101
|
Orchard P, Blackwell TW, Kachuri L, Castaldi PJ, Cho MH, Christenson SA, Durda P, Gabriel S, Hersh CP, Huntsman S, Hwang S, Joehanes R, Johnson M, Li X, Lin H, Liu CT, Liu Y, Mak ACY, Manichaikul AW, Paik D, Saferali A, Smith JD, Taylor KD, Tracy RP, Wang J, Wang M, Weinstock JS, Weiss J, Wheeler HE, Zhou Y, Zoellner S, Wu JC, Mestroni L, Graw S, Taylor MRG, Ortega VE, Johnson CW, Gan W, Abecasis G, Nickerson DA, Gupta N, Ardlie K, Woodruff PG, Zheng Y, Bowler RP, Meyers DA, Reiner A, Kooperberg C, Ziv E, Ramachandran VS, Larson MG, Cupples LA, Burchard EG, Silverman EK, Rich SS, Heard-Costa N, Tang H, Rotter JI, Smith AV, Levy D, Aguet F, Scott L, Raffield LM, Parker SCJ. Cross-cohort analysis of expression and splicing quantitative trait loci in TOPMed. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.19.25322561. [PMID: 40034763 PMCID: PMC11875316 DOI: 10.1101/2025.02.19.25322561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Most genetic variants associated with complex traits and diseases occur in non-coding genomic regions and are hypothesized to regulate gene expression. To understand the genetics underlying gene expression variability, we characterize 14,324 ancestrally diverse RNA-sequencing samples from the NHLBI Trans-Omics for Precision Medicine (TOPMed) program and integrate whole genome sequencing data to perform cis and trans expression and splicing quantitative trait locus (cis-/trans-e/sQTL) analyses in six tissues and cell types, most notably whole blood (N=6,454) and lung (N=1,291). We show this dataset enables greater detection of secondary cis-e/sQTL signals than was achieved in previous studies, and that secondary cis-eQTL and primary trans-eQTL signal discovery is not saturated even though eGene discovery is. Most TOPMed trans-eQTL signals colocalize with cis-e/sQTL signals, suggesting many trans signals are mediated by cis signals. We fine-map European UK BioBank GWAS signals from 164 traits and colocalize the resulting 34,107 fine-mapped GWAS signals with TOPMed e/sQTL signals, finding that of 10,611 GWAS signals with a colocalization, 7,096 GWAS signals colocalize with at least one secondary e/sQTL signal. These results demonstrate that larger e/sQTL analyses will continue to uncover secondary e/sQTL signals, and that these new signals will benefit GWAS interpretation.
Collapse
Affiliation(s)
- Peter Orchard
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Thomas W Blackwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, School of Medicine, Stanford University, Stanford, CA, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephanie A Christenson
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | | | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Scott Huntsman
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Seungyong Hwang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health and Boston University, Framingham, MA, USA
| | - Mari Johnson
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Xingnan Li
- Division of Data Driven and Digital Medicine, Department of Medicine, Icahn School of Medicine at Mount Sanai, New York, NY, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sanai, New York, NY, USA
| | - Honghuang Lin
- Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health and Boston University, Framingham, MA, USA
- Department of Medicine, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Yongmei Liu
- Department of Medicine, School of Medicine, Duke University, Durham, NC, USA
| | - Angel C Y Mak
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ani W Manichaikul
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - David Paik
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Aabida Saferali
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua D Smith
- Northwest Genomics Center, University of Washington, Seattle, WA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Russell P Tracy
- Laboratory for Clinical Biochemistry Research, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Jiongming Wang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Mingqiang Wang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Joshua S Weinstock
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey Weiss
- Northwest Genomics Center, University of Washington, Seattle, WA, USA
| | - Heather E Wheeler
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, USA
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Ying Zhou
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sebastian Zoellner
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Luisa Mestroni
- Department of Medicine, Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sharon Graw
- Department of Medicine, Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew R G Taylor
- Department of Medicine, Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Victor E Ortega
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Phoenix, AZ, USA
| | - Craig W Johnson
- Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, USA
| | - Weiniu Gan
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Goncalo Abecasis
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Deborah A Nickerson
- Northwest Genomics Center, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Namrata Gupta
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | | | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Russell P Bowler
- Department of Genomic Sciences and Systems Biology, Cleveland Clinic, Cleveland, OH, USA
| | - Deborah A Meyers
- Department of Medicine, Division of Genetics, Genomics and Precision Medicine, University of Arizona, Tucson, AZ, USA
| | - Alex Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elad Ziv
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Vasan S Ramachandran
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University, Framingham, MA, USA
- Department of Medicine, School of Medicine, Boston University, Boston, MA, USA
| | - Martin G Larson
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University, Framingham, MA, USA
| | - L Adrienne Cupples
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Esteban G Burchard
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Nancy Heard-Costa
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Hua Tang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Albert V Smith
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health and Boston University, Framingham, MA, USA
| | - François Aguet
- Illumina Artificial Intelligence Laboratory, Illumina, Foster City, CA, USA
| | - Laura Scott
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
102
|
Hecker D, Song X, Baumgarten N, Diagel A, Katsaouni N, Li L, Li S, Maji RK, Behjati Ardakani F, Ma L, Tews D, Wabitsch M, Björkegren JL, Schunkert H, Chen Z, Schulz MH. Cell type-specific epigenetic regulatory circuitry of coronary artery disease loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639228. [PMID: 40027824 PMCID: PMC11870499 DOI: 10.1101/2025.02.20.639228] [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: 03/05/2025]
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Recently, hundreds of genomic loci have been shown to increase CAD risk, however, the molecular mechanisms underlying signals from CAD risk loci remain largely unclear. We sought to pinpoint the candidate causal coding and non-coding genes of CAD risk loci in a cell type-specific fashion. We integrated the latest statistics of CAD genetics from over one million individuals with epigenetic data from 45 relevant cell types to identify genes whose regulation is affected by CAD-associated single nucleotide variants (SNVs) via epigenetic mechanisms. Applying two statistical approaches, we identified 1,580 genes likely involved in CAD, about half of which have not been associated with the disease so far. Enrichment analysis and phenome-wide association studies linked the novel candidate genes to disease-specific pathways and CAD risk factors, corroborating their disease relevance. We showed that CAD-SNVs are enriched to regulate gene expression by affecting the binding of transcription factors (TFs) with cellular specificity. Of all the candidate genes, 23.5% represented non-coding RNAs (ncRNA), which likewise showed strong cell type specificity. We conducted a proof-of-concept biological validation for the novel CAD ncRNA gene IQCH-AS1 . CRISPR/Cas9-based gene knockout of IQCH-AS1 , in a human preadipocyte strain, resulted in reduced preadipocyte proliferation, less adipocyte lipid accumulation, and atherogenic cytokine profile. The cellular data are in line with the reduction of IQCH-AS1 in adipose tissues of CAD patients and the negative impact of risk alleles on its expression, suggesting IQCH-AS1 to be protective for CAD. Our study not only pinpoints CAD candidate genes in a cell type-specific fashion but also spotlights the roles of the understudied ncRNA genes in CAD genetics.
Collapse
Affiliation(s)
- Dennis Hecker
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Xiaoning Song
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nina Baumgarten
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Anastasiia Diagel
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nikoletta Katsaouni
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Ling Li
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Shuangyue Li
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Ranjan Kumar Maji
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Fatemeh Behjati Ardakani
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Lijiang Ma
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | - Daniel Tews
- German Center for Child and Adolescent Health (DZKJ), Partner Site Ulm
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075 Ulm, Germany
| | - Martin Wabitsch
- German Center for Child and Adolescent Health (DZKJ), Partner Site Ulm
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075 Ulm, Germany
| | - Johan L.M. Björkegren
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York 10029, USA
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, 14157 Huddinge, Sweden
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Zhifen Chen
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Marcel H. Schulz
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| |
Collapse
|
103
|
Su Y, Feng C, Ye W, Xiao J, Meng Q, Yang X, Wang Y, Huang T, Lan L, Chen S, Ding Z, Su S, Wei S, Shan Q. Exploring the dynamic responses of group 3 innate lymphoid cells at different times in response to LPS challenge. Int Immunopharmacol 2025; 148:114162. [PMID: 39889415 DOI: 10.1016/j.intimp.2025.114162] [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: 11/24/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Group 3 innate lymphoid cells (ILC3s) have clear roles in regulating mucosal immunity and tissue homeostasis in the intestine, though the immunological functions in lungs remain unclear. This study aimed to demonstrate the dynamic responses of ILC3s to acute inflammation upon LPS challenge. Microarray data and single-cell RNA sequencing (scRNA-seq) data obtained from the GEO database were combined to analyze the function of ILC3 subset, confirmed by flow cytometry assay and qRT-PCR. The gene enrichment analysis of intersected genes identified between microarray data in bacterial pneumonia and single-cell RNA sequencing of intestinal ILC3s were closely related to TNF-alpha effects on cytokine activity, cell motility and apoptosis pathway, indicating the possibility of intestinal ILC3s migration to the lung. Furthermore, the cellular landscapes of ILC3s in lung and intestine at different times after pulmonary infection exhibited varied ILC3 statuses. ILC3s in lung expanded a lot at 48 h while intestinal ILC3s decreased at 72 h response to LPS challenge, with higher expression of marked genes related to TNF-alpha effects on cytokine activity, cell motility and apoptosis pathway. The main findings in our study may serve as valuable resources for understanding the roles that ILC3s play upon LPS challenge, which may offer opportunities for translating ILC3s as therapeutic targets to regulate LPS-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Ying Su
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Caixia Feng
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Wenyu Ye
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Juan Xiao
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Qi Meng
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Xia Yang
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Yongcai Wang
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Ting Huang
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture Guangxi Academy of Fishery Sciences Nanning China
| | - Liancheng Lan
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Sixing Chen
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Ziting Ding
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Shiqi Su
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Sumei Wei
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Qingwen Shan
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China.
| |
Collapse
|
104
|
Nie J, Zhang S, Guo Y, Liu C, Shi J, Wu H, Na R, Liang Y, Yu S, Quan F, Liu K, Li M, Zhou M, Zhao Y, Li X, Luo S, Zhang Q, Wang G, Zhang Y, Yao Y, Xiao Y, Tai S, Zheng T. Mapping of the T-cell Landscape of Biliary Tract Cancer Unravels Anatomic Subtype-Specific Heterogeneity. Cancer Res 2025; 85:704-722. [PMID: 39570809 DOI: 10.1158/0008-5472.can-24-1173] [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: 04/10/2024] [Revised: 08/24/2024] [Accepted: 11/13/2024] [Indexed: 02/18/2025]
Abstract
Biliary tract cancer (BTC), encompassing diseases such as intrahepatic (ICC), extrahepatic cholangiocarcinoma (ECC), and gallbladder cancer, is not only increasing but also poses a significant and urgent health threat due to its high malignancy. Genomic differences point to the possibility that these subtypes represent distinct diseases. Elucidation of the specific distribution of T-cell subsets, critical to cancer immunity, across these diseases could provide better insights into the unique biology of BTC subtypes and help identify potential precision medicine strategies. To address this, we conducted single-cell RNA sequencing and T-cell receptor sequencing on CD3+ T cells from 36 samples from 16 patients with BTC across all subtypes and analyzed 355 pathologic slides to examine the spatial distribution of T cells and tertiary lymphoid structures. Compared with ICC and gallbladder cancer, ECC possessed a unique immune profile characterized by T-cell exhaustion, elevated CXCL13 expression in CD4+ T helper-like and CD8+CXCL13+ exhausted T cells, more mature tertiary lymphoid structures, and fewer desert immunophenotypes. Conversely, ICC displayed an inflamed immunophenotype with an enrichment of IFN-related pathways and high expression of LGALS1 in activated regulatory T cells, associated with immunosuppression. Inhibition of LGALS1 reduced tumor growth and regulatory T-cell prevalence in ICC mouse models. Overall, this study unveils T-cell diversity across BTC subtypes at the single-cell and spatial level that could open paths for tailored immunotherapies. Significance: Single-cell and spatial analyses detailed the T-cell characteristics specific to anatomic subtypes of biliary tract cancer, identifying unique immunologic features that could potentially be harnessed to improve patient outcomes.
Collapse
Affiliation(s)
- Jianhua Nie
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Shuyuan Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Ying Guo
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Caiqi Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Jiaqi Shi
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
- Department of Phase 1 Trials Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Haotian Wu
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ruisi Na
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Yingjian Liang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shan Yu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fei Quan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Kun Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Meng Zhou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Ying Zhao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Xuehan Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Shengnan Luo
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
| | - Qian Zhang
- Department of Abdominal Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guangyu Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Yuanfei Yao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Sheng Tai
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Molecular Oncology in Heilongjiang, Harbin, China
- Department of Phase 1 Trials Center, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
105
|
Batool F, Shireen H, Malik MF, Abrar M, Abbasi AA. The combinatorial binding syntax of transcription factors in forebrain-specific enhancers. Biol Open 2025; 14:BIO061751. [PMID: 39976127 PMCID: PMC11876843 DOI: 10.1242/bio.061751] [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: 09/30/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
Tissue-specific gene regulation in mammals involves the coordinated binding of multiple transcription factors (TFs). Using the forebrain as a model, we investigated the syntax of TF occupancy to determine tissue-specific enhancer regions. We analyzed forebrain-exclusive enhancers from the VISTA Enhancer Browser and a curated set of 23 TFs relevant to forebrain development and disease. Our findings revealed multiple distinct patterns of combinatorial TF binding, with the HES5-FOXP2-GATA3 triad being the most frequent in forebrain-specific enhancers. This syntactic structure was detected in 2614 enhancers from a genome-wide catalog of 25,000 predicted human forebrain enhancers. Notably, this catalog represents a computationally predicted dataset, distinct from the in vivo validated set of enhancers obtained from the VISTA Enhancer Browser. The shortlisted 2614 enhancers were further analyzed using genome-wide epigenetic data and evaluated for evolutionary conservation and disease relevance. Our findings highlight the value of these 2614 enhancers in forebrain-specific gene regulation and provide a framework for discovering tissue-specific enhancers, enhancing the understanding of enhancer function.
