1
|
Ji L, Chen Y, Chen X. Circular RNA Circ_0002762 promotes cell migration and invasion in cervical squamous cell carcinoma via activating RelA/nuclear factor kappa B (Nf-kB) signalling pathway. RNA Biol 2025; 22:1-13. [PMID: 40083243 PMCID: PMC11934174 DOI: 10.1080/15476286.2025.2478539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 02/18/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Cervical cancer is a leading cause of cancer-related deaths, with cervical squamous cell carcinoma (CSCC) accounting for a majority of cases. Circular RNAs (circRNAs) have been repeatedly suggested as crucial effectors in modulating the development of multiple malignancies. The expression of circ_0002762 was predicted to be high in CSCC tissues in GEO dataset, but the functional role and underlying regulatory mechanism of circ_0002762 in CSCC was unclear. By series of functional assays and mechanism assays, supported by bioinformatics analysis, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) analysis and western blot assays, we identified that circ_0002762 aberrantly up-regulated in CSCC, promoting CSCC cell migration and invasion. Mechanically, circ_0002762 was transcriptionally activated by Fork head box A1 (FOXA1). Moreover, the involvement of nuclear factor kappa B (NF-kB) signalling in circ_0002762 regulation mechanism in CSCC cells was ascertained. Additionally, circ_0002762, predominantly accumulated in cell cytoplasm, was proved to recruit Mov10 RISC complex RNA helicase (MOV10) to enhance RelA mRNA stability, thus affecting CSCC cell migration and invasion. In summary, FOXA1-mediated circ_0002762 up-regulation could enhance the migratory and invasive abilities of CSCC cells via the MOV10/RelA/NF-kB pathway.
Collapse
Affiliation(s)
- Lei Ji
- Department of Obstetrics and Gynecology, Yancheng First People’s Hospital of Jiangsu Province, Yancheng, Jiangsu, China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaoping Chen
- Department of Obstetrics and Gynecology, Yancheng First People’s Hospital of Jiangsu Province, Yancheng, Jiangsu, China
| |
Collapse
|
2
|
Mahjoubin-Tehran M, Rezaei S, Kesharwani P, Karav S, Sahebkar A. Decoy oligodeoxynucleotides targeting STATs in non-cancer gene therapy. Gene 2025; 957:149482. [PMID: 40216342 DOI: 10.1016/j.gene.2025.149482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
The Signal Transducer and Activator of Transcription (STAT) protein family is crucial for organizing the epigenetic configuration of immune cells and controlling various fundamental cell physiological functions including apoptosis, development, inflammation, immunological responses, and cell proliferation and differentiation. The human genome has seven known STAT genes, named 1, 2, 3, 4, 5a, 5b, and 6. Aberrant activation of STAT signaling pathways is associated with many human disorders, particularly cardiovascular diseases (CVDs), making these proteins promising therapeutic targets. Improved understanding of altered and pathological gene expression and its role in the pathophysiology of various hereditary and acquired disorders has enabled the development of novel treatment approaches based on gene expression modulation. One such promising development is the oligodeoxynucleotide decoy method, which may allow researchers to specifically influence gene activation or repression. Various oligodeoxynucleotide decoys target STATs and affect the expression of its downstream genes. We summarized cell culture and preclinical research, which evaluated the effects of oligodeoxynucleotide decoys target STATs in different types of non-cancer illnesses.
