regulate gene expression by binding to their DNA binding sites, many of which are situated within nucleosomes. To thoroughly examine the nucleosome-binding abilities of the TP53 family, we used Pioneer-seq, a technique that assesses a transcription factor's binding affinity to its DNA-binding sites at all possible positions within the nucleosome core particle. Using Pioneer-seq, we analyzed the binding affinities of TP53, TP63, and TP73 to 10 TP53 family binding sites across the nucleosome core particle. We find that the affinities of TP53, TP63, and TP73 for nucleosomes are primarily determined by the positioning of TP53 family binding sites within nucleosomes; TP53 family members bind strongly to the more accessible edges of nucleosomes but weakly to the less accessible centers of nucleosomes. Our results further show that the DNA-helical orientation of TP53 family binding sites within nucleosomal DNA impacts the nucleosome-binding affinities of TP53 family members, with binding-site composition impacting the affinity of each TP53 family member only when the binding-site location is accessible. Taken together, our results show that the accessibility, composition, and helical orientation of TP53 family binding sites collectively determine the nucleosome-binding affinities of TP53, TP63, and TP73. These findings help explain the rules underlying TP53 family-nucleosome binding and thus provide requisite insight into how we may better control gene expression changes involved in development and tumor suppression.
Collapse
Affiliation(s)
- Patrick D Wilson
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
| | - Xinyang Yu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
| | - Christopher R Handelmann
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
| | - Michael J Buck
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA;
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
| |
Collapse
|
26
|
Ishigohoka J, Liedvogel M. High-recombining genomic regions affect demography inference based on ancestral recombination graphs. Genetics 2025; 229:iyaf004. [PMID: 39790013 PMCID: PMC11912872 DOI: 10.1093/genetics/iyaf004] [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: 02/05/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
Multiple methods of demography inference are based on the ancestral recombination graph. This powerful approach uses observed mutations to model local genealogies changing along chromosomes by historical recombination events. However, inference of underlying genealogies is difficult in regions with high recombination rate relative to mutation rate due to the lack of mutations representing genealogies. Despite the prevalence of high-recombining genomic regions in some organisms, such as birds, its impact on demography inference based on ancestral recombination graphs has not been well studied. Here, we use population genomic simulations to investigate the impact of high-recombining regions on demography inference based on ancestral recombination graphs. We demonstrate that inference of effective population size and the time of population split events is systematically affected when high-recombining regions cover wide breadths of the chromosomes. Excluding high-recombining genomic regions can practically mitigate this impact, and population genomic inference of recombination maps is informative in defining such regions although the estimated values of local recombination rate can be biased. Finally, we confirm the relevance of our findings in empirical analysis by contrasting demography inferences applied for a bird species, the Eurasian blackcap (Sylvia atricapilla), using different parts of the genome with high and low recombination rates. Our results suggest that demography inference methods based on ancestral recombination graphs should be carried out with caution when applied in species whose genomes contain long stretches of high-recombining regions.
Collapse
Affiliation(s)
- Jun Ishigohoka
- Max Planck Research Group Behavioural Genomics, Max Planck Institute for Evolutionary Biology, August-Thienemann-Straße 2, Plön 24306, Germany
| | - Miriam Liedvogel
- Max Planck Research Group Behavioural Genomics, Max Planck Institute for Evolutionary Biology, August-Thienemann-Straße 2, Plön 24306, Germany
- Institute of Avian Research, An der Vogelwarte 21, Wilhelmshaven 26386, Germany
- Department of Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, Oldenburg 26129, Germany
| |
Collapse
|
27
|
Gohl P, Oliva B. SNPeBoT: a tool for predicting transcription factor allele specific binding. BMC Bioinformatics 2025; 26:81. [PMID: 40065237 PMCID: PMC11895208 DOI: 10.1186/s12859-025-06094-4] [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: 12/24/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Mutations in non-coding regulatory regions of DNA may lead to disease through the disruption of transcription factor binding. However, our understanding of binding patterns of transcription factors and the effects that changes to their binding sites have on their action remains limited. To address this issue we trained a Deep learning model to predict the effects of Single Nucleotide Polymorphisms (SNP) on transcription factor binding. Allele specific binding (ASB) data from Chromatin Immunoprecipitation sequencing (ChIP-seq) experiments were paired with high sequence-identity DNA binding Domains assessed in Protein Binding Microarray (PBM) experiments. For each transcription factor a paired DNA binding Domain was selected from which we derived E-score profiles for reference and alternate DNA sequences of ASB events. A Convolutional Neural Network (CNN) was trained to predict whether these profiles were indicative of ASB gain/loss or no change in binding. 18211 E-score profiles from 113 transcription factors were split into train, validation and test data. We compared the performance of the trained model with other available platforms for predicting the effect of SNP on transcription factor binding. Our model demonstrated increased accuracy and ASB recall in comparison to the best scoring benchmark tools. CONCLUSION In this paper we present our model SNPeBoT (Single Nucleotide Polymorphism effect on Binding of Transcription Factors) in its standalone and web server form. The increased recovery and prediction accuracy of allele specific binding events could prove useful in discovering non-coding mutations relevant to disease.
Collapse
Affiliation(s)
- Patrick Gohl
- Department of Medicine and Life Sciences, SBI-GRIB, Universitat Pompeu Fabra, 08003, Barcelona, Catalonia, Spain
| | - Baldo Oliva
- Department of Medicine and Life Sciences, SBI-GRIB, Universitat Pompeu Fabra, 08003, Barcelona, Catalonia, Spain.
| |
Collapse
|
28
|
Zytnicki M. Assessing genome conservation on pangenome graphs with PanSel. BIOINFORMATICS ADVANCES 2025; 5:vbaf018. [PMID: 40092526 PMCID: PMC11908644 DOI: 10.1093/bioadv/vbaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/21/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025]
Abstract
Motivation With more and more telomere-to-telomere genomes assembled, pangenomes make it possible to capture the genomic diversity of a species. Because they introduce less biases, pangenomes, represented as graphs, tend to supplant the usual linear representation of a reference genome, augmented with variations. However, this major change requires new tools adapted to this data structure. Among the numerous questions that can be addressed to a pangenome graph is the search for conserved or divergent genes. Results In this article, we present a new tool, named PanSel, which computes a conservation score for each segment of the genome, and finds genomic regions that are significantly conserved, or divergent. PanSel can be used on prokaryotes and eukaryotes, with a sequence identity not less than 98%. Availability and implementation PanSel, written in C++11 with no dependency, is available at https://github.com/mzytnicki/pansel.