Collapse
Affiliation(s)
- Fatima Batool
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Huma Shireen
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Muhammad Faizan Malik
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Muhammad Abrar
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Amir Ali Abbasi
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|
106
|
Ventura-Gomes A, Carmo-Fonseca M. The spatial choreography of mRNA biosynthesis. J Cell Sci 2025; 138:JCS263504. [PMID: 40019352 DOI: 10.1242/jcs.263504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025] Open
Abstract
Properly timed gene expression is essential for all aspects of organismal physiology. Despite significant progress, our understanding of the complex mechanisms governing the dynamics of gene regulation in response to internal and external signals remains incomplete. Over the past decade, advances in technologies like light and cryo-electron microscopy (Cryo-EM), cryo-electron tomography (Cryo-ET) and high-throughput sequencing have spurred new insights into traditional paradigms of gene expression. In this Review, we delve into recent concepts addressing 'where' and 'when' gene transcription and RNA splicing occur within cells, emphasizing the dynamic spatial and temporal organization of the cell nucleus.
Collapse
Affiliation(s)
- André Ventura-Gomes
- Gulbenkian Institute for Molecular Medicine, Av. Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Maria Carmo-Fonseca
- Gulbenkian Institute for Molecular Medicine, Av. Professor Egas Moniz, 1649-028 Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisbon, Portugal
| |
Collapse
|
107
|
Fu Y, Yang X, Li S, Ma C, An Y, Cheng T, Liang Y, Sun S, Cheng T, Zhao Y, Wang J, Wang X, Xu P, Yin Y, Liang H, Liu N, Zou W, Chen B. Dynamic properties of transcriptional condensates modulate CRISPRa-mediated gene activation. Nat Commun 2025; 16:1640. [PMID: 39952932 PMCID: PMC11828908 DOI: 10.1038/s41467-025-56735-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 01/28/2025] [Indexed: 02/17/2025] Open
Abstract
CRISPR activation (CRISPRa) is a powerful tool for endogenous gene activation, yet the mechanisms underlying its optimal transcriptional activation remain unclear. By monitoring real-time transcriptional bursts, we find that CRISPRa modulates both burst duration and amplitude. Our quantitative imaging reveals that CRISPR-SunTag activators, with three tandem VP64-p65-Rta (VPR), form liquid-like transcriptional condensates and exhibit high activation potency. Although visible CRISPRa condensates are associated with some RNA bursts, the overall levels of phase separation do not correlate with transcriptional bursting or activation strength in individual cells. When the number of SunTag scaffolds is increased to 10 or more, solid-like condensates form, sequestering co-activators such as p300 and MED1. These condensates display low dynamicity and liquidity, resulting in ineffective gene activation. Overall, our studies characterize various phase-separated CRISPRa systems for gene activation, highlighting the foundational principles for engineering CRISPR-based programmable synthetic condensates with appropriate properties to effectively modulate gene expression.
Collapse
Affiliation(s)
- Yujuan Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Xiaoxuan Yang
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Sihui Li
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Chenyang Ma
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao An
- Center of Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Cheng
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Liang
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Shengbai Sun
- Center of Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianyi Cheng
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Yongyang Zhao
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Jianghu Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- The State Key Laboratory of Southwest Karst Mountain Biodiversity Conservation of Forestry Administration, School of Life Science, Guizhou Normal University, Guiyang, China
| | - Xiaoyue Wang
- The State Key Laboratory of Southwest Karst Mountain Biodiversity Conservation of Forestry Administration, School of Life Science, Guizhou Normal University, Guiyang, China
| | - Pengfei Xu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yafei Yin
- Center of Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongqing Liang
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nan Liu
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China.
| | - Wei Zou
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China.
- Insititute of Translational Medicine, Zhejiang University, Hangzhou, China.
| | - Baohui Chen
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China.
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China.
| |
Collapse
|
108
|
Sobański D, Sobańska M, Staszkiewicz R, Strojny D, Grabarek BO. Changes in the Expression Profile of Growth-Associated Protein 43 in Degenerative Lumbosacral Stenosis. J Clin Med 2025; 14:1223. [PMID: 40004753 PMCID: PMC11856692 DOI: 10.3390/jcm14041223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Degenerative spinal stenosis is a common condition associated with structural degeneration and pain, yet its molecular underpinnings remain incompletely understood. Growth-associated protein 43 (GAP-43), a key player in neuronal plasticity and regeneration, may serve as a biomarker for disease progression and pain severity. This study investigates the expression of GAP-43 at the mRNA and protein levels in the ligamentum flavum of affected patients. Methods: Samples were collected from 96 patients with degenerative spinal stenosis and 85 controls. GAP-43 mRNA expression was analyzed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), while protein levels were quantified via enzyme-linked immunosorbent assay (ELISA) and Western blot. Pain severity was assessed using the visual analog scale (VAS), and associations with lifestyle factors were analyzed. Results:GAP-43 mRNA expression was significantly downregulated in the study group compared to the controls (fold change = 0.58 ± 0.12, p < 0.05), with an inverse correlation to VAS pain severity (fold change = 0.76 at VAS 4 vs. 0.36 at VAS 10). Conversely, GAP-43 protein levels were markedly elevated in the study group (5.57 ± 0.21 ng/mL) when compared to controls (0.54 ± 0.87 ng/mL, p < 0.0001). Protein levels were also correlated with lifestyle factors, including smoking and alcohol consumption (p < 0.05). Conclusions: GAP-43 shows potential as a biomarker for pain severity and disease progression in degenerative spinal stenosis, in a manner influenced by lifestyle factors. Further research is needed to explore its diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Dawid Sobański
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, 30-693 Cracow, Poland;
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
| | - Małgorzata Sobańska
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, 30-693 Cracow, Poland;
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
| | - Rafał Staszkiewicz
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
- Department of Neurosurgery, 5th Military Clinical Hospital with the SP ZOZ Polyclinic in Krakow, 30-901 Krakow, Poland
- Department of Neurosurgery, Faculty of Medicine in Zabrze, Academy of Silesia, 40-555 Katowice, Poland
| | - Damian Strojny
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
- Institute of Health Care, National Academy of Applied Sciences in Przemyśl, 37-700 Przemyśl, Poland
- New Medical Techniques Specialist Hospital of St. Family in Rudna Mała, 36-060 Rzeszów, Poland
| | - Beniamin Oskar Grabarek
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
| |
Collapse
|
109
|
Pitzen SP, Rudenick AN, Qiu Y, Zhang W, Munro SA, McCluskey BM, Forster C, Bergom HE, Ali A, Boytim E, Lafin JT, Linder S, Ismail M, Devlies W, Sessions CJ, Claessens F, Joniau S, Attard G, Zwart W, Nelson PS, Corey E, Wang Y, Lang JM, Beltran H, Strand D, Antonarakis ES, Hwang J, Murugan P, Huang RS, Dehm SM. Comparative transcriptomics reveals a mixed basal, club, and hillock epithelial cell identity in castration-resistant prostate cancer. Proc Natl Acad Sci U S A 2025; 122:e2415308122. [PMID: 39913208 PMCID: PMC11831193 DOI: 10.1073/pnas.2415308122] [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: 07/30/2024] [Accepted: 01/06/2025] [Indexed: 02/19/2025] Open
Abstract
Inhibiting the androgen receptor (AR) is effective for treatment of advanced prostate cancers because of their AR-dependent luminal epithelial cell identity. Tumors progress during therapy to castration-resistant prostate cancer (CRPC) by restoring AR signaling and maintaining luminal identity or by converting through lineage plasticity to a neuroendocrine (NE) identity or double-negative CRPC (DNPC) lacking luminal or NE identities. Here, we show that DNPC cells express genes defining basal, club, and hillock epithelial cells from benign prostate. We identified KLF5 as a regulator of genes defining this mixed basal, club, and hillock cell identity in DNPC models. KLF5-mediated upregulation of RARG uncovered a DNPC sensitivity to growth inhibition by retinoic acid receptor agonists, which down-regulated KLF5 and up-regulated AR. These findings offer CRPC classifications based on prostate epithelial cell identities and nominate KLF5 and RARG as therapeutic targets for CRPC displaying a mixed basal, club, and hillock identity.
Collapse
Affiliation(s)
- Samuel P. Pitzen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Graduate Program in Molecular, Cellular, and Developmental Biology and Genetics, University of Minnesota, Minneapolis, MN55455
| | - Amber N. Rudenick
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
| | - Yinjie Qiu
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN55455
| | - Weijie Zhang
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN55455
| | - Sarah A. Munro
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN55455
| | - Braedan M. McCluskey
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN55455
| | - Colleen Forster
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN55455
| | - Hannah E. Bergom
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - Atef Ali
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - Ella Boytim
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - John T. Lafin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Simon Linder
- Division on Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands1066 CX
| | - Mazlina Ismail
- Department of Oncology, University College London Cancer Institute, London, United KingdomWC1E 6BT
| | - Wout Devlies
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium
- Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Leuven3000, Belgium
| | | | - Frank Claessens
- Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Leuven3000, Belgium
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium
- Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven3000, Belgium
| | - Gerhardt Attard
- Department of Oncology, University College London Cancer Institute, London, United KingdomWC1E 6BT
- University College London Hospitals, LondonWC1E 6DN, United Kingdom
| | - Wilbert Zwart
- Division on Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands1066 CX
| | - Peter S. Nelson
- Division of Hematology and Oncology, University of Washington, Fred Hutchinson Cancer Center, SeattleWA98109
- Human Biology Division, Fred Hutchinson Cancer Center, SeattleWA98109
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA98195
| | - Yuzhuo Wang
- Department of Urologic Sciences, Faculty of Medicine, Vancouver Prostate Centre, University of British Columbia, Vancouver, BCV6H 3Z6, Canada
- Department of Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BCV5Z 1L3, Canada
| | - Joshua M. Lang
- Department of Medicine, University of Wisconsin-Madison, Madison, WI53792
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI53792
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, MA02115
| | - Douglas Strand
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Emmanuel S. Antonarakis
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - Justin Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - Paari Murugan
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN55455
| | - R. Stephanie Huang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN55455
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN55455
- Department of Urology, University of Minnesota, Minneapolis, MN55455
| |
Collapse
|
110
|
Li J, Zhang P, Xi X, Liu L, Wei L, Wang X. Modeling and designing enhancers by introducing and harnessing transcription factor binding units. Nat Commun 2025; 16:1469. [PMID: 39922842 PMCID: PMC11807178 DOI: 10.1038/s41467-025-56749-2] [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: 08/16/2024] [Accepted: 01/24/2025] [Indexed: 02/10/2025] Open
Abstract
Enhancers serve as pivotal regulators of gene expression throughout various biological processes by interacting with transcription factors (TFs). While transcription factor binding sites (TFBSs) are widely acknowledged as key determinants of TF binding and enhancer activity, the significant role of their surrounding context sequences remains to be quantitatively characterized. Here we propose the concept of transcription factor binding unit (TFBU) to modularly model enhancers by quantifying the impact of context sequences surrounding TFBSs using deep learning models. Based on this concept, we develop DeepTFBU, a comprehensive toolkit for enhancer design. We demonstrate that designing TFBS context sequences can significantly modulate enhancer activities and produce cell type-specific responses. DeepTFBU is also highly efficient in the de novo design of enhancers containing multiple TFBSs. Furthermore, DeepTFBU enables flexible decoupling and optimization of generalized enhancers. We prove that TFBU is a crucial concept, and DeepTFBU is highly effective for rational enhancer design.
Collapse
Affiliation(s)
- Jiaqi Li
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division, Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, Beijing, China
| | - Pengcheng Zhang
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division, Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, Beijing, China
| | - Xi Xi
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division, Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, Beijing, China
| | - Liyang Liu
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division, Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, Beijing, China
| | - Lei Wei
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division, Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, Beijing, China
| | - Xiaowo Wang
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division, Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, Beijing, China.
| |
Collapse
|
111
|
Russo T, Plessis-Belair J, Sher R, Riessland M. Regulatory Network Inference of Induced Senescent Midbrain Cell Types Reveals Cell Type-Specific Senescence-Associated Transcriptional Regulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636893. [PMID: 39975267 PMCID: PMC11839108 DOI: 10.1101/2025.02.06.636893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Cellular senescence of brain cell types has become an increasingly important perspective for both aging and neurodegeneration, specifically in the context of Parkinson's Disease (PD). The characterization of classical hallmarks of senescence is a widely debated topic, whereby the context in which a senescence phenotype is being investigated, such as the cell type, the inducing stressor, and/or the model system, is an extremely important aspect to consider when defining a senescent cell. Here, we describe a cell type-specific profile of senescence through the investigation of various canonical senescence markers in five human midbrain cell lines using chronic 5-Bromodeoxyuridine (BrdU) treatment as a model of DNA damage-induced senescence. We used principal component analysis (PCA) and subsequent regulatory network inference to define both unique and common senescence profiles in the cell types investigated, as well as revealed senescence-associated transcriptional regulators (SATRs). Functional characterization of one of the identified regulators, transcription factor AP4 (TFAP4), further highlights the cell type-specificity of the expression of the various senescence hallmarks. Our data indicates that SATRs modulate cell type-specific profiles of induced senescence in key midbrain cell types that play an important role in the context of aging and PD.
Collapse
Affiliation(s)
- Taylor Russo
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Jonathan Plessis-Belair
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Roger Sher
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Markus Riessland
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
112
|
Chang L, Ren B. Efficient, scalable, and near-nucleotide-resolution profiling of protein occupancy in the genome with deaminases. Proc Natl Acad Sci U S A 2025; 122:e2425203122. [PMID: 39869813 PMCID: PMC11804588 DOI: 10.1073/pnas.2425203122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025] Open
Affiliation(s)
- Lei Chang
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA92093
| | - Bing Ren
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA92093
- Center for Epigenomics, University of California, San Diego School of Medicine, La Jolla, CA92093
- Moores Cancer Center, University of California, San Diego School of Medicine, La Jolla, CA92093
| |
Collapse
|
113
|
Morin A, Chu CP, Pavlidis P. Identifying Reproducible Transcription Regulator Coexpression Patterns with Single Cell Transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.15.580581. [PMID: 38559016 PMCID: PMC10979919 DOI: 10.1101/2024.02.15.580581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The proliferation of single cell transcriptomics has potentiated our ability to unveil patterns that reflect dynamic cellular processes such as the regulation of gene transcription. In this study, we leverage a broad collection of single cell RNA-seq data to identify the gene partners whose expression is most coordinated with each human and mouse transcription regulator (TR). We assembled 120 human and 103 mouse scRNA-seq datasets from the literature (>28 million cells), constructing a single cell coexpression network for each. We aimed to understand the consistency of TR coexpression profiles across a broad sampling of biological contexts, rather than examine the preservation of context-specific signals. Our workflow therefore explicitly prioritizes the patterns that are most reproducible across cell types. Towards this goal, we characterize the similarity of each TR's coexpression within and across species. We create single cell coexpression rankings for each TR, demonstrating that this aggregated information recovers literature curated targets on par with ChIP-seq data. We then combine the coexpression and ChIP-seq information to identify candidate regulatory interactions supported across methods and species. Finally, we highlight interactions for the important neural TR ASCL1 to demonstrate how our compiled information can be adopted for community use.