Collapse
Affiliation(s)
- Maryam Mahjoubin-Tehran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Rezaei
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Fang Y, Niu X, Zhao W, Zhang H. Cardioprotective potential of transcription factor PRRX1 silencing against myocardial ischemia/reperfusion injury by regulating excessive mitophagy and ferroptosis through FKBP5-p38 MAPK axis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167766. [PMID: 40044064 DOI: 10.1016/j.bbadis.2025.167766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/21/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a major cause of various adverse cardiovascular outcomes associated with excessive mitophagy and cardiomyocyte ferroptosis. Paired-related homeobox 1 (PRRX1) is a transcriptional factor involved in cardiovascular injury. However, whether and how PRRX1 regulates excessive mitophagy and cardiomyocyte ferroptosis during myocardial I/R injury remains unclear. Oxygen-glucose deprivation and reperfusion (OGD/R)-treated AC16 cardiomyocytes and myocardial I/R-induced rats were used as in vitro and in vivo models. Our results showed that PRRX1 expression was upregulated in AC16 cells after OGD/R treatment. PRRX1 silencing mitigated OGD/R-induced excessive mitophagy by increasing the mitochondrial membrane potential, adenosine triphosphate and p62 levels, and reducing LC3 II/I level in AC16 cells. In addition, PRRX1 knockdown attenuated OGD/R-induced lactate dehydrogenase (LDH) release and cardiomyocyte ferroptosis by decreasing reactive oxygen species, Fe2+ and acyl-CoA synthetase long-chain family member 4 (ACSL4) levels, and increasing glutathione (GSH) and glutathione peroxidase 4 (GPX4) levels. Furthermore, PRRX1 transcriptionally promoted FK506 binding protein 5 (FKBP5), and increased p38 MAPK activation in AC16 cells. FKBP5 overexpression reversed the effects of PRRX1 silencing on excessive mitophagy and cardiomyocyte ferroptosis in OGD/R-treated AC16 cells. These effects were mitigated by a p38 MAPK inhibitor. Finally, PRRX1 downregulation mitigated myocardial I/R injury by reducing heart infarction and creatine kinase-myocardial band (CK-MB) levels in rat models. These findings demonstrate that PRRX1 silencing attenuates OGD/R-induced excessive mitophagy and cardiomyocyte ferroptosis by decreasing FKBP5 expression and inactivating p38 MAPK signaling, indicating the cardioprotective potential of PRRX1 silencing in myocardial I/R injury.
Collapse
Affiliation(s)
- Yongpeng Fang
- Department of Geriatrics, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - Xudong Niu
- Department of Internal Medicine, Yinchuan Maternal and Child Health Hospital, Yinchuan 750004, China
| | - Weifang Zhao
- Cardiac Function Examination Room, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266041, China
| | - Huali Zhang
- Medical Examination Center, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou 730050, China.
| |
Collapse
|
4
|
Liu C, Wang X, Xu S, Liu M, Cao X. Regulation of autophagy: Insights into O-GlcNAc modification mechanisms. Life Sci 2025; 369:123547. [PMID: 40058573 DOI: 10.1016/j.lfs.2025.123547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/30/2025]
Abstract
Autophagy is a "self-eating" biological process that degrades cytoplasmic contents to ensure cellular homeostasis. Its response to stimuli occurs in two stages: Within a few to several hours of exposure to a stress condition, autophagic flow rapidly increases, which is mediated by post-translational modification (PTM). Subsequently, the transcriptional program is activated and mediates the persistent autophagic response. O-linked β-N-acetylglucosamine (O-GlcNAc) modification is an inducible and dynamically cycling PTM; mounting evidence suggests that O-GlcNAc modification participates in the total autophagic process, including autophagy initiation, autophagosome formation, autophagosome-lysosome fusion, and transcriptional process. In this review, we summarize the current knowledge on the emerging role of O-GlcNAc modification in regulating autophagy-associated proteins and explain the different regulatory effects on autophagy exerted by O-GlcNAc modification.