Collapse
Affiliation(s)
- Matthias Zytnicki
- Unité de Mathématiques et Informatique Appliquées, INRAE, 31 326 Castanet-Tolosan, France
| |
Collapse
|
29
|
Srivastava J, Ovcharenko I. Regulatory Plasticity of the Human Genome. Mol Biol Evol 2025; 42:msaf050. [PMID: 40056383 PMCID: PMC11934273 DOI: 10.1093/molbev/msaf050] [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: 07/31/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 03/10/2025] Open
Abstract
Evolutionary turnover in noncoding regions has driven phenotypic divergence during past speciation events and continues to facilitate environmental adaptation through variants. We used a deep learning model to identify the substrates of regulatory turnover using genome-wide mutations mimicking three evolutionary pathways: recent history (human-chimp substitutions), modern population (human population variation), and mutational susceptibility (random mutations). We observed enhancer turnover in approximately 6% of the whole genome, with more than 80% of the novel activity arising from repurposing of enhancers between cell types. Frequency of turnover in a cell type is remarkably similar across the three pathways, despite only ∼19% overlap in the source regions. The majority of turnover loci were found to be localized within 100 kb of a gene, with the highest turnover occurring near neurodevelopmental genes including CNTNAP2, NPAS3, and AUTS2. Flanking enhancers of these genes undergo high turnover irrespective of the mutational model pathway, suggesting a high plasticity in neurocognitive evolution. Based on susceptibility to random mutations, these enhancers were identified as vulnerable by nature and feature a higher abundance of cell type-specific transcription factor binding sites. Our findings suggest that enhancer repurposing within vulnerable loci drives regulatory innovation while keeping the core regulatory networks intact.
Collapse
Affiliation(s)
- Jaya Srivastava
- Division of Intramural Research, National Library of Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ivan Ovcharenko
- Division of Intramural Research, National Library of Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
30
|
Lim CS, Gibbon AK, Tran Nguyen AT, Chieng GSW, Brown CM. RIBOSS detects novel translational events by combining long- and short-read transcriptome and translatome profiling. Brief Bioinform 2025; 26:bbaf164. [PMID: 40221960 PMCID: PMC11994033 DOI: 10.1093/bib/bbaf164] [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: 11/08/2024] [Revised: 03/18/2025] [Accepted: 03/23/2025] [Indexed: 04/15/2025] Open
Abstract
Ribosome profiling is a high-throughput sequencing technique that captures the positions of translating ribosomes on RNAs. Recent advancements in ribosome profiling include achieving highly phased ribosome footprints for plant translatomes and more recently for bacterial translatomes. This substantially increases the specificity of detecting open reading frames (ORFs) that can be translated, such as small ORFs located upstream and downstream of the annotated ORFs. However, most genomes (e.g. bacterial genomes) lack the annotations for the transcription start and termination sites. This hinders the systematic discovery of novel ORFs in the 'untranslated' regions in ribosome profiling data. Here, we develop a new computational pipeline called RIBOSS to discover noncanonical ORFs and assess their translational potential against annotated ORFs. The RIBOSS Python modules are versatile, and we use them to analyse both prokaryotic and eukaryotic data. We present a resulting list of noncanonical ORFs with high translational potential in Homo sapiens, Arabidopsis thaliana, and Salmonella enterica. We further illustrate RIBOSS utility when studying organisms with incomplete transcriptome annotations. We leverage long-read and short-read data for reference-guided transcriptome assembly and highly phased ribosome profiling data for detecting novel translational events in the assembled transcriptome for S. enterica. In sum, RIBOSS is the first integrated computational pipeline for noncanonical ORF detection and translational potential assessment that incorporates long- and short-read sequencing technologies to investigate translation. RIBOSS is freely available at https://github.com/lcscs12345/riboss.
Collapse
Affiliation(s)
- Chun Shen Lim
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
- Genetics Otago, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
| | - Alexandra K Gibbon
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
- Genetics Otago, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
| | - Anh Thu Tran Nguyen
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
- Genetics Otago, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
| | - Gabrielle S W Chieng
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
- Genetics Otago, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
| | - Chris M Brown
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
- Genetics Otago, University of Otago, 710 Cumberland Street, Dunedin North, Dunedin 9016, New Zealand
| |
Collapse
|
31
|
Wang G, Zhang D, He Z, Mao B, Hu X, Chen L, Yang Q, Zhou Z, Zhang Y, Linghu K, Tang C, Xu Z, Liu D, Song J, Wang H, Lin Y, Li R, Lin JW, Chen L. Machine learning-based prediction reveals kinase MAP4K4 regulates neutrophil differentiation through phosphorylating apoptosis-related proteins. PLoS Comput Biol 2025; 21:e1012877. [PMID: 40096134 PMCID: PMC11957395 DOI: 10.1371/journal.pcbi.1012877] [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: 11/10/2024] [Revised: 03/31/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Neutrophils, an essential innate immune cell type with a short lifespan, rely on continuous replenishment from bone marrow (BM) precursors. Although it is established that neutrophils are derived from the granulocyte-macrophage progenitor (GMP), the molecular regulators involved in the differentiation process remain poorly understood. Here we developed a random forest-based machine-learning pipeline, NeuRGI (Neutrophil Regulatory Gene Identifier), which utilized Positive-Unlabeled Learning (PU-learning) and neural network-based in silico gene knockout to identify neutrophil regulators. We interrogated features including gene expression dynamics, physiological characteristics, pathological relatedness, and gene conservation for the model training. Our identified pipeline leads to identifying Mitogen-Activated Protein Kinase-4 (MAP4K4) as a novel neutrophil differentiation regulator. The loss of MAP4K4 in hematopoietic stem cells and progenitors in mice induced neutropenia and impeded the differentiation of neutrophils in the bone marrow. By modulating the phosphorylation level of proteins involved in cell apoptosis, such as STAT5A, MAP4K4 delicately regulates cell apoptosis during the process of neutrophil differentiation. Our work presents a novel regulatory mechanism in neutrophil differentiation and provides a robust prediction model that can be applied to other cellular differentiation processes.