Collapse
Affiliation(s)
- Alexander Morin
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, BC, Canada
| | - C. Pan Chu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, BC, Canada
| | - Paul Pavlidis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
114
|
Jeong R, Bulyk ML. Meta-analysis reveals transcription factors and DNA binding domain variants associated with congenital heart defect and orofacial cleft. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.30.25321274. [PMID: 39974057 PMCID: PMC11838631 DOI: 10.1101/2025.01.30.25321274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Many structural birth defect patients lack genetic diagnoses because there are many disease genes as yet to be discovered. We applied a gene burden test incorporating de novo predicted-loss-of-function (pLoF) and likely damaging missense variants together with inherited pLoF variants to a collection of congenital heart defect (CHD) and orofacial cleft (OC) parent-offspring trio cohorts (n = 3,835 and 1,844, respectively). We identified 17 novel candidate CHD genes and 10 novel candidate OC genes, of which many were known developmental disorder genes. Shorter genes were more powered in a "de novo only" analysis as compared to analysis including inherited pLoF variants. TFs were enriched among the significant genes; 14 and 8 transcription factor (TF) genes showed significant variant burden for CHD and OC, respectively. In total, 30 affected children had a de novo missense variant in a DNA binding domain of a known CHD, OC, and other developmental disorder TF genes. Our results suggest candidate pathogenic variants in CHD and OC and their potentially pleiotropic effects in other developmental disorders.
Collapse
Affiliation(s)
- Raehoon Jeong
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Bioinformatics and Integrative Genomics Graduate Program, Harvard University, Cambridge, MA 02138, USA
| | - Martha L. Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Bioinformatics and Integrative Genomics Graduate Program, Harvard University, Cambridge, MA 02138, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
115
|
Ascui G, Cedillo-Castelan V, Mendis A, Phung E, Liu HY, Verstichel G, Chandra S, Murray MP, Luna C, Cheroutre H, Kronenberg M. Innateness transcriptome gradients characterize mouse T lymphocyte populations. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:223-237. [PMID: 40073243 PMCID: PMC11878997 DOI: 10.1093/jimmun/vkae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 11/01/2024] [Indexed: 03/14/2025]
Abstract
A fundamental dichotomy in lymphocytes separates adaptive T and B lymphocytes, with clonally expressed antigen receptors, from innate lymphocytes, which carry out more rapid responses. Some T cell populations, however, are intermediates between these 2 poles, with the capacity to respond rapidly through T cell receptor activation or by cytokine stimulation. Here, using publicly available datasets, we constructed linear mixed models that not only define a gradient of innate gene expression in common for mouse innate-like T cells, but also are applicable to other mouse T lymphoid populations. A similar gradient could be identified for chromatin landscape based on ATAC-seq (assay for transposase-accessible chromatin using sequencing) data. The gradient included increased transcripts related to many traits of innate immune responses, with increased scores related to evidence for antigen experience. While including genes typical for T helper 1 (Th1) responses, the innateness gene program could be separated from Th1, Th2, and Th17 responses. Lymphocyte populations with higher innateness scores correlated with lower calcium-dependent T cell receptor-mediated cell activation, with some downstream signaling proteins dependent on calcium or affecting metabolism prephosphorylation. Therefore, as a group, different mouse innate-like T cell populations had related gene expression programs and activation pathways that are different from naive CD4 and CD8 T cells.
Collapse
Affiliation(s)
- Gabriel Ascui
- La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| | | | - Alba Mendis
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Eleni Phung
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Hsin-Yu Liu
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | - Shilpi Chandra
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | - Cindy Luna
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Hilde Cheroutre
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
116
|
Leib L, Juli J, Jurida L, Mayr-Buro C, Priester J, Weiser H, Wirth S, Hanel S, Heylmann D, Weber A, Schmitz ML, Papantonis A, Bartkuhn M, Wilhelm J, Linne U, Meier-Soelch J, Kracht M. The proximity-based protein interactome and regulatory logics of the transcription factor p65 NF-κB/RELA. EMBO Rep 2025; 26:1144-1183. [PMID: 39753783 PMCID: PMC11850942 DOI: 10.1038/s44319-024-00339-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 02/26/2025] Open
Abstract
The protein interactome of p65/RELA, the most active subunit of the transcription factor (TF) NF-κB, has not been previously determined in living cells. Using p65-miniTurbo fusion proteins and biotin tagging, we identify >350 RELA interactors from untreated and IL-1α-stimulated cells, including many TFs (47% of all interactors) and >50 epigenetic regulators belonging to different classes of chromatin remodeling complexes. A comparison with the interactomes of two point mutants of p65 reveals that the interactions primarily require intact dimerization rather than DNA-binding properties. A targeted RNAi screen for 38 interactors and subsequent functional transcriptome and bioinformatics studies identify gene regulatory (sub)networks, each controlled by RELA in combination with one of the TFs ZBTB5, GLIS2, TFE3/TFEB, or S100A8/A9. The large, dynamic and versatile high-resolution interactome of RELA and its gene regulatory logics provides a rich resource and a new framework for explaining how RELA cooperativity determines gene expression patterns.
Collapse
Affiliation(s)
- Lisa Leib
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Jana Juli
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Liane Jurida
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Christin Mayr-Buro
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Jasmin Priester
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Hendrik Weiser
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Stefanie Wirth
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Simon Hanel
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Daniel Heylmann
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Axel Weber
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | | | - Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Marek Bartkuhn
- Biomedical Informatics and Systems Medicine, Justus Liebig University Giessen, Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Jochen Wilhelm
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- German Center for Lung Research (DZL) and Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Uwe Linne
- Mass Spectrometry Facility of the Department of Chemistry, Philipps University, Marburg, Germany
| | - Johanna Meier-Soelch
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany.
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany.
- Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany.
- German Center for Lung Research (DZL) and Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.
| |
Collapse
|
117
|
Parisot N, Ribeiro Lopes M, Peignier S, Baa-Puyoulet P, Charles H, Calevro F, Callaerts P. Annotation of transcription factors, chromatin-associated factors, and basal transcription machinery in the pea aphid, Acyrthosiphon pisum, and development of the ATFdb database, a resource for studies of transcriptional regulation. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 177:104217. [PMID: 39579797 DOI: 10.1016/j.ibmb.2024.104217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/15/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
The pea aphid, Acyrthosiphon pisum, is an emerging model system in functional and comparative genomics, in part due to the availability of new genomic approaches and the different sequencing and annotation efforts that the community has dedicated to this important crop pest insect. The pea aphid is also used as a model to study fascinating biological traits of aphids, such as their extensive polyphenisms, their bacteriocyte-confined nutritional symbiosis, or their adaptation to the highly unbalanced diet represented by phloem sap. To get insights into the molecular basis of all these processes, it is important to have an appropriate annotation of transcription factors (TFs), which would enable the reconstruction/inference of gene regulatory networks in aphids. Using the latest version of the A. pisum genome assembly and annotation, which represents the first chromosome-level pea aphid genome, we annotated the complete repertoire of A. pisum TFs and complemented this information by annotating genes encoding chromatin-associated and basal transcription machinery proteins. These annotations were done combining information from the model Drosophila melanogaster, for which we also provide a revisited list of these proteins, and de novo prediction. The comparison between the two model systems allowed the identification of major losses or expansions in each genome, while a deeper analysis was made of ZNF TFs (with certain families expanded in the pea aphid), and the Hox gene cluster (showing reorganization in gene position in the pea aphid compared to D. melanogaster). All annotations are available to the community through the Aphid Transcription Factors database (ATFdb), consolidating the various annotations we generated. ATFdb serves as a valuable resource for gene regulation studies in aphids.
Collapse
Affiliation(s)
- Nicolas Parisot
- INSA Lyon, INRAE, BF2I, UMR0203, F-69621, Villeurbanne, France.
| | | | - Sergio Peignier
- INSA Lyon, INRAE, BF2I, UMR0203, F-69621, Villeurbanne, France
| | | | - Hubert Charles
- INSA Lyon, INRAE, BF2I, UMR0203, F-69621, Villeurbanne, France
| | | | - Patrick Callaerts
- KU Leuven, University of Leuven, Department of Human Genetics, Laboratory of Behavioral and Developmental Genetics, B-3000, Leuven, Belgium.
| |
Collapse
|
118
|
He J, Zhang Y, Liu Y, Zhou Z, Li T, Zhang Y, Xie B. BCDB: A dual-branch network based on transformer for predicting transcription factor binding sites. Methods 2025; 234:141-151. [PMID: 39701486 DOI: 10.1016/j.ymeth.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/28/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024] Open
Abstract
Transcription factor binding sites (TFBSs) are critical in regulating gene expression. Precisely locating TFBSs can reveal the mechanisms of action of different transcription factors in gene transcription. Various deep learning methods have been proposed to predict TFBS; however, these models often need help demonstrating ideal performance under limited data conditions. Furthermore, these models typically have complex structures, which makes their decision-making processes difficult to transparentize. Addressing these issues, we have developed a framework named BCDB. This framework integrates multi-scale DNA information and employs a dual-branch output strategy. Integrating DNABERT, convolutional neural networks (CNN), and multi-head attention mechanisms enhances the feature extraction capabilities, significantly improving the accuracy of predictions. This innovative method aims to balance the extraction of global and local information, enhancing predictive performance while utilizing attention mechanisms to provide an intuitive way to explain the model's predictions, thus strengthening the overall interpretability of the model. Prediction results on 165 ChIP-seq datasets show that BCDB significantly outperforms other existing deep learning methods in terms of performance. Additionally, since the BCDB model utilizes transfer learning methods, it can transfer knowledge learned from many unlabeled data to specific cell line prediction tasks, allowing our model to achieve cross-cell line TFBS prediction. The source code for BCDB is available on https://github.com/ZhangLab312/BCDB.
Collapse
Affiliation(s)
- Jia He
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Yupeng Zhang
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Yuhang Liu
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Zhigan Zhou
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Tianhao Li
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Yongqing Zhang
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Boqia Xie
- Department of Cardiology, Cardiovascualr Imaging Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
119
|
Karaoglu B, Gur Dedeoglu B. A Regulatory Circuits Analysis Tool, "miRCuit," Helps Reveal Breast Cancer Pathways: Toward Systems Medicine in Oncology. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025; 29:49-59. [PMID: 39853230 DOI: 10.1089/omi.2024.0201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
A systems medicine understanding of the regulatory molecular circuits that underpin breast cancer is essential for early cancer detection and precision/personalized medicine in clinical oncology. Transcription factors (TFs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) control gene expression and cell biology, and by extension, serve as pillars of the regulatory circuits that determine human health and disease. We report here the development of a regulatory circuit analysis program, miRCuit, constructing 10 different types of regulatory elements involving messenger RNA, miRNA, lncRNA, and TFs. Using the miRCuit, we analyzed expression profiling data from 179 invasive ductal breast carcinoma and 51 normal tissue samples from the Gene Expression Omnibus database. We identified eight circuit types along with two special types of circuits, one of which highlighted the significant roles of lncRNA CASC15, miR-130b-3p, and TF KLF5 in breast cancer development and progression. These findings advance our understanding of the regulatory molecules associated with breast cancer. Moreover, miRCuit offers a new avenue for users to construct circuits from regulatory molecules for potential applications to decipher disease pathogenesis.
Collapse
Affiliation(s)
- Begum Karaoglu
- Biotechnology Institute, Ankara University, Ankara, Turkey
- Intergen Genetics and Rare Diseases Diagnosis Center, Ankara, Turkey
| | | |
Collapse
|
120
|
Wei J, Gao C, Lu C, Wang L, Dong D, Sun M. The E2F family: a ray of dawn in cardiomyopathy. Mol Cell Biochem 2025; 480:825-839. [PMID: 38985251 DOI: 10.1007/s11010-024-05063-4] [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: 03/21/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024]
Abstract
Cardiomyopathies are a group of heterogeneous diseases, characterized by abnormal structure and function of the myocardium. For many years, it has been a hot topic because of its high morbidity and mortality as well as its complicated pathogenesis. The E2Fs, a group of transcription factors found extensively in eukaryotes, play a crucial role in governing cell proliferation, differentiation, and apoptosis, meanwhile their deregulated activity can also cause a variety of diseases. Based on accumulating evidence, E2Fs play important roles in cardiomyopathies. In this review, we describe the structural and functional characteristics of the E2F family and its role in cardiomyocyte processes, with a focus on how E2Fs are associated with the onset and development of cardiomyopathies. Moreover, we discuss the great potential of E2Fs as biomarkers and therapeutic targets, aiming to provide a reference for future research.
Collapse
Affiliation(s)
- Jinwen Wei
- College of Exercise and Health, Shenyang Sport University, No.36 Jinqiansong East Road, Shenyang, 110102, Liaoning, People's Republic of China
| | - Can Gao
- College of Exercise and Health, Shenyang Sport University, No.36 Jinqiansong East Road, Shenyang, 110102, Liaoning, People's Republic of China
| | - Changxu Lu
- College of Exercise and Health, Shenyang Sport University, No.36 Jinqiansong East Road, Shenyang, 110102, Liaoning, People's Republic of China
| | - Lijie Wang
- Department of Cardiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110033, Liaoning, People's Republic of China
| | - Dan Dong
- College of Basic Medical Science, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, No.36 Jinqiansong East Road, Shenyang, 110102, Liaoning, People's Republic of China.
| |
Collapse
|
121
|
Patalano SD, Fuxman Bass P, Fuxman Bass JI. Transcription factors in the development and treatment of immune disorders. Transcription 2025; 16:118-140. [PMID: 38100543 PMCID: PMC11970766 DOI: 10.1080/21541264.2023.2294623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Immune function is highly controlled at the transcriptional level by the binding of transcription factors (TFs) to promoter and enhancer elements. Several TF families play major roles in immune gene expression, including NF-κB, STAT, IRF, AP-1, NRs, and NFAT, which trigger anti-pathogen responses, promote cell differentiation, and maintain immune system homeostasis. Aberrant expression, activation, or sequence of isoforms and variants of these TFs can result in autoimmune and inflammatory diseases as well as hematological and solid tumor cancers. For this reason, TFs have become attractive drug targets, even though most were previously deemed "undruggable" due to their lack of small molecule binding pockets and the presence of intrinsically disordered regions. However, several aspects of TF structure and function can be targeted for therapeutic intervention, such as ligand-binding domains, protein-protein interactions between TFs and with cofactors, TF-DNA binding, TF stability, upstream signaling pathways, and TF expression. In this review, we provide an overview of each of the important TF families, how they function in immunity, and some related diseases they are involved in. Additionally, we discuss the ways of targeting TFs with drugs along with recent research developments in these areas and their clinical applications, followed by the advantages and disadvantages of targeting TFs for the treatment of immune disorders.