Collapse
Affiliation(s)
- Chengzhi Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinyu Wang
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Shengnan Xu
- College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Mingyue Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xusheng Cao
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China.
| |
Collapse
|
5
|
Wang X, Xie C, Shen K, Li D, Xie XS. Quantification and potential functional relevance of binding cooperativity of adjacent transcription factors on DNA. Proc Natl Acad Sci U S A 2025; 122:e2422555122. [PMID: 40305050 DOI: 10.1073/pnas.2422555122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/23/2025] [Indexed: 05/02/2025] Open
Abstract
In eukaryotes, the expression of specific genes is regulated by a combination of transcription factors (TFs) bound on regulatory regions of the genomic DNA (promoters and enhancers). Recent advances in genomic sequencing technology have enabled the measurements of TFs' footprints and binding affinities on DNA at the single-molecule level, facilitating the probing of binding cooperativity among adjacent TFs. This necessitates quantitative descriptions of TFs' binding cooperativity and understanding of its potential functional relevance. In this study, we show that the binding cooperativities between two adjacent TFs can be quantified by the [Formula: see text] coefficient, which can be experimentally determined. Under thermodynamic equilibrium, the binding affinities of two TFs can either increase together (positive cooperativity) or decrease together (negative cooperativity), but not in opposing directions (one increases while the other decreases). Within the framework of thermodynamics, we investigate the functional relevance of cooperativity. The functional relevance of positive cooperativity, which has been extensively discussed in the literature, is the sigmoidal binding curve around a TF concentration threshold (analogous to oxygen binding to hemoglobin), whereas the functional relevance of negative cooperativity is twofold. First, mutual exclusion of the two TFs enables bidirectional gene switching, akin to the CI-Cro system in phage [Formula: see text]. Second, while TFs often exhibit intranuclear concentration fluctuations, negative binding cooperativity assures fast TF dissociation from DNA and hence rapid response for gene expression regulation. Furthermore, the nonequilibrium steady states of living cells can lead to either positive or negative cooperativity, which can also be quantified by the [Formula: see text] coefficient.
Collapse
Affiliation(s)
- Xinyao Wang
- Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, People's Republic of China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, People's Republic of China
| | - Chen Xie
- Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, People's Republic of China
- Changping Laboratory, Beijing 102206, People's Republic of China
| | - Ke Shen
- Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, People's Republic of China
- Changping Laboratory, Beijing 102206, People's Republic of China
- School of Life Sciences, Peking University, Beijing 100871, People's Republic of China
| | - Dubai Li
- Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, People's Republic of China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, People's Republic of China
- Changping Laboratory, Beijing 102206, People's Republic of China
| | - Xiaoliang Sunney Xie
- Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, People's Republic of China
- Changping Laboratory, Beijing 102206, People's Republic of China
| |
Collapse
|
6
|
Abe K. Dynamic activity changes in transcription factors: Unlocking the mechanisms regulating physiological changes in the brain. Neurosci Res 2025; 214:16-22. [PMID: 39134224 DOI: 10.1016/j.neures.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 07/30/2024] [Accepted: 08/04/2024] [Indexed: 08/18/2024]
Abstract
Transcription factors (TFs) regulate the establishment and modulation of the transcriptome within cells, thereby playing a crucial role in various aspects of cellular physiology throughout the body. Quantitative measurement of TF activity during the development, function, and dysfunction of the brain is essential for gaining a deeper understanding of the regulatory mechanisms governing gene expression during these processes. Due to their role as regulators of gene expression, assessing and modulating detailed TF activity contributes to the development of practical methods to intervene in these processes, potentially offering more efficient treatments for diseases. Recent methodologies have revealed that TF activity is dynamically regulated within cells and organisms, including the adult brain. This review summarizes the regulatory mechanisms of TF activities and the methodologies used to assess them, emphasizing their importance in both fundamental research and clinical applications.
Collapse
Affiliation(s)
- Kentaro Abe
- Lab of Brain Development, Graduate School of Life Sciences, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai, Miyagi 980-8577, Japan; Division for the Establishment of Frontier Sciences of the Organization for Advanced Studies, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai, Miyagi 980-8577, Japan.