Collapse
Affiliation(s)
- Guihua Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhifeng He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Mao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiao Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Chen
- Biosafety Laboratory of West China Hospital, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingxin Yang
- Biosafety Laboratory of West China Hospital, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yating Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kepan Linghu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao Tang
- Biosafety Laboratory of West China Hospital, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zijie Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Defu Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junwei Song
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huiying Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yishan Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruihan Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing-Wen Lin
- Biosafety Laboratory of West China Hospital, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
32
|
Edwards DL, Huang M, Wang TT. Soluble Factors and Mechanisms Regulated by Sialylated IgG Signaling. Immunol Rev 2025; 330:e70021. [PMID: 40084926 PMCID: PMC12042769 DOI: 10.1111/imr.70021] [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: 11/01/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Inflammation is a complex biological response that can be both induced and actively suppressed by IgG-Fc gamma receptor (FcγR) interactions. This review explores the role of IgG sialylation in reducing or blocking inflammatory responses. We first revisit foundational studies that established the anti-inflammatory properties of sialylated IgG1 Fc. These early investigations revealed that the sialylated fraction is crucial for intravenous immunoglobulin's (IVIg's) ability to reduce inflammation in many autoinflammatory diseases and defined a paracrine signaling mechanism underlying this activity. Next, we discuss a recently identified mechanism whereby sialylated IgG directly induces RE1-Silencing Transcription Factor (REST) which functions as a transcriptional repressor of NF-κB1. This mechanism suggests a very broad role for sialylated IgG signaling in inflammation control since NF-κB is a central mediator of responses downstream of diverse activating receptors on both adaptive and innate immune cells. Finally, we review a set of soluble factors that are suppressed by sialylated IgG signaling in the murine airway and in purified human macrophages, providing additional insight into mechanisms by which sialylated IgG contributes to broad inflammatory control.
Collapse
Affiliation(s)
- Desmond L. Edwards
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | - Min Huang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | - Taia T. Wang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305
- Department of Medicine, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
33
|
MacNish TR, Al‐Mamun HA, Bayer PE, McPhan C, Fernandez CGT, Upadhyaya SR, Liu S, Batley J, Parkin IAP, Sharpe AG, Edwards D. Brassica Panache: A multi-species graph pangenome representing presence absence variation across forty-one Brassica genomes. THE PLANT GENOME 2025; 18:e20535. [PMID: 39648684 PMCID: PMC11730171 DOI: 10.1002/tpg2.20535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/20/2024] [Accepted: 11/01/2024] [Indexed: 12/10/2024]
Abstract
Brassicas are an economically important crop species that provide a source of healthy oil and vegetables. With the rising population and the impact of climate change on agriculture, there is an increasing need to improve agronomically important traits of crops such as Brassica. The genomes of plant species have significant sequence presence absence variation (PAV), which is a source of genetic variation that can be used for crop improvement, and this species variation can be captured through the construction of pangenomes. Graph pangenomes are a recent reference format that represent the genomic variation with a species or population as alternate paths in a sequence graph. Graph pangenomes contain information on alignment, PAV, and annotation. Here we present the first multi-species graph pangenome for Brassica visualized with pangenome analyzer with chromosomal exploration (Panache).
Collapse
Affiliation(s)
- Tessa R. MacNish
- School of Biological SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
- Center for Applied BioinformaticsThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Hawlader A. Al‐Mamun
- School of Biological SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
- Center for Applied BioinformaticsThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Philipp E. Bayer
- School of Biological SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
- Center for Applied BioinformaticsThe University of Western AustraliaPerthWestern AustraliaAustralia
- Minderoo FoundationPerthWestern AustraliaAustralia
| | - Connor McPhan
- School of Biological SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
- Center for Applied BioinformaticsThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Cassandria G. Tay Fernandez
- School of Biological SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
- Center for Applied BioinformaticsThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Shriprabha R. Upadhyaya
- School of Biological SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
- Center for Applied BioinformaticsThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Shengyi Liu
- Oil Crops Research Institute, CAASWuhanChina
| | - Jacqueline Batley
- School of Biological SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | | | | | - David Edwards
- School of Biological SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
- Center for Applied BioinformaticsThe University of Western AustraliaPerthWestern AustraliaAustralia
| |
Collapse
|
34
|
Lu W, Liu Y, Li J, Huang S, Wen Z, Su C, Lu Z, Mo Z, Yu Z. Single-cell assay for transposase-accessible chromatin sequencing of human clear cell renal cell carcinoma. Sci Data 2025; 12:334. [PMID: 40000710 PMCID: PMC11861977 DOI: 10.1038/s41597-025-04666-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Regulating the occurrence and progression of tumor cells at the epigenetic level is a new insight of clear cell renal cell carcinoma (ccRCC). Chromatin accessibility is an important pathway of epigenetic regulation, which may explain the mystery of tumor occurrence. Assay for transposase-accessible chromatin sequencing (ATAC-seq) provides insight into the epigenetic regulatory features of ccRCC, especially at the single-cell level. In this study, we performed scATAC-seq of 3 ccRCC samples and captured a total of 18,703 high-quality cell nuclei and 104,818 unique peaks. Our protocol for nuclear extraction was reliable and stable, which can be used to deal with fresh and frozen single-cell suspensions. We presented basic methods for scATAC-seq data analysis, such as cell clustering, gene activity scoring, cell subtype specific peaks, transcription factors, motif and motif footprinting analysis. Taken together, our data indicated the valuable epigenetic features of ccRCC, which will provide more references for the study of ccRCC.
Collapse
Affiliation(s)
- Wenhao Lu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yixuan Liu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiaping Li
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zheng Wen
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Cheng Su
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zheng Lu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Zengnan Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Zhenyuan Yu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
35
|
Miao Z, Yue JX. Interactive visualization and interpretation of pangenome graphs by linear reference-based coordinate projection and annotation integration. Genome Res 2025; 35:296-310. [PMID: 39805704 PMCID: PMC11874961 DOI: 10.1101/gr.279461.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
With the increasing availability of high-quality genome assemblies, pangenome graphs emerged as a new paradigm in the genomic field for identifying, encoding, and presenting genomic variation at both the population and species level. However, it remains challenging to truly dissect and interpret pangenome graphs via biologically informative visualization. To facilitate better exploration and understanding of pangenome graphs toward novel biological insights, here we present a web-based interactive visualization and interpretation framework for linear reference-projected pangenome graphs (VRPG). VRPG provides efficient and intuitive support for exploring and annotating pangenome graphs along a linear-genome-based coordinate system (e.g., that of a primary linear reference genome). Moreover, VRPG offers many unique features such as in-graph path highlighting for graph-constituent input assemblies, copy number characterization for graph-embedding nodes, and graph-based mapping for query sequences, all of which are highly valuable for researchers working with pangenome graphs. Additionally, VRPG enables side-by-side visualization between the graph-based pangenome representation and the conventional primary linear reference genome-based feature annotations, therefore seamlessly bridging the graph and linear genomic contexts. To further demonstrate its functionality and scalability, we applied VRPG to the cutting-edge yeast and human reference pangenome graphs derived from hundreds of high-quality genome assemblies via a dedicated web portal and examined their local genome diversity in the graph contexts.