Collapse
Affiliation(s)
- Samantha D. Patalano
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Paula Fuxman Bass
- Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I. Fuxman Bass
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
122
|
Shea A, Eyal-Lubling Y, Guerrero-Romero D, Manzano Garcia R, Greenwood W, O’Reilly M, Georgopoulou D, Callari M, Lerda G, Wix S, Giovannetti A, Masina R, Esmaeilishirazifard E, Cope W, Martin AG, Nagano A, Young L, Kupczak S, Cheng Y, Bardwell H, Provenzano E, Kane J, Lay J, Grybowicz L, McAdam K, Caldas C, Abraham J, Rueda OM, Bruna A. Modeling Drug Responses and Evolutionary Dynamics Using Patient-Derived Xenografts Reveals Precision Medicine Strategies for Triple-Negative Breast Cancer. Cancer Res 2025; 85:567-584. [PMID: 39514406 PMCID: PMC7617242 DOI: 10.1158/0008-5472.can-24-1703] [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: 05/22/2024] [Revised: 08/09/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
The intertumor and intratumor heterogeneity of triple-negative breast cancers, which is reflected in diverse drug responses, interplays with tumor evolution. In this study, we developed a preclinical experimental and analytical framework using patient-derived tumor xenografts (PDTX) from patients with treatment-naïve triple-negative breast cancers to test their predictive value in personalized cancer treatment approaches. Patients and their matched PDTXs exhibited concordant drug responses to neoadjuvant therapy using two trial designs and dosing schedules. This platform enabled analysis of nongenetic mechanisms involved in relapse dynamics. Treatment resulted in permanent phenotypic changes, with functional and therapeutic consequences. High-throughput drug screening methods in ex vivo PDTX cells revealed patient-specific drug response changes dependent on first-line therapy. This was validated in vivo, as exemplified by a change in olaparib sensitivity in tumors previously treated with clinically relevant cycles of standard-of-care chemotherapy. In summary, PDTXs provide a robust tool to test patient drug responses and therapeutic regimens and to model evolutionary trajectories. However, high intermodel variability and permanent nongenomic transcriptional changes constrain their use for personalized cancer therapy. This work highlights important considerations associated with preclinical drug response modeling and potential uses of the platform to identify efficacious and preferential sequential therapeutic regimens. Significance: Patient-derived tumor xenografts from treatment-naïve breast cancer samples can predict patient drug responses and model treatment-induced phenotypic and functional evolution, making them valuable preclinical tools.
Collapse
Affiliation(s)
- Abigail Shea
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Institute of Cancer Research, London, United Kingdom
| | - Yaniv Eyal-Lubling
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Guerrero-Romero
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Raquel Manzano Garcia
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Wendy Greenwood
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Martin O’Reilly
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Dimitra Georgopoulou
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Maurizio Callari
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Fondazione Michelangelo, Milan, Italy
| | - Giulia Lerda
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Sophia Wix
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Agnese Giovannetti
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Clinical Genomics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Riccardo Masina
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Elham Esmaeilishirazifard
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Wei Cope
- Department of Histopathology, Cambridge University NHS Foundation Trust, Cambridge, United Kingdom
| | - Alistair G. Martin
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ai Nagano
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Lisa Young
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Steven Kupczak
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Yi Cheng
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Helen Bardwell
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Elena Provenzano
- Department of Histopathology, Cambridge University NHS Foundation Trust, Cambridge, United Kingdom
- Cambridge NIH Biomedical Research Centre, Cambridge, United Kingdom
| | - Justine Kane
- Department of Oncology, Precision Breast Cancer Institute, University of Cambridge, Cambridge, United Kingdom
| | - Jonny Lay
- Department of Oncology, Precision Breast Cancer Institute, University of Cambridge, Cambridge, United Kingdom
| | - Louise Grybowicz
- Cambridge University NHS Foundation Trust, Cambridge, United Kingdom
| | - Karen McAdam
- Cambridge University NHS Foundation Trust, Cambridge, United Kingdom
| | - Carlos Caldas
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Clinical Biochemistry, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Jean Abraham
- Department of Oncology, Precision Breast Cancer Institute, University of Cambridge, Cambridge, United Kingdom
| | - Oscar M. Rueda
- MRC-Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
123
|
Datta RR, Akdogan D, Tezcan EB, Onal P. Versatile roles of disordered transcription factor effector domains in transcriptional regulation. FEBS J 2025. [PMID: 39888268 DOI: 10.1111/febs.17424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/25/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
Transcription, a crucial step in the regulation of gene expression, is tightly controlled and involves several essential processes, such as chromatin organization, recognition of the specific genomic sequences, DNA binding, and ultimately recruiting the transcriptional machinery to facilitate transcript synthesis. At the center of this regulation are transcription factors (TFs), which comprise at least one DNA-binding domain (DBD) and an effector domain (ED). Although the structure and function of DBDs have been well studied, our knowledge of the structure and function of effector domains is limited. EDs are of particular importance in generating distinct transcriptional responses between protein members of the same TF family that have similar DBDs and specificities. The study of transcriptional activity conferred by effector domains has traditionally been conducted through examining protein-protein interactions. However, recent research has uncovered alternative mechanisms by which EDs regulate gene expression, such as the formation of condensates that increase the local concentration of transcription factors, cofactors, and coregulated genes, as well as DNA binding. Here, we provide a comprehensive overview of the known roles of transcription factor EDs, with a specific focus on disordered regions. Additionally, we emphasize the significance of intrinsically disordered regions (IDRs) during transcriptional regulation. We examine the mechanisms underlying the establishment and maintenance of transcriptional specificity through the structural properties of predominantly disordered EDs. We then provide a comprehensive overview of the current understanding of these domains, including their physical and chemical characteristics, as well as their functional roles.
Collapse
Affiliation(s)
| | - Dilan Akdogan
- Molecular Biology and Genetics Department, Ihsan Dogramaci Bilkent University, Ankara, Turkey
| | - Elif B Tezcan
- Molecular Biology and Genetics Department, Ihsan Dogramaci Bilkent University, Ankara, Turkey
| | - Pinar Onal
- Molecular Biology and Genetics Department, Ihsan Dogramaci Bilkent University, Ankara, Turkey
| |
Collapse
|
124
|
Ru Y, Ma M, Zhou X, Kriti D, Cohen N, D'Souza S, Schaniel C, Motch Perrine SM, Kuo S, Pichurin O, Pinto D, Housman G, Holmes G, Schadt E, van Bakel H, Zhang B, Jabs EW, Wu M. Integrated transcriptomic analysis of human induced pluripotent stem cell-derived osteogenic differentiation reveals a regulatory role of KLF16. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.11.579844. [PMID: 38405902 PMCID: PMC10888757 DOI: 10.1101/2024.02.11.579844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Osteogenic differentiation is essential for bone development, metabolism, and repair; however, the underlying regulatory relationships among genes remain poorly understood. To elucidate the transcriptomic changes and identify novel regulatory genes involved in osteogenic differentiation, we differentiated mesenchymal stem cells (MSCs) derived from 20 human iPSC lines into preosteoblasts (preOBs) and osteoblasts (OBs). We then performed transcriptome profiling of MSCs, preOBs and OBs. The iPSC-derived MSCs and OBs showed similar transcriptome profiles to those of primary human MSCs and OBs, respectively. Differential gene expression analysis revealed global changes in the transcriptomes from MSCs to preOBs, and then to OBs, including the differential expression of 840 genes encoding transcription factors (TFs). TF regulatory network analysis uncovered a network comprising 451 TFs, organized into five interactive modules. Multiscale embedded gene co-expression network analysis (MEGENA) identified gene co-expression modules and key network regulators (KNRs). From these analyses, KLF16 emerged as an important TF in osteogenic differentiation. We demonstrate that overexpression of Klf16 in vitro inhibited osteogenic differentiation and mineralization, while Klf16 +/- mice exhibited increased bone mineral density, trabecular number, and cortical bone area. Our study underscores the complexity of osteogenic differentiation and identifies novel regulatory genes such as KLF16, which plays an inhibitory role in osteogenic differentiation both in vitro and in vivo.
Collapse
Affiliation(s)
- Ying Ru
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Meng Ma
- Mount Sinai Genomics, Sema4, Stamford, CT, 06902, USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Divya Kriti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Present address: Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC V6T 2G3, Canada
| | - Ninette Cohen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Present address: Division of Cytogenetics and Molecular Pathology, Zucker School of Medicine at Hofstra/Northwell, Northwell Health Laboratories, Lake Success, NY, 11030, USA
| | - Sunita D'Souza
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Present address: St Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Christoph Schaniel
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Susan M Motch Perrine
- Department of Anthropology, Pennsylvania State University, University Park, PA, 16802, USA
| | - Sharon Kuo
- Department of Biomedical Sciences, University of Minnesota, Duluth, MN, 55812, USA
- Technological Primates Research Group, Max Planck Institute for Evolutionary Anthropology, Leipzig, 04103, Germany
| | - Oksana Pichurin
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Dalila Pinto
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Genevieve Housman
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary Anthropology, Leipzig, 04103, Germany
| | - Greg Holmes
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ethylin Wang Jabs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Meng Wu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
125
|
Li S, Noroozizadeh S, Moayedpour S, Kogler-Anele L, Xue Z, Zheng D, Montoya FU, Agarwal V, Bar-Joseph Z, Jager S. mRNA-LM: full-length integrated SLM for mRNA analysis. Nucleic Acids Res 2025; 53:gkaf044. [PMID: 39898548 PMCID: PMC11962594 DOI: 10.1093/nar/gkaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/07/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025] Open
Abstract
The success of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) messenger RNA (mRNA) vaccine has led to increased interest in the design and use of mRNA for vaccines and therapeutics. Still, selecting the most appropriate mRNA sequence for a protein remains a challenge. Several recent studies have shown that the specific mRNA sequence can have a significant impact on the translation efficiency, half-life, degradation rates, and other issues that play a major role in determining vaccine efficiency. To enable the selection of the most appropriate sequence, we developed mRNA-LM, an integrated small language model for modeling the entire mRNA sequence. mRNA-LM uses the contrastive language-image pretraining integration technology to combine three separate language models for the different mRNA segments. We trained mRNA-LM on millions of diverse mRNA sequences from several different species. The unsupervised model was able to learn meaningful biology related to evolution and host-pathogen interactions. Fine-tuning of mRNA-LM allowed us to use it in several mRNA property prediction tasks. As we show, using the full-length integrated model led to accurate predictions, improving on prior methods proposed for this task.
Collapse
Affiliation(s)
- Sizhen Li
- Digital R&D, Sanofi, Cambridge, MA 02141, United States
| | - Shahriar Noroozizadeh
- Digital R&D, Sanofi, Cambridge, MA 02141, United States
- Machine Learning Department, Carnegie Mellon University, Pittsburgh, PA 15213, United States
- Heinz College, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | | | | | - Zexin Xue
- Digital R&D, Sanofi, Cambridge, MA 02141, United States
- Department of Computer Science, University of Toronto, Toronto, ON M5S 2E4, Canada
| | - Dinghai Zheng
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, United States
| | | | - Vikram Agarwal
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, United States
| | | | - Sven Jager
- Digital R&D, Sanofi, Cambridge, MA 02141, United States
| |
Collapse
|
126
|
Wang S, Wang Z, Zang C. Genomic clustering tendency of transcription factors reflects phase-separated transcriptional condensates at super-enhancers. Nucleic Acids Res 2025; 53:gkaf015. [PMID: 39868536 PMCID: PMC11760973 DOI: 10.1093/nar/gkaf015] [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: 09/18/2023] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025] Open
Abstract
Many transcription factors (TFs) have been shown to bind to super-enhancers, forming transcriptional condensates to activate transcription in various cellular systems. However, the genomic and epigenomic determinants of phase-separated transcriptional condensate formation remain poorly understood. Questions regarding which TFs tend to associate with transcriptional condensates and what factors influence their association are largely unanswered. Here we systematically analyzed 571 DNA sequence motifs across the human genome and 6650 TF binding profiles across different cell types to identify the molecular features contributing to the formation of transcriptional condensates. We found that the genomic distributions of sequence motifs for different TFs exhibit distinct clustering tendencies. Notably, TF motifs with a high genomic clustering tendency are significantly associated with super-enhancers. TF binding profiles showing a high genomic clustering tendency are further enriched at cell-type-specific super-enhancers. TFs with a high binding clustering tendency also possess high liquid-liquid phase separation abilities. Compared to nonclustered TF binding, densely clustered TF binding sites are more enriched at cell-type-specific super-enhancers with higher chromatin accessibility, elevated chromatin interaction and stronger association with cancer outcomes. Our results indicate that the clustered genomic binding patterns and the phase separation properties of TFs collectively contribute to the formation of transcriptional condensates.