| |
Collapse
|
7
|
Hu D, Zhang Z, Wang Y, Li S, Zhang J, Wu Z, Sun M, Jiang J, Liu D, Ji X, Wang S, Wang Y, Luo X, Huang W, Xia L. Transcription factor ELF4 in physiology and diseases: Molecular roles and clinical implications. Genes Dis 2025; 12:101394. [PMID: 40083328 PMCID: PMC11904542 DOI: 10.1016/j.gendis.2024.101394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/21/2024] [Accepted: 07/28/2024] [Indexed: 03/16/2025] Open
Abstract
Transcription factor E74 like ETS transcription factor 4 (ELF4), a member of the ETS family, is highly expressed in normal human hematopoietic tissue, ovary, placenta, colon, and certain pathological cell lines. During normal physiological processes, ELF4 regulates differentiation in osteogenic, adipocyte, and neuronal types. It also exerts a critical impact on the development of the immune system. However, its function is dysregulated through posttranslational modifications, gene fusions, and complex signaling crosstalk under pathological conditions. Furthermore, serving as a double-edged sword in cancer, ELF4 exhibits both tumor-suppressing and tumor-promoting effects. Specifically, ELF4 plays a critical role in cancer metastasis, proliferation, and modulation of the tumor microenvironment. This review provides an in-depth overview of the molecular structure and post-translational modifications of ELF4. It also summarizes the hallmarks of ELF4 in physiology and diseases, with a particular focus on its significance in oncology. Notably, this review underscores the potential of ELF4 as a prognostic biomarker, highlighting its clinical relevance. Finally, it discusses unresolved questions and future research directions of ELF4. An in-depth understanding of ELF4 biology could facilitate its clinical translation and offer promising targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dian Hu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zerui Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yijun Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhangfan Wu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Junqing Jiang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Danfei Liu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaoyu Ji
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiangyuan Luo
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shannxi 710032, China
| |
Collapse
|
8
|
Xu B, Zhou L, Zhang Q. Curcumin Inhibits the Progression of Non-small Cell Lung Cancer by Regulating DMRT3/SLC7A11 Axis. Mol Biotechnol 2025; 67:1880-1892. [PMID: 38744789 DOI: 10.1007/s12033-024-01166-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/03/2024] [Indexed: 05/16/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a fatal malignancy all over the world. Emerging studies have shown that curcumin might repress NSCLC progression by regulating ferroptosis, but the underlying mechanism remains unclear. 16HBE, LK-2, and H1650 cell viability was detected using Cell Counting Kit-8 assay. LK-2 and H1650 cell proliferation, apoptosis, and angiopoiesis were measured using 5-ethynyl-2'-deoxyuridine, flow cytometry, and tube formation assay. Superoxide dismutase, Malondialdehyde, Glutathione, and lactate dehydrogenase levels in LK-2 and H1650 cells were examined using special assay kits. Fe+ level was assessed using an iron assay kit. Doublesex and Mab-3 related Transcription Factor 3 (DMRT3) and solute carrier family 7 member 11 (SLC7A11) protein levels were detected using western in NSCLC tissues, adjacent matched normal tissues, 16HBE cells, LK-2 cells, H1650 cells, and xenograft tumor tissues. Glutathione peroxidase 4, Acyl-CoA Synthetase Long Chain Family Member 4, and transferrin receptor 1 protein levels in LK-2 and H1650 cells were examined by western blot assay. DMRT3 and SLC7A11 levels were determined using real-time quantitative polymerase chain reaction. After JASPAR prediction, binding between DMRT3 and SLC7A11 promoter was verified using Chromatin immunoprecipitation and dual-luciferase reporter assays in LK-2 and H1650 cells. Role of curcumin on NSCLC tumor growth was assessed using the xenograft tumor model in vivo. Curcumin blocked NSCLC cell proliferation and angiopoiesis, and induced apoptosis and ferroptosis. DMRT3 or SLC7A11 upregulation partly abolished the suppressive role of curcumin on NSCLC development. In mechanism, DMRT3 was a transcription factor of SLC7A11 and increased the transcription of SLC7A11 via binding to its promoter region. Curcumin inhibited NSCLC growth in vivo by modulating DMRT3. Curcumin might constrain NSCLC cell malignant phenotypes partly through the DMRT3/SLC7A11 axis, providing a promising therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Bin Xu
- Department of TCM, Changzhou Cancer Hospital, No.68, Honghe Road, Xinbei District, Changzhou City, 213000, Jiangsu, China
| | - Li Zhou
- Department of TCM, Changzhou Cancer Hospital, No.68, Honghe Road, Xinbei District, Changzhou City, 213000, Jiangsu, China
| | - Qian Zhang
- Department of TCM, Changzhou Cancer Hospital, No.68, Honghe Road, Xinbei District, Changzhou City, 213000, Jiangsu, China.