Collapse
Affiliation(s)
- Zepu Miao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jia-Xing Yue
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
36
|
Negi S, Stenton SL, Berger SI, Canigiula P, McNulty B, Violich I, Gardner J, Hillaker T, O'Rourke SM, O'Leary MC, Carbonell E, Austin-Tse C, Lemire G, Serrano J, Mangilog B, VanNoy G, Kolmogorov M, Vilain E, O'Donnell-Luria A, Délot E, Miga KH, Monlong J, Paten B. Advancing long-read nanopore genome assembly and accurate variant calling for rare disease detection. Am J Hum Genet 2025; 112:428-449. [PMID: 39862869 PMCID: PMC11866955 DOI: 10.1016/j.ajhg.2025.01.002] [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: 08/21/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
More than 50% of families with suspected rare monogenic diseases remain unsolved after whole-genome analysis by short-read sequencing (SRS). Long-read sequencing (LRS) could help bridge this diagnostic gap by capturing variants inaccessible to SRS, facilitating long-range mapping and phasing and providing haplotype-resolved methylation profiling. To evaluate LRS's additional diagnostic yield, we sequenced a rare-disease cohort of 98 samples from 41 families, using nanopore sequencing, achieving per sample ∼36× average coverage and 32-kb read N50 from a single flow cell. Our Napu pipeline generated assemblies, phased variants, and methylation calls. LRS covered, on average, coding exons in ∼280 genes and ∼5 known Mendelian disease-associated genes that were not covered by SRS. In comparison to SRS, LRS detected additional rare, functionally annotated variants, including structural variants (SVs) and tandem repeats, and completely phased 87% of protein-coding genes. LRS detected additional de novo variants and could be used to distinguish postzygotic mosaic variants from prezygotic de novos. Diagnostic variants were established by LRS in 11 probands, with diverse underlying genetic causes including de novo and compound heterozygous variants, large-scale SVs, and epigenetic modifications. Our study demonstrates LRS's potential to enhance diagnostic yield for rare monogenic diseases, implying utility in future clinical genomics workflows.
Collapse
Affiliation(s)
- Shloka Negi
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Sarah L Stenton
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Seth I Berger
- Children's National Research Institute, Washington, DC, USA
| | | | - Brandy McNulty
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Ivo Violich
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Joshua Gardner
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Todd Hillaker
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Sara M O'Rourke
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Melanie C O'Leary
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elizabeth Carbonell
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christina Austin-Tse
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gabrielle Lemire
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jillian Serrano
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian Mangilog
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Grace VanNoy
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mikhail Kolmogorov
- Cancer Data Science Laboratory, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Eric Vilain
- Institute for Clinical and Translational Science, University of California, Irvine, Irvine, CA, USA
| | - Anne O'Donnell-Luria
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emmanuèle Délot
- Institute for Clinical and Translational Science, University of California, Irvine, Irvine, CA, USA
| | - Karen H Miga
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Jean Monlong
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.
| | - Benedict Paten
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
37
|
Law CT, Burns KH. Comparative Genomics Reveals LINE-1 Recombination with Diverse RNAs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.02.635956. [PMID: 39975348 PMCID: PMC11838501 DOI: 10.1101/2025.02.02.635956] [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: 02/21/2025]
Abstract
Long interspersed element-1 (LINE-1, L1) retrotransposons are the most abundant protein-coding transposable elements (TE) in mammalian genomes, and have shaped genome content over 170 million years of evolution. LINE-1 is self-propagating and mobilizes other sequences, including Alu elements. Occasionally, LINE-1 forms chimeric insertions with non-coding RNAs and mRNAs. U6 spliceosomal small nuclear RNA/LINE-1 chimeras are best known, though there are no comprehensive catalogs of LINE-1 chimeras. To address this, we developed TiMEstamp, a computational pipeline that leverages multiple sequence alignments (MSA) to estimate the age of LINE-1 insertions and identify candidate chimeric insertions where an adjacent sequence arrives contemporaneously. Candidates were refined by detecting hallmark features of L1 retrotransposition, such as target site duplication (TSD). Applying this pipeline to the human genome, we recovered all known species of LINE-1 chimeras and discovered new chimeric insertions involving small RNAs, Alu elements, and mRNA fragments. Some insertions are compatible with known mechanisms, such as RNA ligation. Other structures nominate novel mechanisms, such as trans-splicing. We also see evidence that LINE-1 loci with defunct promoters can acquire regulatory elements from nearby genes to restore retrotransposition activity. These discoveries highlight the recombinatory potential of LINE-1 RNA with implications for genome evolution and TE domestication.
Collapse
Affiliation(s)
- Cheuk-Ting Law
- Corresponding authors: Cheuk-Ting Law (), Kathleen H. Burns ()
| | | |
Collapse
|
38
|
Lori A, Patel AV, Westmaas JL, Diver WR. A novel smoking cessation behavior based on quit attempts may identify new genes associated with long-term abstinence. Addict Behav 2025; 161:108192. [PMID: 39504611 DOI: 10.1016/j.addbeh.2024.108192] [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: 04/19/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Smoking cessation at any age has been shown to improve quality of life, decrease illness, and reduce mortality. About half of smokers attempt to quit each year, but only ∼ 7 % maintain long-term abstinence unaided. Few genetic factors have been consistently associated with smoking cessation, possibly due to poor phenotype definition. METHODS We performed a genome-wide association study (GWAS) with an alternative phenotype based on the difficulty of quitting smoking (DQS) in the Cancer Prevention Study-3 cohort. Difficult quitters were defined as having made at least ten quit attempts, whether successful or not, and easy quitters as having quit after only one attempt. Only individuals of European ancestry were selected for the study. Among 10,004 smokers (5,071 difficult quitters, 4,933 easy quitters), we assessed the genetic heritability of DQS and evaluated associations between DQS and each genome-wide variant using logistic regression while adjusting for confounders, including smoking intensity (cigarettes per day). RESULTS The genetic heritability of the DQS phenotype was 13 %, comparable to, or higher than, the reported heritability of other smoking behaviors (e.g., smoking intensity, cessation). Although no variants were genome-wide significant, several genes were identified at a subthreshold level (p < 10-4). A variant in MEGF9 (rs149760032), a transmembrane protein largely expressed in the central nervous system, showed the strongest association with DQS (OR = 0.60, p = 1.3x10-7). Additional variants associated with DQS independently by smoking intensity were also detected in GLRA3 (rs73006492, OR = 0.77, p = 5.6x10-7) and FOCAD (rs112251973, OR = 1.96, p = 1.8x10-6) and are plausibly related to smoking cessation through pathways in the brain and respiratory system. CONCLUSIONS The use of an alternative cessation phenotype based on difficulty quitting smoking facilitated the identification of new pathways that could lead to unique smoking treatments.