Collapse
Affiliation(s)
- Shengyuan Wang
- Department of Genome Sciences, University of Virginia, PO Box 800717, Charlottesville, VA 22908, USA
| | - Zhenjia Wang
- Department of Genome Sciences, University of Virginia, PO Box 800717, Charlottesville, VA 22908, USA
| | - Chongzhi Zang
- Department of Genome Sciences, University of Virginia, PO Box 800717, Charlottesville, VA 22908, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, PO Box 800733, Charlottesville, VA 22908, USA
- Department of Biomedical Engineering, University of Virginia, PO Box 800759, Charlottesville, VA 22908, USA
- UVA Comprehensive Cancer Center, University of Virginia, PO Box 800334, Charlottesville, VA 22908, USA
| |
Collapse
|
127
|
Lan YZ, Wu Z, Chen WJ, Yu XN, Wu HT, Liu J. Sine oculis homeobox homolog family function in gastrointestinal cancer: Progression and comprehensive analysis. World J Clin Oncol 2025; 16:97163. [PMID: 39867730 PMCID: PMC11528897 DOI: 10.5306/wjco.v16.i1.97163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/20/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024] Open
Abstract
The sine oculis homeobox homolog (SIX) family, a group of transcription factors characterized by a conserved DNA-binding homology domain, plays a critical role in orchestrating embryonic development and organogenesis across various organisms, including humans. Comprising six distinct members, from SIX1 to SIX6, each member contributes uniquely to the development and differentiation of diverse tissues and organs, underscoring the versatility of the SIX family. Dysregulation or mutations in SIX genes have been implicated in a spectrum of developmental disorders, as well as in tumor initiation and progression, highlighting their pivotal role in maintaining normal developmental trajectories and cellular functions. Efforts to target the transcriptional complex of the SIX gene family have emerged as a promising strategy to inhibit tumor development. While the development of inhibitors targeting this gene family is still in its early stages, the significant potential of such interventions holds promise for future therapeutic advances. Therefore, this review aimed to comprehensively explore the advancements in understanding the SIX family within gastrointestinal cancers, focusing on its critical role in normal organ development and its implications in gastrointestinal cancers, including gastric, pancreatic, colorectal cancer, and hepatocellular carcinomas. In conclusion, this review deepened the understanding of the functional roles of the SIX family and explored the potential of utilizing this gene family for the diagnosis, prognosis, and treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Yang-Zheng Lan
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Zheng Wu
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Wen-Jia Chen
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Xin-Ning Yu
- Department of General Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Hua-Tao Wu
- Department of General Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Jing Liu
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
128
|
Andersen RE, Talukdar M, Sakamoto T, Song JH, Qian X, Lee S, Delgado RN, Zhao S, Eichfeld G, Harms J, Walsh CA. Autism-Associated Genes and Neighboring lncRNAs Converge on Key Gene Regulatory Networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.634000. [PMID: 39896631 PMCID: PMC11785016 DOI: 10.1101/2025.01.20.634000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The diversity of genes implicated in autism spectrum disorder (ASD) creates challenges for identifying core pathophysiological mechanisms. Aggregation of seven different classes of genetic variants implicated in ASD, in a database we call Consensus-ASD, reveals shared features across distinct types of ASD variants. Functional interrogation of 19 ASD genes and 9 neighboring long non-coding RNAs (lncRNAs) using CRISPR-Cas13 strikingly revealed differential gene expression profiles that were significantly enriched for other ASD genes. Furthermore, construction of a gene regulatory network (GRN) enabled the identification of central regulators that exhibit convergently altered activity upon ASD gene disruption. Thus, this study reveals how perturbing distinct ASD-associated genes can lead to shared, broad dysregulation of GRNs with critical relevance to ASD. This provides a crucial framework for understanding how diverse genes, including lncRNAs, can play convergent roles in key neurodevelopmental processes and ultimately contribute to ASD.
Collapse
Affiliation(s)
- Rebecca E. Andersen
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Allen Discovery Center for Human Brain Evolution, Boston, MA, USA
| | - Maya Talukdar
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT MD/PhD Program, Program in Biomedical Informatics, Boston, MA, USA
| | - Tyler Sakamoto
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Janet H.T. Song
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Allen Discovery Center for Human Brain Evolution, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| | - Xuyu Qian
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Allen Discovery Center for Human Brain Evolution, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| | - Seungil Lee
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Ryan N. Delgado
- Department of Genetics, Blavatnik Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Sijing Zhao
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Harvard BBS PhD Program, Boston, MA, USA
| | - Gwenyth Eichfeld
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Colgate University, Hamilton, NY, USA
| | - Julia Harms
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- University of California Berkeley, Berkeley, CA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics and Manton Center for Orphan Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Allen Discovery Center for Human Brain Evolution, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA, USA
| |
Collapse
|
129
|
Cihan M, Schmauck G, Sprang M, Andrade-Navarro MA. Unveiling cell-type-specific microRNA networks through alternative polyadenylation in glioblastoma. BMC Biol 2025; 23:15. [PMID: 39838429 PMCID: PMC11752630 DOI: 10.1186/s12915-024-02104-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is characterized by its cellular complexity, with a microenvironment consisting of diverse cell types, including oligodendrocyte precursor cells (OPCs) and neoplastic CD133 + radial glia-like cells. This study focuses on exploring the distinct cellular transitions in GBM, emphasizing the role of alternative polyadenylation (APA) in modulating microRNA-binding and post-transcriptional regulation. RESULTS Our research identified unique APA profiles that signify the transitional phases between neoplastic cells and OPCs, underscoring the importance of APA in cellular identity and transformation in GBM. A significant finding was the disconnection between differential APA events and gene expression alterations, indicating that APA operates as an independent regulatory mechanism. We also highlighted the specific genes in neoplastic cells and OPCs that lose microRNA-binding sites due to APA, which are crucial for maintaining stem cell characteristics and DNA repair, respectively. The constructed networks of microRNA-transcription factor-target genes provide insights into the cellular mechanisms influencing cancer cell survival and therapeutic resistance. CONCLUSIONS This study elucidates the APA-driven regulatory framework within GBM, spotlighting its influence on cell state transitions and microRNA network dynamics. Our comprehensive analysis using single-cell RNA sequencing data to investigate the microRNA-binding sites altered by APA profiles offers a robust foundation for future research, presenting a novel approach to understanding and potentially targeting the complex molecular interplay in GBM.
Collapse
Affiliation(s)
- Mert Cihan
- Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Greta Schmauck
- Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Maximilian Sprang
- Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | | |
Collapse
|
130
|
Yang Z, Kan W, Wang Z, Tang C, Cheng Y, Wang D, Gao Y, Wu L. Genome-wide identification and expression analysis of phytochrome gene family in Aikang58 wheat ( Triticum aestivum L.). FRONTIERS IN PLANT SCIENCE 2025; 15:1520457. [PMID: 39906238 PMCID: PMC11790602 DOI: 10.3389/fpls.2024.1520457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/27/2024] [Indexed: 02/06/2025]
Abstract
Phytochromes are essential photoreceptors in plants that sense red and far-red light, playing a vital role in regulating plant growth and development through light signal transduction. Despite extensive research on phytochromes in model plants like Arabidopsis and rice, they have received relatively little attention in wheat. In this study, we employed bioinformatics methods to identify eight TaAkPHY genes in the Aikang58 wheat variety. Based on gene structure, conserved domains, and phylogenetic relationships, the TaAkPHY gene family exhibits a high degree of conservation. Synteny analysis revealed the evolutionary history of the PHY genes in Aikang58 and Chinese Spring wheat (Triticum aestivum L.), barley (Hordeum vulgare L.), rice (Oryza sativa L.), maize (Zea mays L.), quinoa (Chenopodium quinoa Willd.), soybean [Glycine max (L.) Merr.], and Arabidopsis [Arabidopsis thaliana (L.) Heynh.]. Among these species, wheat is most closely related to barley, followed by rice and maize. The cis-acting element analysis indicates that the promoter regions of TaAkPHY genes contain a large number of CAT-box, CGTCA-motif, GC-motif, etc., which are mainly involved in plant development, hormone response, and stress response. Gene expression profiling demonstrated that TaAkPHY genes exhibit varying expression levels across different tissues and are induced by various stress conditions and plant hormone treatments. Co-expression network analysis suggested that TaAkPHY genes may specifically regulate downstream genes associated with stress responses, chloroplast development, and circadian rhythms. Additionally, the least absolute shrinkage and selection operator (LASSO) regression algorithm in machine learning was used to screen transcription factors such as bHLH, WRKY, and MYB that influenced the expression of TaAkPHY genes. This method helps to quickly extract key influencing factors from a large amount of complex data. Overall, these findings provide new insights into the role of phytochromes in wheat growth, development, and stress responses, laying a foundation for future research on phytochromes in wheat.
Collapse
Affiliation(s)
- Zhu Yang
- Science Island Branch, University of Science and Technology of China, Hefei, Anhui, China
- The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Wenjie Kan
- Science Island Branch, University of Science and Technology of China, Hefei, Anhui, China
- The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Ziqi Wang
- The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Caiguo Tang
- The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Yuan Cheng
- Science Island Branch, University of Science and Technology of China, Hefei, Anhui, China
- The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Dacheng Wang
- Science Island Branch, University of Science and Technology of China, Hefei, Anhui, China
- The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Yameng Gao
- The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Lifang Wu
- Science Island Branch, University of Science and Technology of China, Hefei, Anhui, China
- The Center for Ion Beam Bioengineering & Green Agriculture, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| |
Collapse
|
131
|
Zhou W, Fang J, Jia Q, Meng H, Liu F, Mao J. Transcription factor specificity protein (SP) family in renal physiology and diseases. PeerJ 2025; 13:e18820. [PMID: 39850832 PMCID: PMC11756367 DOI: 10.7717/peerj.18820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 12/15/2024] [Indexed: 01/25/2025] Open
Abstract
Dysregulated specificity proteins (SPs), members of the C2H2 zinc-finger family, are crucial transcription factors (TFs) with implications for renal physiology and diseases. This comprehensive review focuses on the role of SP family members, particularly SP1 and SP3, in renal physiology and pathology. A detailed analysis of their expression and cellular localization in the healthy human kidney is presented, highlighting their involvement in fatty acid metabolism, electrolyte regulation, and the synthesis of important molecules. The review also delves into the diverse roles of SPs in various renal diseases, including renal ischemia/reperfusion injury, diabetic nephropathy, renal interstitial fibrosis, and lupus nephritis, elucidating their molecular mechanisms and potential as therapeutic targets. The review further discusses pharmacological modulation of SPs and its implications for treatment. Our findings provide a comprehensive understanding of SPs in renal health and disease, offering new avenues for targeted therapeutic interventions and precision medicine in nephrology.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jiaxi Fang
- Department of Ultrasound, Taizhou Central Hospital, Taizhou, Zhejiang, China
| | - Qingqing Jia
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Hanyan Meng
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Fei Liu
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jianhua Mao
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| |
Collapse
|
132
|
Liu Y, Rao S, Hoskins I, Geng M, Zhao Q, Chacko J, Ghatpande V, Qi K, Persyn L, Wang J, Zheng D, Zhong Y, Park D, Cenik ES, Agarwal V, Ozadam H, Cenik C. Translation efficiency covariation across cell types is a conserved organizing principle of mammalian transcriptomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.11.607360. [PMID: 39149359 PMCID: PMC11326257 DOI: 10.1101/2024.08.11.607360] [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: 08/17/2024]
Abstract
Characterization of shared patterns of RNA expression between genes across conditions has led to the discovery of regulatory networks and novel biological functions. However, it is unclear if such coordination extends to translation, a critical step in gene expression. Here, we uniformly analyzed 3,819 ribosome profiling datasets from 117 human and 94 mouse tissues and cell lines. We introduce the concept of Translation Efficiency Covariation (TEC), identifying coordinated translation patterns across cell types. We nominate potential mechanisms driving shared patterns of translation regulation. TEC is conserved across human and mouse cells and helps uncover gene functions. Moreover, our observations indicate that proteins that physically interact are highly enriched for positive covariation at both translational and transcriptional levels. Our findings establish translational covariation as a conserved organizing principle of mammalian transcriptomes.
Collapse
Affiliation(s)
- Yue Liu
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Shilpa Rao
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Ian Hoskins
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Michael Geng
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Qiuxia Zhao
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jonathan Chacko
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Vighnesh Ghatpande
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Kangsheng Qi
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Logan Persyn
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jun Wang
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA
| | - Dinghai Zheng
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA
| | - Yochen Zhong
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Dayea Park
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Elif Sarinay Cenik
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Vikram Agarwal
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA
| | - Hakan Ozadam
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
- Present address: Sail Biomedicines, Cambridge, MA, 02141, USA
| | | |
Collapse
|
133
|
Song W, Ovcharenko I. Abundant repressor binding sites in human enhancers are associated with the fine-tuning of gene regulation. iScience 2025; 28:111658. [PMID: 39868043 PMCID: PMC11761325 DOI: 10.1016/j.isci.2024.111658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/04/2024] [Accepted: 11/25/2024] [Indexed: 01/28/2025] Open
Abstract
The regulation of gene expression relies on the coordinated action of transcription factors (TFs) at enhancers, including both activator and repressor TFs. We employed deep learning (DL) to dissect HepG2 enhancers into positive (PAR), negative (NAR), and neutral activity regions. Sharpr-MPRA and STARR-seq highlight the dichotomy impact of NARs and PARs on modulating and catalyzing the activity of enhancers, respectively. Approximately 22% of HepG2 enhancers, termed "repressive impact enhancers" (RIEs), are predominantly populated by NARs and transcriptional repression motifs. Genes flanking RIEs exhibit a stage-specific decline in expression during late development, suggesting RIEs' role in trimming enhancer activities. About 16.7% of human NARs emerge from neutral rhesus macaque DNA. This gain of repressor binding sites in RIEs is associated with a 30% decrease in the average expression of flanking genes in humans compared to rhesus macaque. Our work reveals modulated enhancer activity and adaptable gene regulation through the evolutionary dynamics of TF binding sites.