| |
Collapse
|
9
|
Fu Z, Feng B, Akogo HY, Ma J, Liu Y, Quan H, Zhang X, Hou Y, Zhang X, Ma J, Cui H. Amyotrophic Lateral Sclerosis and Parkinson's Disease: Brain Tissue Transcriptome Analysis Reveals Interactions. Mol Neurobiol 2025; 62:6383-6396. [PMID: 39792201 DOI: 10.1007/s12035-024-04681-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
This study utilises amyotrophic lateral sclerosis (ALS) and Parkinson's disease (PD) human brain samples from the GEO database and employs differential expression gene (DEG) analysis to identify genes that are pivotal in both neurodegenerative diseases. Through in depth GO and KEGG enrichment analyses, we elucidated the biological functions and potential pathways associated with these DEGs. Furthermore, by constructing protein‒protein interaction networks, we highlight the significance of shared DEGs in both cellular physiology and disease contexts. Analysis of drug‒gene associations revealed potential therapeutic compounds linked to ALS and PD treatment. Additionally, we explored the interactions between transcription factors, miRNAs, and common DEGs, revealing aspects of gene regulatory networks. This study provides insights into the molecular mechanisms of ALS and PD, offering valuable contributions to ongoing research and potential therapeutic avenues.
Collapse
Affiliation(s)
- Zewei Fu
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Baofeng Feng
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, PMB UCC, Cape Coast, Ghana
| | - Herman Yao Akogo
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Human Anatomy Department, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
| | - Jiajia Ma
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Yukun Liu
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Hezhi Quan
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Xiaohan Zhang
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Yu Hou
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Xuecong Zhang
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Jun Ma
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China.
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China.
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China.
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China.
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, PMB UCC, Cape Coast, Ghana.
| | - Huixian Cui
- Hebei Medical University-Galway University Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, PMB UCC, Cape Coast, Ghana
| |
Collapse
|
10
|
Xu S, Yin K, Xu X, Fu L, Wu R. O-GlcNAcylation reduces proteome solubility and regulates the formation of biomolecular condensates in human cells. Nat Commun 2025; 16:4068. [PMID: 40307207 PMCID: PMC12043995 DOI: 10.1038/s41467-025-59371-4] [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: 11/20/2023] [Accepted: 04/22/2025] [Indexed: 05/02/2025] Open
Abstract
O-GlcNAcylation plays critical roles in the regulation of protein functions and cellular activities, including protein interactions with other macromolecules. While the formation of biomolecular condensates (or biocondensates) regulated by O-GlcNAcylation in a few individual proteins has been reported, systematic investigation of O-GlcNAcylation on the regulation of biocondensate formation remains to be explored. Here we systematically study the roles of O-GlcNAcylation in regulating protein solubility and its impacts on RNA-protein condensates using mass spectrometry-based chemoproteomics. Unexpectedly, we observe a system-wide decrease in the solubility of proteins modified by O-GlcNAcylation, with glycoproteins involved in focal adhesion and actin binding exhibiting the most significant decrease. Furthermore, O-GlcNAcylation sites located in disordered regions and with fewer acidic and aromatic residues nearby are related to a greater drop in protein solubility. Additionally, we discover that a specific group of O-GlcNAcylation events promotes the dissociation of RNA-protein condensates under heat stress, while some enhance the formation of RNA-protein condensates during the recovery phase. Using site mutagenesis, inhibition of O-GlcNAc transferase, and fluorescence microscopy, we validate that O-GlcNAcylation regulates the formation of biocondensates for YTHDF3 and NUFIP2. This work advances our understanding of the functions of protein O-GlcNAcylation and its roles in the formation of biomolecular condensates.