Collapse
Affiliation(s)
- Adriana Lori
- Department of Population Science, American Cancer Society, Atlanta, GA, USA.
| | - Alpa V Patel
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - J Lee Westmaas
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - W Ryan Diver
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| |
Collapse
|
39
|
Varshosaz P, O'Connor C, Moise AR. Feedback regulation of retinaldehyde reductase DHRS3, a critical determinant of retinoic acid homeostasis. FEBS Lett 2025; 599:340-351. [PMID: 39420244 PMCID: PMC11808460 DOI: 10.1002/1873-3468.15038] [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: 08/21/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Retinoic acid is crucial for vertebrate embryogenesis, influencing anterior-posterior patterning and organogenesis through its interaction with nuclear hormone receptors comprising heterodimers of retinoic acid receptors (RARα, β, or γ) and retinoid X receptors (RXRα, β, or γ). Tissue retinoic acid levels are tightly regulated since both its excess and deficiency are deleterious. Dehydrogenase/reductase 3 (DHRS3) plays a critical role in this regulation by converting retinaldehyde to retinol, preventing excessive retinoic acid formation. Mutations in DHRS3 can result in embryonic lethality and congenital defects. This study shows that mouse Dhrs3 expression is responsive to vitamin A status and is directly regulated by the RAR/RXR complex through cis-regulatory elements. This highlights a negative feedback mechanism that ensures retinoic acid homeostasis.
Collapse
Affiliation(s)
- Parisa Varshosaz
- Biology and Biomolecular Sciences Ph.D. Program, Northern Ontario School of MedicineLaurentian UniversitySudburyCanada
| | - Catherine O'Connor
- Medical Sciences DivisionNorthern Ontario School of MedicineSudburyCanada
| | - Alexander R. Moise
- Medical Sciences DivisionNorthern Ontario School of MedicineSudburyCanada
- Department of Biology and Biomolecular Sciences ProgramLaurentian UniversitySudburyCanada
| |
Collapse
|
40
|
Liu Y, Li Z, Chen X, Cui X, Gao Z, Jiang R. INSTINCT: Multi-sample integration of spatial chromatin accessibility sequencing data via stochastic domain translation. Nat Commun 2025; 16:1247. [PMID: 39893190 PMCID: PMC11787322 DOI: 10.1038/s41467-025-56535-0] [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: 05/31/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
Recent advances in spatial epigenomic techniques have given rise to spatial assay for transposase-accessible chromatin using sequencing (spATAC-seq) data, enabling the characterization of epigenomic heterogeneity and spatial information simultaneously. Integrative analysis of multiple spATAC-seq samples, for which no method has been developed, allows for effective identification and elimination of unwanted non-biological factors within the data, enabling comprehensive exploration of tissue structures and providing a holistic epigenomic landscape, thereby facilitating the discovery of biological implications and the study of regulatory processes. In this article, we present INSTINCT, a method for multi-sample INtegration of Spatial chromaTIN accessibility sequencing data via stochastiC domain Translation. INSTINCT can efficiently handle the high dimensionality of spATAC-seq data and eliminate the complex noise and batch effects of samples through a stochastic domain translation procedure. We demonstrate the superiority and robustness of INSTINCT in integrating spATAC-seq data across multiple simulated scenarios and real datasets. Additionally, we highlight the advantages of INSTINCT in spatial domain identification, visualization, spot-type annotation, and various downstream analyses, including motif enrichment analysis, expression enrichment analysis, and partitioned heritability analysis.
Collapse
Affiliation(s)
- Yuyao Liu
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Zhen Li
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Xiaoyang Chen
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Xuejian Cui
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Zijing Gao
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Rui Jiang
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
41
|
Chan PP, Holmes AD, Lowe TM. Analyzing, visualizing, and annotating tRNA-derived RNAs using tRAX and tDRnamer. Methods Enzymol 2025; 711:103-133. [PMID: 39952700 DOI: 10.1016/bs.mie.2024.11.016] [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: 02/17/2025]
Abstract
tRNA-derived RNAs (tDRs) are known for their diverse regulatory roles in many organisms. These small RNA transcripts have been identified mainly by high-throughput RNA sequencing, numbering hundreds to thousands of unique molecules in any given biological sample. As such, bioinformatic analysis is essential in understanding the features, complexity, and unexplored biological patterns of tDRs. This chapter describes use of tRAX: tRNA Analysis of eXpression, a specially designed comprehensive end-to-end software pipeline for tDR abundance estimation, differential expression comparison, and inference of RNA modifications from raw small RNA sequencing data. We also demonstrate tDRnamer, a web- and command-line-based companion tool that provides automated, standardized tDR naming and annotations based on source tRNAs and related tDRs.