Collapse
Affiliation(s)
- Wei Song
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Ivan Ovcharenko
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
134
|
Vo K, Sharma Y, Chakravarthi VP, Mohamadi R, Bahadursingh ES, Mohamadi A, Dahiya V, Rosales CY, Pei GJ, Fields PE, Rumi MAK. Altered Expression of Epigenetic and Transcriptional Regulators in ERβ Knockout Rat Ovaries During Postnatal Development. Int J Mol Sci 2025; 26:760. [PMID: 39859473 PMCID: PMC11765817 DOI: 10.3390/ijms26020760] [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: 11/15/2024] [Revised: 01/04/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
We analyzed the transcriptome data of wildtype and estrogen receptor β knockout (ErβKO) rat ovaries during the early postnatal period and detected remarkable changes in epigenetic regulators and transcription factors. Compared with postnatal day (PD) 4.5 ovaries, PD 6.5 wildtype ovaries possessed 581 differentially expressed downstream transcripts (DEDTs), including 17 differentially expressed epigenetic regulators (DEERs) and 23 differentially expressed transcription factors (DETFs). Subsequently, compared with PD 6.5 ovaries, PD 8.5 wildtype ovaries showed 920 DEDTs, including 24 DEERs and 68 DETFs. The DEDTs, DEERs, and DETFs in wildtype ovaries represented the gene expression during primordial follicle activation and the gradual development of primary follicles of first-wave origin because the second-wave follicles remained dormant during this developmental period. When we compared the transcriptome data of age-matched ErβKO ovaries, we observed that PD 6.5 ErβKO ovaries had 744 DEDTs compared with PD 4.5 ovaries, including 46 DEERs and 55 DETFs. The loss of ERβ rapidly activated the primordial follicles of both first- and second-wave origin on PD 6.5 and showed a remarkable increase in DEDTs (744 vs. 581). However, compared with PD 6.5 ovaries, PD 8.5 ErβKO ovaries showed only 191 DEDTs, including 8 DEERs and 10 DETFs. This finding suggests that the PD 8.5 ErβKO ovaries did not undergo remarkable ovarian follicle activation greater than that had already occurred in PD 6.5 ErβKO ovaries. The results also showed that the numbers of DEERs and DETFs were associated with increased changes in DEDTs; the greater the number of DEERs or DETFs, the larger the number of DEDTs. In addition to the quantitative differences in DEERs and DETFs between the wildtype and ErβKO ovaries, the differentially expressed regulators showed distinct patterns. We identified that 17 transcripts were tied to follicle assembly, 6 to follicle activation, and 12 to steroidogenesis. Our observations indicate that a loss of ERβ dysregulates the epigenetic regulators and transcription factors in ErβKO ovaries, which disrupts the downstream genes in ovarian follicles and increases follicle activation. Further studies are required to clarify if ERβ directly or indirectly regulates DEDTs, including DEERs and DETFs, during the neonatal development of rat ovarian follicles.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (K.V.); (Y.S.); (V.P.C.); (R.M.); (E.S.B.); (A.M.); (V.D.); (C.Y.R.); (P.E.F.)
| |
Collapse
|
135
|
Zhao F, Jiang X, Li Y, Huang T, Xiahou Z, Nie W, Li Q. Characterizing tumor biology and immune microenvironment in high-grade serous ovarian cancer via single-cell RNA sequencing: insights for targeted and personalized immunotherapy strategies. Front Immunol 2025; 15:1500153. [PMID: 39896800 PMCID: PMC11782144 DOI: 10.3389/fimmu.2024.1500153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
Background High-grade serous ovarian cancer (HGSOC), the predominant subtype of epithelial ovarian cancer, is frequently diagnosed at an advanced stage due to its nonspecific early symptoms. Despite standard treatments, including cytoreductive surgery and platinum-based chemotherapy, significant improvements in survival have been limited. Understanding the molecular mechanisms, immune landscape, and drug sensitivity of HGSOC is crucial for developing more effective and personalized therapies. This study integrates insights from cancer immunology, molecular profiling, and drug sensitivity analysis to identify novel therapeutic targets and improve treatment outcomes. Utilizing single-cell RNA sequencing (scRNA-seq), the study systematically examines tumor heterogeneity and immune microenvironment, focusing on biomarkers influencing drug response and immune activity, aiming to enhance patient outcomes and quality of life. Methods scRNA-seq data was obtained from the GEO database in this study. Differential gene expression was analyzed using gene ontology and gene set enrichment methods. InferCNV identified malignant epithelial cells, while Monocle, Cytotrace, and Slingshot software inferred subtype differentiation trajectories. The CellChat software package predicted cellular communication between malignant cell subtypes and other cells, while pySCENIC analysis was utilized to identify transcription factor regulatory networks within malignant cell subtypes. Finally, the analysis results were validated through functional experiments, and a prognostic model was developed to assess prognosis, immune infiltration, and drug sensitivity across various risk groups. Results This study investigated the cellular heterogeneity of HGSOC using scRNA-seq, focusing on tumor cell subtypes and their interactions within the tumor microenvironment. We confirmed the key role of the C2 IGF2+ tumor cell subtype in HGSOC, which was significantly associated with poor prognosis and high levels of chromosomal copy number variations. This subtype was located at the terminal differentiation of the tumor, displaying a higher degree of malignancy and close association with stage IIIC tissue types. The C2 subtype was also associated with various metabolic pathways, such as glycolysis and riboflavin metabolism, as well as programmed cell death processes. The study highlighted the complex interactions between the C2 subtype and fibroblasts through the MK signaling pathway, which may be closely related to tumor-associated fibroblasts and tumor progression. Elevated expression of PRRX1 was significantly connected to the C2 subtype and may impact disease progression by modulating gene transcription. A prognostic model based on the C2 subtype demonstrated its association with adverse prognosis outcomes, emphasizing the importance of immune infiltration and drug sensitivity analysis in clinical intervention strategies. Conclusion This study integrates molecular oncology, immunotherapy, and drug sensitivity analysis to reveal the mechanisms driving HGSOC progression and treatment resistance. The C2 IGF2+ tumor subtype, linked to poor prognosis, offers a promising target for future therapies. Emphasizing immune infiltration and drug sensitivity, the research highlights personalized strategies to improve survival and quality of life for HGSOC patients.
Collapse
MESH Headings
- Female
- Tumor Microenvironment/immunology
- Tumor Microenvironment/genetics
- Humans
- Single-Cell Analysis
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/therapy
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/pathology
- Precision Medicine
- Immunotherapy/methods
- Biomarkers, Tumor/genetics
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/immunology
- Cystadenocarcinoma, Serous/therapy
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/mortality
- Gene Expression Regulation, Neoplastic
- Sequence Analysis, RNA
- Neoplasm Grading
- Gene Expression Profiling
- Carcinoma, Ovarian Epithelial/genetics
- Carcinoma, Ovarian Epithelial/immunology
- Carcinoma, Ovarian Epithelial/therapy
- Transcriptome
Collapse
Affiliation(s)
- Fu Zhao
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaojing Jiang
- Affiliated Hospital of Shandong Academy of Traditional Chinese Medicine, Jinan, China
| | - Yumeng Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianjiao Huang
- The First School of Clinical Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Wenyang Nie
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Li
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
136
|
Zhang S, Deng S, Liu J, Liu S, Chen Z, Liu S, Xue C, Zeng L, Zhao H, Xu Z, Zhao S, Zhou Y, Peng X, Wu X, Bai R, Wu S, Li M, Zheng J, Lin D, Zhang J, Huang X. Targeting MXD1 sensitises pancreatic cancer to trametinib. Gut 2025:gutjnl-2024-333408. [PMID: 39819860 DOI: 10.1136/gutjnl-2024-333408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/29/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND The resistance of pancreatic ductal adenocarcinoma (PDAC) to trametinib therapy limits its clinical use. However, the molecular mechanisms underlying trametinib resistance in PDAC remain unclear. OBJECTIVE We aimed to illustrate the mechanisms of resistance to trametinib in PDAC and identify trametinib resistance-associated druggable targets, thus improving the treatment efficacy of trametinib-resistant PDAC. DESIGN We established patient-derived xenograft (PDX) models and primary cell lines to conduct functional experiments. We also applied single-cell RNA sequencing, Assay for Transposase-accessible Chromatin with sequencing and Cleavage Under Targets and Tagmentation sequencing to explore the relevant molecular mechanism. RESULTS We have identified a cancer cell subpopulation featured by hyperactivated viral mimicry response in trametinib-resistant PDXs. We have demonstrated that trametinib treatment of PDAC PDXs induces expression of transcription factor MAX dimerisation protein 1 (MXD1), which acts as a cofactor of histone methyltransferase mixed lineage leukaemia 1 to increased H3K4 trimethylation in transposable element (TE) loci, enhancing chromatin accessibility and thus the transcription of TEs. Mechanistically, enhanced transcription of TEs produces excessive double-stranded RNAs, leading to the activation of viral mimicry response and downstream oncogenic interferon-stimulated genes. Inhibiting MXD1 expression can recover the drug vulnerability of trametinib-resistant PDAC cells to trametinib. CONCLUSIONS Our study has discovered an important mechanism for trametinib resistance and identified MXD1 as a druggable target in treatment of trametinib-resistant PDAC.
Collapse
Affiliation(s)
- Shaoping Zhang
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shuang Deng
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ji Liu
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shuang Liu
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ziming Chen
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shaoqiu Liu
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chunling Xue
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Lingxing Zeng
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Hongzhe Zhao
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zilan Xu
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Sihan Zhao
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yifan Zhou
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xinyi Peng
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xiaoyu Wu
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ruihong Bai
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shaojia Wu
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Mei Li
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jian Zheng
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Dongxin Lin
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jialiang Zhang
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xudong Huang
- State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
137
|
Pagani F, Orzan F, Lago S, De Bacco F, Prelli M, Cominelli M, Somenza E, Gryzik M, Balzarini P, Ceresa D, Marubbi D, Isella C, Crisafulli G, Poli M, Malatesta P, Galli R, Ronca R, Zippo A, Boccaccio C, Poliani PL. Concurrent RB1 and P53 pathway disruption predisposes to the development of a primitive neuronal component in high-grade gliomas depending on MYC-driven EBF3 transcription. Acta Neuropathol 2025; 149:8. [PMID: 39821672 DOI: 10.1007/s00401-025-02845-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
The foremost feature of glioblastoma (GBM), the most frequent malignant brain tumours in adults, is a remarkable degree of intra- and inter-tumour heterogeneity reflecting the coexistence within the tumour bulk of different cell populations displaying distinctive genetic and transcriptomic profiles. GBM with primitive neuronal component (PNC), recently identified by DNA methylation-based classification as a peculiar GBM subtype (GBM-PNC), is a poorly recognized and aggressive GBM variant characterised by nodules containing cells with primitive neuronal differentiation along with conventional GBM areas. In addition, the presence of a PNC component has been also reported in IDH-mutant high-grade gliomas (HGGs), and to a lesser extent to other HGGs, suggesting that regardless from being IDH-mutant or IDH-wildtype, peculiar genetic and/or epigenetic events may contribute to the phenotypic skewing with the emergence of the PNC phenotype. However, a clear hypothesis on the mechanisms responsible for this phenotypic skewing is still lacking. We assumed that the biphasic nature of these entities represents a unique model to investigate the relationships between genetic alterations and their phenotypic manifestations. In this study we show that in HGGs with PNC features both components are highly enriched in genetic alterations directly causing cell cycle deregulation (RB inactivation or CDK4 amplification) and p53 pathway inactivation (TP53 mutations or MDM2/4 amplification). However, the PNC component displays further upregulation of transcriptional pathways associated with proliferative activity, including overexpression of MYC target genes. Notably, the PNC phenotype relies on the expression of EBF3, an early neurogenic transcription factor, which is directly controlled by MYC transcription factors in accessible chromatin sites. Overall our findings indicate that the concomitant presence of genetic alterations, impinging on both cell cycle and p53 pathway control, strongly predisposes GBM to develop a concomitant poorly differentiated primitive phenotype depending on MYC-driven EBF3 transcription in a subset of glioma stem-like progenitor cells.
Collapse
Affiliation(s)
- Francesca Pagani
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francesca Orzan
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060, Turin, Italy
- Department of Oncology, University of Turin Medical School, Candiolo, 10060, Turin, Italy
| | - Sara Lago
- Laboratory for Chromatin Biology and Epigenetics, CIBIO-Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Francesca De Bacco
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060, Turin, Italy
- Department of Oncology, University of Turin Medical School, Candiolo, 10060, Turin, Italy
| | - Marta Prelli
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060, Turin, Italy
- Department of Oncology, University of Turin Medical School, Candiolo, 10060, Turin, Italy
| | - Manuela Cominelli
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elena Somenza
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Experimental Oncology and Immunology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Magdalena Gryzik
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Biochemistry Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Piera Balzarini
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Davide Ceresa
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela Marubbi
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy
| | - Claudio Isella
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060, Turin, Italy
- Department of Oncology, University of Turin Medical School, Candiolo, 10060, Turin, Italy
| | | | - Maura Poli
- Biochemistry Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Paolo Malatesta
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy
| | - Rossella Galli
- Neural Stem Cell Biology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Roberto Ronca
- Experimental Oncology and Immunology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessio Zippo
- Laboratory for Chromatin Biology and Epigenetics, CIBIO-Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Carla Boccaccio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060, Turin, Italy
- Department of Oncology, University of Turin Medical School, Candiolo, 10060, Turin, Italy
| | - Pietro Luigi Poliani
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
- Pathology Unit, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
138
|
Feng X, Gao Y, Chu F, Shan Y, Liu M, Wang Y, Zhu Y, Lu Q, Li M. Cortical arealization of interneurons defines shared and distinct molecular programs in developing human and macaque brains. Nat Commun 2025; 16:672. [PMID: 39809789 PMCID: PMC11733295 DOI: 10.1038/s41467-025-56058-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Cortical interneurons generated from ganglionic eminence via a long-distance journey of tangential migration display evident cellular and molecular differences across brain regions, which seeds the heterogeneous cortical circuitry in primates. However, whether such regional specifications in interneurons are intrinsically encoded or gained through interactions with the local milieu remains elusive. Here, we recruit 685,692 interneurons from cerebral cortex and subcortex including ganglionic eminence within the developing human and macaque species. Our integrative and comparative analyses reveal that less transcriptomic alteration is accompanied by interneuron migration within the ganglionic eminence subdivisions, in contrast to the dramatic changes observed in cortical tangential migration, which mostly characterize the transcriptomic specification for different destinations and for species divergence. Moreover, the in-depth survey of temporal regulation illustrates species differences in the developmental dynamics of cell types, e.g., the employment of CRH in primate interneurons during late-fetal stage distinguishes from their postnatal emergence in mice, and our entropy quantifications manifest the interneuron diversities gradually increase along the developmental ages in human and macaque cerebral cortices. Overall, our analyses depict the spatiotemporal features appended to cortical interneurons, providing a new proxy for understanding the relationship between cellular diversity and functional progression.