Collapse
Affiliation(s)
- Senhan Xu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- The Scripps Research Institute, La Jolla, CA, USA
| | - Kejun Yin
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Incyte Corporation, Wilmington, DE, USA
| | - Xing Xu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Longping Fu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Ronghu Wu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
11
|
Yang S, Li Y, Ruan R, Yu J, Zhu B, Lou H, Zhang X, Wang S. Exogenous TSG-6 treatment alleviates DSS-induced colitis in mice by modulating Pou2f3 and promoting tuft cells differentiation. Mol Med 2025; 31:157. [PMID: 40301757 PMCID: PMC12042439 DOI: 10.1186/s10020-025-01230-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/24/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Whereas intestinal epithelial barrier dysfunction is implicated in inflammatory bowel disease (IBD), the underlying mechanisms remain elusive. Tumor necrosis factor α stimulated gene 6 (TSG-6) is a secretory protein with anti-inflammatory properties. Our previous research demonstrated TSG-6 can relieve intestinal inflammation and mucosal damage. However, the underlying mechanism and targets remain unclear. This research sought to explore how TSG-6 regulates the intestinal epithelial barrier and its mechanistic role in experimental colitis. METHODS IBD mouse model was generated using dextran sodium sulfate (DSS), with or without intraperitoneal injection of TSG-6(100 µg/kg or 200 µg/kg). The effects of TSG-6 on colonic inflammation and intestinal barrier function were investigated. Label-free quantitative proteomic analysis was performed on intestinal samples to explore the mechanism and therapeutic target of TSG-6. Molecular interactions were determined by co-immunoprecipitation (Co-IP) and immunofluorescence colocalization. RESULTS TSG-6 treatment significantly attenuated DSS-induced colitis symptoms and inflammatory cell infiltration. Microarray analysis revealed that TSG-6 decreased pro-inflammatory cytokine levels in colon tissue. TSG-6 restored the intestinal epithelial barrier through the promotion of intestinal epithelial cells (IECs) proliferation and mitigation of tight junctions (TJs) damage. Mechanistically, TSG-6 promoted tuft cells differentiation and increased interleukin-25 (IL-25) levels by directly binding to Pou class 2 homeobox 3(Pou2f3) and up-regulating its expression in the gut. CONCLUSIONS This study demonstrated TSG-6 as a positive regulator of tuft cells differentiation by interacting with Pou2f3, and the effectiveness of exogenous TSG-6 treatment on maintaining intestinal barrier integrity showed a promising potential for its clinical application.
Collapse
Affiliation(s)
- Shaopeng Yang
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yuqi Li
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Rongwei Ruan
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Jiangping Yu
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Bo Zhu
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Haibin Lou
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xiaolan Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Shi Wang
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
12
|
Chen W, Zeng S, Zhong J, Zou J, Lei Y, Chen X, Mei Q, Luo Q. Mapping immune cell dynamics and macrophage plasticity in breast cancer tumor microenvironment through single-cell analysis. Discov Oncol 2025; 16:625. [PMID: 40293603 PMCID: PMC12037460 DOI: 10.1007/s12672-025-02419-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Breast cancer (BRCA) is a complex disease influenced by the tumor microenvironment, where interactions between immune cells and cancer cells play a crucial role in tumor progression and response to therapy. Understanding the intricacies of these interactions requires detailed analysis at the single-cell level, enabling the identification of specific immune cell subpopulations and their functional roles within the tumor milieu. This study comprehensively analyzed immune cell subpopulations and macrophage subtypes in BRCA using single-cell RNA sequencing technology and various computational tools. Initially, Sc-Type software accurately identified and annotated immune cell subpopulations, followed by CNV analysis using infercnv software, revealing significant CNV variations in epithelial cells. Subsequently, macrophages were re-clustered into 5 clusters, and their biological significance and functional features were assessed. CellChat analysis elucidated potential interactions between macrophage subtypes and BRCA cells, primarily through SPP1-CD44 and LGALS9-CD44 signaling networks. Additionally, CytoTRACE and Monocle were employed to analyze cellular plasticity and differentiation trajectories of macrophage subtypes. Furthermore, efferocytosis-related gene set scoring, transcription factor analysis, and risk score development were conducted, followed by immune infiltration and tumor mutation burden analysis, revealing increased immune infiltration and higher TMB levels in the high-risk group. These findings offer crucial insights into the interaction mechanisms of immune cells and macrophage subtypes within the BRCA tumor microenvironment, aiding in the understanding of tumor progression and therapeutic interventions.