Collapse
Affiliation(s)
- Patricia P Chan
- Department of Biomolecular Engineering, University of California, Santa Cruz, CA, United States
| | - Andrew D Holmes
- Department of Biomolecular Engineering, University of California, Santa Cruz, CA, United States
| | - Todd M Lowe
- Department of Biomolecular Engineering, University of California, Santa Cruz, CA, United States.
| |
Collapse
|
42
|
Ehn E, Eisfeldt J, Laffita-Mesa JM, Thonberg H, Schoumans J, Portaankorva AM, Viitanen M, Lindstrand A, Nennesmo I, Graff C. A de novo, mosaic and complex chromosome 21 rearrangement causes APP triplication and familial autosomal dominant early onset Alzheimer disease. Sci Rep 2025; 15:2912. [PMID: 39849058 PMCID: PMC11759332 DOI: 10.1038/s41598-025-86645-0] [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: 09/06/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
Copy number variation (CNV) of the amyloid-β precursor protein gene (APP) is a known cause of autosomal dominant Alzheimer disease (ADAD), but de novo genetic variants causing ADAD are rare. We report a mother and daughter with neuropathologically confirmed definite Alzheimer disease (AD) and extensive cerebral amyloid angiopathy (CAA). Copy number analysis identified an increased number of APP copies and genome sequencing (GS) revealed the underlying complex genomic rearrangement (CGR) including a triplication of APP with two unique breakpoint junctions (BPJs). The mosaic state in the mother had likely occurred de novo. Digital droplet PCR (ddPCR) on 42 different tissues, including 17 different brain regions, showed the derivative chromosome at varying mosaic levels (20-96%) in the mother who had symptom onset at age 58 years. In contrast, the derivative chromosome was present in all analyzed cells in the daughter whose symptom onset was at 34 years. This study reveals the architecture of a de novo CGR causing APP triplication and ADAD with a striking difference in age at onset between the fully heterozygous daughter compared to the mosaic mother. The GS analysis identified the complexity of the CGR illustrating its usefulness in identifying structural variants (SVs) in neurodegenerative disorders.
Collapse
Affiliation(s)
- Emma Ehn
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Unit for Hereditary Dementias, Karolinska University Hospital Solna, Stockholm, Sweden.
| | - Jesper Eisfeldt
- Department for Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Jose M Laffita-Mesa
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Unit for Hereditary Dementias, Karolinska University Hospital Solna, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Thonberg
- Department for Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics and Genomics, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jacqueline Schoumans
- Département de Médicine de Laboratoire et Pathologie, Centre Universitaire Hospitalier Vaudois (CHUV), Lausanne, Switzerland
| | - Anne M Portaankorva
- Faculty of Medicine, Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Matti Viitanen
- Division for Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Anna Lindstrand
- Department for Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics and Genomics, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Inger Nennesmo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Caroline Graff
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Unit for Hereditary Dementias, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
43
|
Smith JR, Tutaj MA, Thota J, Lamers L, Gibson AC, Kundurthi A, Gollapally VR, Brodie KC, Zacher S, Laulederkind SJF, Hayman GT, Wang SJ, Tutaj M, Kaldunski ML, Vedi M, Demos WM, De Pons JL, Dwinell MR, Kwitek AE. Standardized pipelines support and facilitate integration of diverse datasets at the Rat Genome Database. Database (Oxford) 2025; 2025:baae132. [PMID: 39841812 PMCID: PMC11753291 DOI: 10.1093/database/baae132] [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: 05/14/2024] [Revised: 11/01/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
The Rat Genome Database (RGD) is a multispecies knowledgebase which integrates genetic, multiomic, phenotypic, and disease data across 10 mammalian species. To support cross-species, multiomics studies and to enhance and expand on data manually extracted from the biomedical literature by the RGD team of expert curators, RGD imports and integrates data from multiple sources. These include major databases and a substantial number of domain-specific resources, as well as direct submissions by individual researchers. The incorporation of these diverse datatypes is handled by a growing list of automated import, export, data processing, and quality control pipelines. This article outlines the development over time of a standardized infrastructure for automated RGD pipelines with a summary of key design decisions and a focus on lessons learned.
Collapse
Affiliation(s)
- Jennifer R Smith
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Marek A Tutaj
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Jyothi Thota
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Logan Lamers
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Adam C Gibson
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Akhilanand Kundurthi
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Varun Reddy Gollapally
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Kent C Brodie
- Clinical and Translational Science Institute, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Stacy Zacher
- Finance and Administration, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Stanley J F Laulederkind
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - G Thomas Hayman
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Shur-Jen Wang
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Monika Tutaj
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Mary L Kaldunski
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Mahima Vedi
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Wendy M Demos
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Jeffrey L De Pons
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Melinda R Dwinell
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| | - Anne E Kwitek
- Rat Genome Database, Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, United States
| |
Collapse
|
44
|
Mcgonigal M, Ito K. From Soil to Surface: Exploring the Impact of Green Infrastructure on Microbial Communities in the Built Environment. J Genomics 2025; 13:10-23. [PMID: 39925382 PMCID: PMC11803137 DOI: 10.7150/jgen.106245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 12/10/2024] [Indexed: 02/11/2025] Open
Abstract
High microbial diversity offers extensive benefits to both the environment and human health, contributing to ecosystem stability, nutrient cycling, and pathogen suppression. In built environments, factors such as building design, human activity, and cleaning protocols influence microbial communities. This study investigates the impact of landscape design on microbial diversity and function within the "Visionary Lab" exhibition in Tokyo, Japan, using 16S rRNA gene amplicon sequencing and shallow shotgun sequencing. Despite the limited sample size, the study suggests that the Visionary Lab samples may exhibit higher microbial diversity compared to other museum areas. Potential distinct microbial community structures may be correlated with sampling locations. However, despite this, no consistent patterns were observed in virulence factors or antimicrobial resistance genes across the samples. Metabolic function analysis showed varied profiles, suggesting diverse ecological interactions influenced that may be by the curated landscape. This suggest that the curated landscape design may have the potential to enhance microbial diversity, highlighting a possible avenue to create healthier and more sustainable built environments. However, the lack of consistent patterns in virulence factors and antimicrobial resistance genes underscores the complexity of microbial community dynamics.
Collapse
Affiliation(s)
- Malin Mcgonigal
- BIOTA Inc., Tokyo, 101-0022, Japan
- Department of Biosciences, Durham University, Durham, DH1 3LE, England
| | | |
Collapse
|
45
|
Jusuf JM, Grosse-Holz S, Gabriele M, Mach P, Flyamer IM, Zechner C, Giorgetti L, Mirny LA, Hansen AS. Genome-wide absolute quantification of chromatin looping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632736. [PMID: 39935886 PMCID: PMC11812599 DOI: 10.1101/2025.01.13.632736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
3D genomics methods such as Hi-C and Micro-C have uncovered chromatin loops across the genome and linked these loops to gene regulation. However, these methods only measure 3D interaction probabilities on a relative scale. Here, we overcome this limitation by using live imaging data to calibrate Micro-C in mouse embryonic stem cells, thus obtaining absolute looping probabilities for 36,804 chromatin loops across the genome. We find that the looped state is generally rare, with a mean probability of 2.3% and a maximum of 26% across the quantified loops. On average, CTCF-CTCF loops are stronger than loops between cis-regulatory elements (3.2% vs. 1.1%). Our findings can be extended to human stem cells and differentiated cells under certain assumptions. Overall, we establish an approach for genome-wide absolute loop quantification and report that loops generally occur with low probabilities, generalizing recent live imaging results to the whole genome.