Collapse
Affiliation(s)
- Xiangling Feng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingjie Gao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Chu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuwen Shan
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meicheng Liu
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaoyi Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingfeng Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
- Innovation center for Brain Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
139
|
Huisman BD, Michelson DA, Rubin SA, Kohlsaat K, Gomarga W, Fang Y, Lee JM, Del Nido P, Nathan M, Benoist C, Zon L, Mathis D. Cross-species analyses of thymic mimetic cells reveal evolutionarily ancient origins and both conserved and species-specific elements. Immunity 2025; 58:108-123.e7. [PMID: 39731911 PMCID: PMC11735279 DOI: 10.1016/j.immuni.2024.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/19/2024] [Accepted: 11/27/2024] [Indexed: 12/30/2024]
Abstract
Thymic mimetic cells are molecular hybrids between medullary-thymic-epithelial cells (mTECs) and diverse peripheral cell types. They are involved in eliminating autoreactive T cells and can perform supplementary functions reflective of their peripheral-cell counterparts. Current knowledge about mimetic cells derives largely from mouse models. To provide the high resolution that proved revelatory for mice, we performed single-cell RNA sequencing on purified mimetic-cell compartments from human pediatric donors. The single-cell profiles of individual donors were surprisingly similar, with diversification of neuroendocrine subtypes and expansion of the muscle subtype relative to mice. Informatic and imaging studies on the muscle-mTEC population highlighted a maturation trajectory suggestive of skeletal-muscle differentiation, some striated structures, and occasional cellular groupings reminiscent of neuromuscular junctions. We also profiled thymic mimetic cells from zebrafish. Integration of data from the three species identified species-specific adaptations but substantial interspecies conservation, highlighting the evolutionarily ancient nature of mimetic mTECs. Our findings provide a landscape view of human mimetic cells, with anticipated relevance in autoimmunity.
Collapse
Affiliation(s)
- Brooke D Huisman
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Daniel A Michelson
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA; PhD Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Sara A Rubin
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA; PhD Program in Immunology, Harvard Medical School, Boston, MA, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Katherine Kohlsaat
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wilson Gomarga
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Yuan Fang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Ji Myung Lee
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro Del Nido
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Meena Nathan
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | | | - Leonard Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Howard Hughes Medical Institute and Boston Children's Hospital, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
140
|
Woyciehowsky M, Larson P, Stephan AR, Dandridge SL, Idonije D, Berg KA, Lanthier A, Acuna SA, Stites SW, Gebhardt WJ, Holtzen SE, Rakshit A, Palmer AE. Systematic characterization of zinc in a series of breast cancer cell lines reveals significant changes in zinc homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.11.632547. [PMID: 39868107 PMCID: PMC11761790 DOI: 10.1101/2025.01.11.632547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
An optimal amount of labile zinc (Zn 2+ ) is essential for proliferation of human cells, where Zn 2+ levels that are too high or too low cause cell cycle exit. Tumors of the breast have been characterized by high levels of total Zn 2+ . Given the role of Zn 2+ in proliferation of human cells and elevation of zinc in breast cancer tumors, we examined the concentration of total and labile Zn 2+ across a panel of 5 breast cancer cell lines, compared to the normal MCF10A cell line. We found that three cell lines (MDA-MB-231, MDA-MB-157, and SK-Br-3) showed elevated labile Zn 2+ in the cytosol, while T-47D showed significantly lower Zn 2+ , and MCF7 showed no change compared to MCF10A cells. There was no change in total Zn 2+ across the cell lines, as measured by ICP-MS, but we did observe a difference in the cells ability to accumulate Zn 2+ when Zn 2+ in the media was elevated. Therefore, we examined how proliferation of each cell line was affected by increases and decreases in the media. We found striking differences, where three cancer cell lines (MDA-MB-231, MDA-MB-157, and MCF7) showed robust proliferation in high Zn 2+ at concentrations that killed MCF10A, T-47D, and SK-Br-3 cells. We also discovered that 4 of the 5 cancer cell lines demonstrate compromised proliferation and increased cell death in low Zn 2+ , suggesting these cells may be addicted to Zn 2+ . Overall, our study suggests significant differences in Zn 2+ homeostasis and regulation in different types of breast cancer cells, with consequences for both proliferation and cell viability.
Collapse
|
141
|
Desterke C, Fu Y, Bonifacio-Mundaca J, Monge C, Pineau P, Mata-Garrido J, Francés R. Single-Cell RNA Sequencing Reveals LEF1-Driven Wnt Pathway Activation as a Shared Oncogenic Program in Hepatoblastoma and Medulloblastoma. Curr Oncol 2025; 32:35. [PMID: 39851951 PMCID: PMC11763369 DOI: 10.3390/curroncol32010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
(1) Background: Hepatoblastoma and medulloblastoma are two types of pediatric tumors with embryonic origins. Both tumor types can exhibit genetic alterations that affect the β-catenin and Wnt pathways; (2) Materials and Methods: This study used bioinformatics and integrative analysis of multi-omics data at both the tumor and single-cell levels to investigate two distinct pediatric tumors: medulloblastoma and hepatoblastoma; (3) Results: The cross-transcriptome analysis revealed a commonly regulated expression signature between hepatoblastoma and medulloblastoma tumors. Among the commonly upregulated genes, the transcription factor LEF1 was significantly expressed in both tumor types. In medulloblastoma, LEF1 upregulation is associated with the WNT-subtype. The analysis of LEF1 genome binding occupancy in H1 embryonic stem cells identified 141 LEF1 proximal targets activated in WNT medulloblastoma, 13 of which are involved in Wnt pathway regulation: RNF43, LEF1, NKD1, AXIN2, DKK4, DKK1, LGR6, FGFR2, NXN, TCF7L1, STK3, YAP1, and NFATC4. The ROC curve analysis of the combined expression of these 13 WNT-related LEF1 targets yielded an area under the curve (AUC) of 1.00, indicating 100% specificity and sensitivity for predicting the WNT subtype in the PBTA medulloblastoma cohort. An expression score based on these 13 WNT-LEF1 targets accurately predicted the WNT subtype in two independent medulloblastoma transcriptome cohorts. At the single-cell level, the WNT-LEF1 expression score was exclusively positive in WNT-medulloblastoma tumor cells. This WNT-LEF1-dependent signature was also confirmed as activated in the hepatoblastoma tumor transcriptome. At the single-cell level, the WNT-LEF1 expression score was higher in tumor cells from both human hepatoblastoma samples and a hepatoblastoma patient-derived xenotransplant model; (4) Discussion: This study uncovered a shared transcriptional activation of a LEF1-dependent embryonic program, which orchestrates the regulation of the Wnt signaling pathway in tumor cells from both hepatoblastoma and medulloblastoma.
Collapse
Affiliation(s)
- Christophe Desterke
- Faculté de Médecine du Kremlin Bicêtre, Université Paris-Saclay, INSERM UMRS-1310, 94805 Villejuif, France;
| | - Yuanji Fu
- Institut Necker Enfants Malades, INSERM, CNRS, Université Paris Cité, 75015 Paris, France;
| | - Jenny Bonifacio-Mundaca
- National Tumor Bank, Department of Pathology, National Institute of Neoplastic Diseases, Lima 15024, Peru;
| | - Claudia Monge
- Institut Pasteur, Université Paris Cité, Unité Organisation Nucléaire et Oncogenèse, INSERM U993, 75015 Paris, France; (C.M.); (P.P.)
| | - Pascal Pineau
- Institut Pasteur, Université Paris Cité, Unité Organisation Nucléaire et Oncogenèse, INSERM U993, 75015 Paris, France; (C.M.); (P.P.)
| | - Jorge Mata-Garrido
- Institut Pasteur, Université Paris Cité, Unité Organisation Nucléaire et Oncogenèse, INSERM U993, 75015 Paris, France; (C.M.); (P.P.)
| | - Raquel Francés
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 75006 Paris, France
| |
Collapse
|
142
|
Ellison EL, Zhou P, Chu YH, Hermanson P, Gomez-Cano L, Myers ZA, Abnave A, Gray J, Hirsch CN, Grotewold E, Springer NM. Transcriptome profiling of maize transcription factor mutants to probe gene regulatory network predictions. G3 (BETHESDA, MD.) 2025; 15:jkae274. [PMID: 39566186 PMCID: PMC11979765 DOI: 10.1093/g3journal/jkae274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Transcription factors play important roles in regulation of gene expression and phenotype. A variety of approaches have been utilized to develop gene regulatory networks to predict the regulatory targets for each transcription factor, such as yeast-1-hybrid screens and gene co-expression network analysis. Here we identified potential transcription factor targets and used a reverse genetics approach to test the predictions of several gene regulatory networks in maize. Loss-of-function mutant alleles were isolated for 22 maize transcription factors. These mutants did not exhibit obvious morphological phenotypes. However, transcriptomic profiling identified differentially expressed genes in each of the mutant genotypes, and targeted metabolic profiling indicated variable phenolic accumulation in some mutants. An analysis of expression levels for predicted target genes based on yeast-1-hybrid screens identified a small subset of predicted targets that exhibit altered expression levels. The analysis of predicted targets from gene co-expression network-based methods found significant enrichments for prediction sets of some transcription factors, but most predicted targets did not exhibit altered expression. This could result from false-positive gene co-expression network predictions, a transcription factor with a secondary regulatory role resulting in minor effects on gene regulation, or redundant gene regulation by other transcription factors. Collectively, these findings suggest that loss-of-function for single uncharacterized transcription factors might have limited phenotypic impacts but can reveal subsets of gene regulatory network predicted targets with altered expression.
Collapse
Affiliation(s)
- Erika L Ellison
- Department of Plant and Microbial Biology, University of Minnesota, Saint Paul, MN 55108, USA
| | - Peng Zhou
- Department of Plant and Microbial Biology, University of Minnesota, Saint Paul, MN 55108, USA
| | - Yi-Hsuan Chu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Peter Hermanson
- Department of Plant and Microbial Biology, University of Minnesota, Saint Paul, MN 55108, USA
| | - Lina Gomez-Cano
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Zachary A Myers
- Department of Plant and Microbial Biology, University of Minnesota, Saint Paul, MN 55108, USA
| | - Ankita Abnave
- Department of Biological Sciences, The University of Toledo, Toledo, OH 43606, USA
| | - John Gray
- Department of Biological Sciences, The University of Toledo, Toledo, OH 43606, USA
| | - Candice N Hirsch
- Department of Agronomy and Plant Genetics, University of Minnesota, Saint Paul, MN 55108, USA
| | - Erich Grotewold
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Nathan M Springer
- Department of Plant and Microbial Biology, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
143
|
Perkins ML, Crocker J, Tkačik G. Chromatin enables precise and scalable gene regulation with factors of limited specificity. Proc Natl Acad Sci U S A 2025; 122:e2411887121. [PMID: 39793086 PMCID: PMC11725945 DOI: 10.1073/pnas.2411887121] [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: 06/13/2024] [Accepted: 11/22/2024] [Indexed: 01/12/2025] Open
Abstract
Biophysical constraints limit the specificity with which transcription factors (TFs) can target regulatory DNA. While individual nontarget binding events may be low affinity, the sheer number of such interactions could present a challenge for gene regulation by degrading its precision or possibly leading to an erroneous induction state. Chromatin can prevent nontarget binding by rendering DNA physically inaccessible to TFs, at the cost of energy-consuming remodeling orchestrated by pioneer factors (PFs). Under what conditions and by how much can chromatin reduce regulatory errors on a global scale? We use a theoretical approach to compare two scenarios for gene regulation: one that relies on TF binding to free DNA alone and one that uses a combination of TFs and chromatin-regulating PFs to achieve desired gene expression patterns. We find, first, that chromatin effectively silences groups of genes that should be simultaneously OFF, thereby allowing more accurate graded control of expression for the remaining ON genes. Second, chromatin buffers the deleterious consequences of nontarget binding as the number of OFF genes grows, permitting a substantial expansion in regulatory complexity. Third, chromatin-based regulation productively co-opts nontarget TF binding for ON genes in order to establish a "leaky" baseline expression level, which targeted activator or repressor binding subsequently up- or down-modulates. Thus, on a global scale, using chromatin simultaneously alleviates pressure for high specificity of regulatory interactions and enables an increase in genome size with minimal impact on global expression error.
Collapse
Affiliation(s)
- Mindy Liu Perkins
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117Heidelberg, Germany
| | - Justin Crocker
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117Heidelberg, Germany
| | - Gašper Tkačik
- Institute of Science and Technology Austria, AT-3400Klosterneuburg, Austria
| |
Collapse
|
144
|
Lu X, Zhu M, Pei X, Ma J, Wang R, Wang Y, Chen S, Yan Y, Zhu Y. Super-enhancers in hepatocellular carcinoma: regulatory mechanism and therapeutic targets. Cancer Cell Int 2025; 25:7. [PMID: 39773719 PMCID: PMC11706108 DOI: 10.1186/s12935-024-03599-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Super-enhancers (SEs) represent a distinct category of cis-regulatory elements notable for their robust transcriptional activation capabilities. In tumor cells, SEs intricately regulate the expression of oncogenes and pivotal cancer-associated signaling pathways, offering significant potential for cancer treatment. However, few studies have systematically discussed the crucial role of SEs in hepatocellular carcinoma (HCC), which is one of the most common liver cancers with late-stage diagnosis and limited treatment methods for advanced disease. Herein, we first summarize the identification methods and the intricate processes of formation and organization of super-enhancers. Subsequently, we delve into the roles and molecular mechanisms of SEs within the framework of HCC. Finally, we discuss the inhibitors targeting the key SE-components and their potential effects on the treatment of HCC. In conclusion, this review meticulously encapsulates the distinctive characteristics of SEs and underscores their pivotal roles in the context of hepatocellular carcinoma, presenting a novel perspective on the potential of super-enhancers as emerging therapeutic targets for HCC.
Collapse
Affiliation(s)
- Xuejin Lu
- Department of Pathophysiology, College of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Meizi Zhu
- Department of Pathophysiology, College of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Xingyue Pei
- Department of Pathophysiology, College of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Jinhu Ma
- Department of Pathophysiology, College of Basic Medical Science, Anhui Medical University, Hefei, China
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Rui Wang
- Department of Pathophysiology, College of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Yi Wang
- Department of Pathophysiology, College of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Shuwen Chen
- Department of Pathophysiology, College of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Yan Yan
- Laboratory Animal Research Center, College of Basic Medical Science, Anhui Medical University, Hefei, China.
| | - Yaling Zhu
- Department of Pathophysiology, College of Basic Medical Science, Anhui Medical University, Hefei, China.