Collapse
Affiliation(s)
- Wang Chen
- Department of Pharmacy, The Affiliated Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, People's Republic of China
| | - Siyu Zeng
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, People's Republic of China
| | - Junyong Zhong
- Department of Oncology, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, People's Republic of China
| | - Jian Zou
- Department of Pharmacy, The Affiliated Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, People's Republic of China
- School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yanli Lei
- Department of Pharmacy, The 2, People's Hospital of Bijie, Bijie, , Guizhou, China
| | - Xiaohan Chen
- Department of Pharmacy, The Affiliated Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, People's Republic of China
| | - Qinghua Mei
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, People's Republic of China.
| | - Qianhua Luo
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, People's Republic of China.
| |
Collapse
|
13
|
Kim C, Smith SN, Gopalan SS, Kerwin SR, Ballard K, Perry BW, Smith CF, Saviola AJ, Adams RH, Mackessy SP, Castoe TA. Unique physiological and regulatory activity drives divergent toxin and non-toxin gene expression in rattlesnake accessory venom glands. Toxicon 2025:108376. [PMID: 40300655 DOI: 10.1016/j.toxicon.2025.108376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/01/2025]
Abstract
Understanding the mechanisms by which organs and tissues evolve new physiological functions is central to understanding the evolution of novelty. This is particularly interesting in the context of related tissues that evolve specialized, yet complementary, functions. Snake venom glands are an attractive system to test hypotheses related to the evolution and specialization of novel physiological function, as these modified salivary glands have evolved over ∼60 MY to synthesize and store venom. Front-fanged venomous snakes (elapids and viperids) possess two types of venom glands: the main and accessory glands. The larger main gland produces greater quantities of venom toxins and has been studied extensively, while the smaller accessory gland has received less attention. Here, we explore gene expression differences between main and accessory venom glands across three rattlesnake species (Crotalus cerberus, C. oreganus concolor and C. viridis). Our findings indicate that accessory glands express most venom genes at significantly lower levels than the main gland, with a few exceptions that may represent biologically relevant contributions of accessory glands to venom. The two glands also exhibit distinct trans-regulatory environments that we link to key differences in their underlying physiology and secretory roles. Our results further suggest that two signaling pathways that regulate venom, the unfolded protein response (UPR) and extracellular signal-regulated kinase (ERK), show significantly lower activation in the accessory gland. These findings provide insight into the physiological and functional diversification of snake venom systems, highlighting how distinct glandular systems have evolved contrasting and complementary roles driven by distinct physiological and regulatory mechanisms.