Collapse
Affiliation(s)
- James M. Jusuf
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA
| | - Simon Grosse-Holz
- Center for Systems Biology Dresden, 01307 Dresden, Germany
- Max-Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany
| | - Michele Gabriele
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA
| | - Pia Mach
- Friedrich Miescher Institute for Biomedical Research, 4065 Basel, Switzerland
- University of Basel, 4001 Basel, Switzerland
| | - Ilya M. Flyamer
- Friedrich Miescher Institute for Biomedical Research, 4065 Basel, Switzerland
| | - Christoph Zechner
- Center for Systems Biology Dresden, 01307 Dresden, Germany
- Scuola Internazionale Superiori di Studi Avanzati, 34136 Trieste, Italy
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Luca Giorgetti
- Friedrich Miescher Institute for Biomedical Research, 4065 Basel, Switzerland
| | - Leonid A. Mirny
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anders S. Hansen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA
| |
Collapse
|
46
|
Kubitschek J, Takhaveev V, Mingard C, Rochlitz M, Reinert P, Keller G, Kloter T, Fernández Cereijo R, Huber S, McKeague M, Sturla S. Single-nucleotide-resolution genomic maps of O6-methylguanine from the glioblastoma drug temozolomide. Nucleic Acids Res 2025; 53:gkae1320. [PMID: 39831306 PMCID: PMC11744188 DOI: 10.1093/nar/gkae1320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 12/20/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
Temozolomide kills cancer cells by forming O6-methylguanine (O6-MeG), which leads to cell cycle arrest and apoptosis. However, O6-MeG repair by O6-methylguanine-DNA methyltransferase (MGMT) contributes to drug resistance. Characterizing genomic profiles of O6-MeG could elucidate how O6-MeG accumulation is influenced by repair, but there are no methods to map genomic locations of O6-MeG. Here, we developed an immunoprecipitation- and polymerase-stalling-based method, termed O6-MeG-seq, to locate O6-MeG across the whole genome at single-nucleotide resolution. We analyzed O6-MeG formation and repair across sequence contexts and functional genomic regions in relation to MGMT expression in a glioblastoma-derived cell line. O6-MeG signatures were highly similar to mutational signatures from patients previously treated with temozolomide. Furthermore, MGMT did not preferentially repair O6-MeG with respect to sequence context, chromatin state or gene expression level, however, may protect oncogenes from mutations. Finally, we found an MGMT-independent strand bias in O6-MeG accumulation in highly expressed genes. These data provide high resolution insight on how O6-MeG formation and repair are impacted by genome structure and nucleotide sequence. Further, O6-MeG-seq is expected to enable future studies of DNA modification signatures as diagnostic markers for addressing drug resistance and preventing secondary cancers.
Collapse
Affiliation(s)
- Jasmina Kubitschek
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Vakil Takhaveev
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Cécile Mingard
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Martha I Rochlitz
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Patricia B Reinert
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Giulia Keller
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Tom Kloter
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Raúl Fernández Cereijo
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Sabrina M Huber
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Maureen McKeague
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
- Department of Chemistry, Faculty of Science, McGill University, 801 Sherbrooke St. West, Montreal, QCH3A 0B8, Canada
- Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, 3655 Prom. Sir William Osler, Montreal, QCH3G 1Y6, Canada
| | - Shana J Sturla
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| |
Collapse
|
47
|
Nobusada T, Yip C, Agrawal S, Severin J, Abugessaisa I, Hasegawa A, Hon C, Ide S, Koido M, Kondo A, Masuya H, Oki S, Tagami M, Takada T, Terao C, Thalhath N, Walker S, Yasuzawa K, Shin J, de Hoon ML, Carninci P, Kawaji H, Kasukawa T. Update of the FANTOM web resource: enhancement for studying noncoding genomes. Nucleic Acids Res 2025; 53:D419-D424. [PMID: 39592010 PMCID: PMC11701582 DOI: 10.1093/nar/gkae1047] [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: 09/17/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
The FANTOM web resource (https://fantom.gsc.riken.jp/) has been a unique resource for studying mammalian genomes, which is built on the research activities conducted in the international collaborative project FANTOM (Functional ANnoTation Of the Mammalian genome). In recent updates, we expanded annotations for long non-coding RNAs (lncRNAs) and transcribed cis-regulatory elements (CREs). The former was derived from the large-scale lncRNA perturbations in induced pluripotent stem cells (iPSCs) and integrative analysis of Hi-C data conducted in the sixth iteration of the project (FANTOM6). The resulting annotations of lncRNAs, according to the impact on cellular and molecular phenotypes and the potential RNA-chromatin interactions, are accessible via the interactive ZENBU-Reports framework. The latter involves a new platform, fanta.bio (https://fanta.bio/), which collects transcribed CREs identified via use of an extended dataset of CAGE profiles. The CREs, with their annotations including genetic and epigenetic information, are accessible via a dedicated interface as well as the UCSC Genome Browser Database. These updates offer enhanced opportunities to investigate the functions of non-coding regions within mammalian genomes.