- Laboratory Animal Research Center, College of Basic Medical Science, Anhui Medical University, Hefei, China.
| |
Collapse
|
145
|
Kang J, Ahn K, Oh J, Lee T, Hwang S, Uh Y, Choi SJ. Identification of Endometriosis Pathophysiologic-Related Genes Based on Meta-Analysis and Bayesian Approach. Int J Mol Sci 2025; 26:424. [PMID: 39796277 PMCID: PMC11720405 DOI: 10.3390/ijms26010424] [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: 11/27/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Endometriosis is a complex disease with diverse etiologies, including hormonal, immunological, and environmental factors; however, its exact pathogenesis remains unknown. While surgical approaches are the diagnostic and therapeutic gold standard, identifying endometriosis-associated genes is a crucial first step. Five endometriosis-related gene expression studies were selected from the available datasets. Approximately, 14,167 genes common to these 5 datasets were analyzed for differential expression. Meta-analyses utilized fold-change values and standard errors obtained from each analysis, with the binomial and continuous datasets contributing to endometriosis presence and endometriosis severity meta-analysis, respectively. Approximately 160 genes showed significant results in both meta-analyses. For Bayesian analysis, endometriosis-related single nucleotide polymorphisms (SNPs), the human transcription factor catalog, uterine SNP-related gene expression, disease-gene databases, and interactome databases were utilized. Twenty-four genes, present in at least three or more databases, were identified. Network analysis based on Pearson's correlation coefficients revealed the HLA-DQB1 gene with both a high score in the Bayesian analysis and a central position in the network. Although ZNF24 had a lower score, it occupied a central position in the network, followed by other ZNF family members. Bayesian analysis identified genes with high confidence that could support discovering key diagnostic biomarkers and therapeutic targets for endometriosis.
Collapse
Affiliation(s)
- Jieun Kang
- Department of Obstetrics and Gynecology, Yonsei University Wonju College of Medicine, 20, Ilsan-ro, Wonju-si 26426, Republic of Korea;
| | - Kwangjin Ahn
- Department of Laboratory Medicine, Yonsei University Wonju College of Medicine, 20, Ilsan-ro, Wonju-si 26426, Republic of Korea;
| | - Jiyeon Oh
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, 20, Ilsan-ro, Wonju-si 26426, Republic of Korea;
| | - Taesic Lee
- Department of Family Medicine, Yonsei University Wonju College of Medicine, 20, Ilsan-ro, Wonju-si 26426, Republic of Korea;
| | - Sangwon Hwang
- Department of Precision Medicine, Yonsei University Wonju College of Medicine, 20, Ilsan-ro, Wonju-si 26426, Republic of Korea;
| | - Young Uh
- Department of Laboratory Medicine, Yonsei University Wonju College of Medicine, 20, Ilsan-ro, Wonju-si 26426, Republic of Korea;
| | - Seong Jin Choi
- Department of Obstetrics and Gynecology, Yonsei University Wonju College of Medicine, 20, Ilsan-ro, Wonju-si 26426, Republic of Korea;
| |
Collapse
|
146
|
Ren X, Shi Y, Xiao B, Su X, Shi H, He G, Chen P, Wu D, Shi Y. Gene Doping Detection From the Perspective of 3D Genome. Drug Test Anal 2025. [PMID: 39757126 DOI: 10.1002/dta.3850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025]
Abstract
Since the early 20th century, the concept of doping was first introduced. To achieve better athletic performance, chemical substances were used. By the mid-20th century, it became gradually recognized that the illegal use of doping substances can seriously endangered athletes' health and compromised the fairness of sports competitions. Over the past 30 years, the World Anti-Doping Agency (WADA) has established corresponding rules and regulations to prohibit athletes from using doping substances or restrict the use of certain drugs, and isotope, chromatography, and mass spectrometry techniques were accredited to detect doping substances. With the development of gene editing technology, many genetic diseases have been effectively treated, but enabled by the same technology, doping has also the potential to pose a threat to sports in the form of gene doping. WADA has explicitly indicated gene doping in the Prohibited List as a prohibited method (M3) and approved qPCR detection. However, gene doping can easily evade detection, if the target genes' upstream regulatory elements are considered, the task became more challenging. Hi-C experiment driven 3D genome technology, through perspectives such as topologically associating domain (TAD) and chromatin loop, provides a more comprehensive and in-depth understanding of gene regulation and expression, thereby better preventing the potential use of 3D genome level gene doping. In this work, we will explore gene doping from a different perspective by analyzing recent studies on gene doping and explore related genes under 3D genome.
Collapse
Affiliation(s)
- Xinyuan Ren
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Shi
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, China
| | - Bo Xiao
- Faculty of Physical Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xianbin Su
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Shi
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, China
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| | - Peijie Chen
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, China
| | - Die Wu
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, China
| | - Yi Shi
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
147
|
Levitsky VG, Raditsa VV, Tsukanov AV, Mukhin AM, Zhimulev IF, Merkulova TI. Asymmetry of Motif Conservation Within Their Homotypic Pairs Distinguishes DNA-Binding Domains of Target Transcription Factors in ChIP-Seq Data. Int J Mol Sci 2025; 26:386. [PMID: 39796242 PMCID: PMC11720554 DOI: 10.3390/ijms26010386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Transcription factors (TFs) are the main regulators of eukaryotic gene expression. The cooperative binding of at least two TFs to genomic DNA is a major mechanism of transcription regulation. Massive analysis of the co-occurrence of overrepresented pairs of motifs for different target TFs studied in ChIP-seq experiments can clarify the mechanisms of TF cooperation. We categorized the target TFs from M. musculus ChIP-seq and A. thaliana ChIP-seq/DAP-seq experiments according to the structure of their DNA-binding domains (DBDs) into classes. We studied homotypic pairs of motifs, using the same recognition model for each motif. Asymmetric and symmetric pairs consist of motifs of remote and close recognition scores. We found that asymmetric pairs of motifs predominate for all TF classes. TFs from the murine/plant 'Basic helix-loop-helix (bHLH)', 'Basic leucine zipper (bZIP)', and 'Tryptophan cluster' classes and murine 'p53 domain' and 'Rel homology region' classes showed the highest enrichment of asymmetric homotypic pairs of motifs. Pioneer TFs, despite their DBD types, have a higher significance of asymmetry within homotypic pairs of motifs compared to other TFs. Asymmetry within homotypic CEs is a promising new feature decrypting the mechanisms of gene transcription regulation.
Collapse
Affiliation(s)
- Victor G. Levitsky
- Department of System Biology, Institute of Cytology and Genetics, Novosibirsk 630090, Russia; (V.V.R.); (A.V.T.); (A.M.M.); (T.I.M.)
- Department of Natural Science, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Vladimir V. Raditsa
- Department of System Biology, Institute of Cytology and Genetics, Novosibirsk 630090, Russia; (V.V.R.); (A.V.T.); (A.M.M.); (T.I.M.)
| | - Anton V. Tsukanov
- Department of System Biology, Institute of Cytology and Genetics, Novosibirsk 630090, Russia; (V.V.R.); (A.V.T.); (A.M.M.); (T.I.M.)
- Institute of Molecular and Cellular Biology, Novosibirsk 630090, Russia;
| | - Aleksey M. Mukhin
- Department of System Biology, Institute of Cytology and Genetics, Novosibirsk 630090, Russia; (V.V.R.); (A.V.T.); (A.M.M.); (T.I.M.)
| | - Igor F. Zhimulev
- Institute of Molecular and Cellular Biology, Novosibirsk 630090, Russia;
| | - Tatyana I. Merkulova
- Department of System Biology, Institute of Cytology and Genetics, Novosibirsk 630090, Russia; (V.V.R.); (A.V.T.); (A.M.M.); (T.I.M.)
- Department of Natural Science, Novosibirsk State University, Novosibirsk 630090, Russia
| |
Collapse
|
148
|
Chi WY, Yoon SH, Mekerishvili L, Ganesan S, Potenski C, Izzo F, Landau D, Raimondi I. Single-cell mapping of regulatory DNA:Protein interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630903. [PMID: 39803441 PMCID: PMC11722406 DOI: 10.1101/2024.12.31.630903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Gene expression is coordinated by a multitude of transcription factors (TFs), whose binding to the genome is directed through multiple interconnected epigenetic signals, including chromatin accessibility and histone modifications. These complex networks have been shown to be disrupted during aging, disease, and cancer. However, profiling these networks across diverse cell types and states has been limited due to the technical constraints of existing methods for mapping DNA:Protein interactions in single cells. As a result, a critical gap remains in understanding where TFs or other chromatin remodelers bind to DNA and how these interactions are perturbed in pathological contexts. To address this challenge, we developed a transformative single-cell immuno-tethering DNA:Protein mapping technology. By coupling a species-specific antibody-binding nanobody to a cytosine base editing enzyme, this approach enables profiling of even weak or transient factor binding to DNA, a task that was previously unachievable in single cells. Thus, our Docking & Deamination followed by sequencing (D&D-seq) technique induces cytosine-to-uracil edits in genomic regions bound by the target protein, offering a novel means to capture DNA:Protein interactions with unprecedented resolution. Importantly, this technique can be seamlessly incorporated into common single-cell multiomics workflows, enabling multimodal analysis of gene regulation in single cells. We tested the ability of D&D-seq to record TF binding both in bulk and at the single-cell level by profiling CTCF and GATA family members, obtaining high specificity and efficiency, with clear identification of TF footprint and signal retention in the targeted cell subpopulations. Furthermore, the deamination reaction showed minimal off-target activity, with high concordance to bulk ChIP-seq reference data. Applied to primary human peripheral blood mononuclear cells (PBMCs), D&D-seq successfully identified CTCF binding sites and enabled integration with advanced machine-learning algorithms for predicting 3D chromatin structure. Furthermore, we integrated D&D-seq with single-cell genotyping to assess the impact of IDH2 mutations on CTCF binding in a human clonal hematopoiesis sample, uncovering altered binding and chromatin co-accessibility patterns in mutant cells. Altogether, D&D-seq represents an important technological advance enabling the direct mapping of TF or chromatin remodeler binding to the DNA in primary human samples, opening new avenues for understanding chromatin and transcriptional regulation in health and disease.
Collapse
Affiliation(s)
- Wei-Yu Chi
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Sang-Ho Yoon
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Levan Mekerishvili
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Saravanan Ganesan
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Catherine Potenski
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Franco Izzo
- Icahn School of Medicine at Mount Sinai, Department of Oncological Sciences, New York, NY, USA
| | - Dan Landau
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Ivan Raimondi
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
149
|
Moorman A, Benitez EK, Cambulli F, Jiang Q, Mahmoud A, Lumish M, Hartner S, Balkaran S, Bermeo J, Asawa S, Firat C, Saxena A, Wu F, Luthra A, Burdziak C, Xie Y, Sgambati V, Luckett K, Li Y, Yi Z, Masilionis I, Soares K, Pappou E, Yaeger R, Kingham TP, Jarnagin W, Paty PB, Weiser MR, Mazutis L, D'Angelica M, Shia J, Garcia-Aguilar J, Nawy T, Hollmann TJ, Chaligné R, Sanchez-Vega F, Sharma R, Pe'er D, Ganesh K. Progressive plasticity during colorectal cancer metastasis. Nature 2025; 637:947-954. [PMID: 39478232 PMCID: PMC11754107 DOI: 10.1038/s41586-024-08150-0] [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: 07/10/2023] [Accepted: 10/02/2024] [Indexed: 11/06/2024]
Abstract
As cancers progress, they become increasingly aggressive-metastatic tumours are less responsive to first-line therapies than primary tumours, they acquire resistance to successive therapies and eventually cause death1,2. Mutations are largely conserved between primary and metastatic tumours from the same patients, suggesting that non-genetic phenotypic plasticity has a major role in cancer progression and therapy resistance3-5. However, we lack an understanding of metastatic cell states and the mechanisms by which they transition. Here, in a cohort of biospecimen trios from same-patient normal colon, primary and metastatic colorectal cancer, we show that, although primary tumours largely adopt LGR5+ intestinal stem-like states, metastases display progressive plasticity. Cancer cells lose intestinal cell identities and reprogram into a highly conserved fetal progenitor state before undergoing non-canonical differentiation into divergent squamous and neuroendocrine-like states, a process that is exacerbated in metastasis and by chemotherapy and is associated with poor patient survival. Using matched patient-derived organoids, we demonstrate that metastatic cells exhibit greater cell-autonomous multilineage differentiation potential in response to microenvironment cues compared with their intestinal lineage-restricted primary tumour counterparts. We identify PROX1 as a repressor of non-intestinal lineage in the fetal progenitor state, and show that downregulation of PROX1 licenses non-canonical reprogramming.
Collapse
Affiliation(s)
- Andrew Moorman
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth K Benitez
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Francesco Cambulli
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Qingwen Jiang
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ahmed Mahmoud
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Program, Weill Cornell Graduate School, New York, NY, USA
| | - Melissa Lumish
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Saskia Hartner
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sasha Balkaran
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan Bermeo
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simran Asawa
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Canan Firat
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Asha Saxena
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fan Wu
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anisha Luthra
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cassandra Burdziak
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yubin Xie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Tri-Institutional PhD Program in Computational Biology and Medicine, New York, NY, USA
| | - Valeria Sgambati
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kathleen Luckett
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Yanyun Li
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Zhifan Yi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ignas Masilionis
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin Soares
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emmanouil Pappou
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T Peter Kingham
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William Jarnagin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Philip B Paty
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin R Weiser
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Linas Mazutis
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael D'Angelica
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jinru Shia
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julio Garcia-Aguilar
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tal Nawy
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Travis J Hollmann
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Ronan Chaligné
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Roshan Sharma
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Karuna Ganesh
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
150
|
Shishkin SS. Moonlighting Proteins of Human and Some Other Eukaryotes. Evolutionary Aspects. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S36-S59. [PMID: 40164152 DOI: 10.1134/s0006297924602855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/02/2025]
Abstract
This review presents materials on formation of the concept of moonlighting proteins and general characteristics of different similar proteins. It is noted that the concept under consideration is based on the data on the existence in different organisms of individual genes, protein products of which have not one, but at least two fundamentally different functions, for example, depending on cellular or extracellular location. An important feature of these proteins is that their functions can be switched. As a result, in different cellular compartments or outside the cells, as well as under a number of other circumstances, one of the possible functions can be carried out, and under other conditions, another. It is emphasized that the significant interest in moonlighting proteins is due to the fact that information is currently accumulating about their involvement in many vital molecular processes (glycolysis, translation, transcription, replication, etc.). Alternative hypotheses on the evolutionary origin of moonlighting proteins are discussed.
Collapse
Affiliation(s)
- Sergei S Shishkin
- Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Moscow, 119071, Russia.
| |
Collapse
|