Collapse
Affiliation(s)
- Tomoe Nobusada
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Chi Wai Yip
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Saumya Agrawal
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Jessica Severin
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Imad Abugessaisa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Akira Hasegawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Chung Chau Hon
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Satoru Ide
- Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Masaru Koido
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 277-0882, Japan
| | - Atsushi Kondo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Hiroshi Masuya
- RIKEN BioResource Research Center, Tsukuba, Ibaraki 305-0074, Japan
| | - Shinya Oki
- Kumamoto University, Kumamoto 860-0811, Japan
| | - Michihira Tagami
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Toyoyuki Takada
- RIKEN BioResource Research Center, Tsukuba, Ibaraki 305-0074, Japan
| | - Chikashi Terao
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka 420-8527, Japan
| | - Nishad Thalhath
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Scott Walker
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Kayoko Yasuzawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Jay W Shin
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Michiel J L de Hoon
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Piero Carninci
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Hideya Kawaji
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Takeya Kasukawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
48
|
Hanna AD, Chang T, Ho KS, Yee RSZ, Walker WC, Agha N, Hsu CW, Jung SY, Dickinson ME, Samee MAH, Ward CS, Lee CS, Rodney GG, Hamilton SL. Mechanisms underlying dilated cardiomyopathy associated with FKBP12 deficiency. J Gen Physiol 2025; 157:e202413583. [PMID: 39661086 PMCID: PMC11633665 DOI: 10.1085/jgp.202413583] [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: 03/21/2024] [Revised: 08/14/2024] [Accepted: 10/22/2024] [Indexed: 12/12/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a highly prevalent and genetically heterogeneous condition that results in decreased contractility and impaired cardiac function. The FK506-binding protein FKBP12 has been implicated in regulating the ryanodine receptor in skeletal muscle, but its role in cardiac muscle remains unclear. To define the effect of FKBP12 in cardiac function, we generated conditional mouse models of FKBP12 deficiency. We used Cre recombinase driven by either the α-myosin heavy chain, (αMHC) or muscle creatine kinase (MCK) promoter, which are expressed at embryonic day 9 (E9) and E13, respectively. Both conditional models showed an almost total loss of FKBP12 in adult hearts compared with control animals. However, only the early embryonic deletion of FKBP12 (αMHC-Cre) resulted in an early-onset and progressive DCM, increased cardiac oxidative stress, altered expression of proteins associated with cardiac remodeling and disease, and sarcoplasmic reticulum Ca2+ leak. Our findings indicate that FKBP12 deficiency during early development results in cardiac remodeling and altered expression of DCM-associated proteins that lead to progressive DCM in adult hearts, thus suggesting a major role for FKBP12 in embryonic cardiac muscle.
Collapse
Affiliation(s)
- Amy D. Hanna
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Ting Chang
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Kevin S. Ho
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Rachel Sue Zhen Yee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Nadia Agha
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Chih-Wei Hsu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Sung Yun Jung
- Department of Biochemistry, Baylor College of Medicine, Houston, TX, USA
| | - Mary E. Dickinson
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Christopher S. Ward
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Chang Seok Lee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - George G. Rodney
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Susan L. Hamilton
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
49
|
Xu J, Wan J, Huang HY, Chen Y, Huang Y, Huang J, Zhang Z, Su C, Zhou Y, Lin X, Lin YCD, Huang HD. miRStart 2.0: enhancing miRNA regulatory insights through deep learning-based TSS identification. Nucleic Acids Res 2025; 53:D138-D146. [PMID: 39578697 PMCID: PMC11701676 DOI: 10.1093/nar/gkae1086] [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: 09/15/2024] [Revised: 10/17/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression by binding to the 3'-untranslated regions of target mRNAs, influencing various biological processes at the post-transcriptional level. Identifying miRNA transcription start sites (TSSs) and transcription factors' (TFs) regulatory roles is crucial for elucidating miRNA function and transcriptional regulation. miRStart 2.0 integrates over 4500 high-throughput datasets across five data types, utilizing a multi-modal approach to annotate 28 828 putative TSSs for 1745 human and 1181 mouse miRNAs, supported by sequencing-based signals. Over 6 million tissue-specific TF-miRNA interactions, integrated from ChIP-seq data, are supplemented by DNase hypersensitivity and UCSC conservation data, with network visualizations. Our deep learning-based model outperforms existing tools in miRNA TSS prediction, achieving the most overlaps with both cell-specific and non-cell-specific validated TSSs. The user-friendly web interface and visualization tools make miRStart 2.0 easily accessible to researchers, enabling efficient identification of miRNA upstream regulatory elements in relation to their TSSs. This updated database provides systems-level insights into gene regulation and disease mechanisms, offering a valuable resource for translational research, facilitating the discovery of novel therapeutic targets and precision medicine strategies. miRStart 2.0 is now accessible at https://awi.cuhk.edu.cn/∼miRStart2.
Collapse
Affiliation(s)
- Jiatong Xu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Jingting Wan
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Hsi-Yuan Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Yigang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Yixian Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Junyang Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Ziyue Zhang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Chang Su
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Yuming Zhou
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Xingqiao Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Yang-Chi-Dung Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Hsien-Da Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- Department of Endocrinology, Key Laboratory of Endocrinology of National Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.9 Dongdansantiao Street, Dongcheng District, Beijing 100730, P.R. China
| |
Collapse
|
50
|
He Y, Bao X, Chen T, Jiang Q, Zhang L, He LN, Zheng J, Zhao A, Ren J, Zuo Z. RPS 2.0: an updated database of RNAs involved in liquid-liquid phase separation. Nucleic Acids Res 2025; 53:D299-D309. [PMID: 39460625 PMCID: PMC11701738 DOI: 10.1093/nar/gkae951] [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: 09/13/2024] [Revised: 10/05/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Liquid-liquid phase separation (LLPS) is a crucial process for the formation of biomolecular condensates such as coacervate droplets, P-bodies and stress granules, which play critical roles in many physiological and pathological processes. Increasing studies have shown that not only proteins but also RNAs play a critical role in LLPS. To host LLPS-associated RNAs, we previously developed a database named 'RPS' in 2021. In this study, we present an updated version RPS 2.0 (https://rps.renlab.cn/) to incorporate the newly generated data and to host new LLPS-associated RNAs driven by post-transcriptional regulatory mechanisms. Currently, RPS 2.0 hosts 171 301 entries of LLPS-associated RNAs in 24 different biomolecular condensates with four evidence types, including 'Reviewed', 'High-throughput (LLPS enrichment)', 'High-throughput (LLPS perturbation)' and 'Predicted', and five event types, including 'Expression', 'APA', 'AS', 'A-to-I' and 'Modification'. Additionally, extensive annotations of LLPS-associated RNAs are provided in RPS 2.0, including RNA sequence and structure features, RNA-protein/RNA-RNA interactions, RNA modifications, as well as diseases related annotations. We expect that RPS 2.0 will further promote research of LLPS-associated RNAs and deepen our understanding of the biological functions and regulatory mechanisms of LLPS.
Collapse
Affiliation(s)
- Yongxin He
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Xiaoqiong Bao
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Tianjian Chen
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Qi Jiang
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Luowanyue Zhang
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Li-Na He
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Jian Zheng
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - An Zhao
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310000, China
| | - Jian Ren
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Zhixiang Zuo
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|