Collapse
Affiliation(s)
- Claire Kim
- Department of Biology, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Sierra N Smith
- Department of Biology, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Siddharth S Gopalan
- Department of Biology, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Samuel R Kerwin
- Department of Biological Sciences, University of Northern Colorado, Greely, CO, 80639, USA
| | - Kaas Ballard
- Department of Biology, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Blair W Perry
- Department of Ecology and Evolutionary Biology, University of California Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Cara F Smith
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Richard H Adams
- Department of Entomology and Plant Pathology, University of Arkansas Agricultural Experimental Station, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Stephen P Mackessy
- Department of Biological Sciences, University of Northern Colorado, Greely, CO, 80639, USA
| | - Todd A Castoe
- Department of Biology, University of Texas at Arlington, Arlington, TX, 76019, USA.
| |
Collapse
|
14
|
Jin H, Zhang D, Ma Y, Meng L, Huang S, Su H, Xu J, Yao Y. YTHDC2 manipulates anti-tumoral macrophage polarization and predicts favorable outcomes in triple negative breast cancer. NPJ Precis Oncol 2025; 9:119. [PMID: 40274959 PMCID: PMC12022267 DOI: 10.1038/s41698-025-00880-2] [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: 05/14/2024] [Accepted: 03/11/2025] [Indexed: 04/26/2025] Open
Abstract
Triple-negative breast cancer (TNBC) possesses high malignant and metastatic rates among all subtypes. Chemotherapy is a standard of care for TNBC but only a small moiety of patients achieved complete relief (CR) after chemotherapy. The recent concept of tumor ecosystem has provided new insights into solutions from an approach of enhancing anti-tumoral immunity of macrophages. We hereby observed a positive correlation of YTHDC2 abundance with anti-tumoral gene markers of macrophages. YTHDC2-high macrophages also exerted interactions with other immune cells such as T helper cells, cytotoxic T cells, and NK cells. Further investigation on the transcriptional regulatory network identified six transcriptional factors upregulated by YTHDC2, and they together influenced the expressions of TWISTNB and the oncogene MYC. Additionally, our survival analysis prompted that YTHDC2 is prognostic of higher chemo-therapeutic efficacy and better survival outcomes. We demonstrated that ample macrophage YTHDC2 indicates anti-tumoral phenotype polarization and propitious survival outcome in post-treatment TNBC patients (Clinical trial registry name: Chinese Clinical Trial Registry, Registration No.: ChiCTR2400084513, Registration Date: 2024-05-20).
Collapse
Affiliation(s)
- Hao Jin
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Dongbo Zhang
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yufan Ma
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lanlan Meng
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, China
| | - Songyin Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Hongjun Su
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jiannan Xu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yandan Yao
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China.
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, China.
| |
Collapse
|
15
|
Manoharan MS, Lee GC, Harper N, Meunier JA, Restrepo MI, Jimenez F, Karekatt S, Branum AP, Gaitan AA, Andampour K, Smith AM, Mader M, Noronha M, Tripathy D, Zhang N, Moreira AG, Pandranki L, Sanchez-Reilly S, Trinh HD, Barnett C, Angel L, Segal LN, Nicholson S, Clark RA, He W, Okulicz JF, Ahuja SK. The 15-Year Survival Advantage: Immune Resilience as a Salutogenic Force in Healthy Aging. Aging Cell 2025:e70063. [PMID: 40264357 DOI: 10.1111/acel.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Human aging presents an evolutionary paradox: while aging rates remain constant, healthspan and lifespan vary widely. We address this conundrum via salutogenesis-the active production of health-through immune resilience (IR), the capacity to resist disease despite aging and inflammation. Analyzing ~17,500 individuals across lifespan stages and inflammatory challenges, we identified a core salutogenic mechanism: IR centered on TCF7, a conserved transcription factor maintaining T-cell stemness and regenerative potential. IR integrates innate and adaptive immunity to counter three aging and mortality drivers: chronic inflammation (inflammaging), immune aging, and cellular senescence. By mitigating these aging mechanisms, IR confers survival advantages: At age 40, individuals with poor IR face a 9.7-fold higher mortality rate-a risk equivalent to that of 55.5-year-olds with optimal IR-resulting in a 15.5-year gap in survival. Optimal IR preserves youthful immune profiles at any age, enhances vaccine responses, and reduces burdens of cardiovascular disease, Alzheimer's, and serious infections. Two key salutogenic evolutionary themes emerge: first, female-predominant IR, including TCF7, likely reflects evolutionary pressures favoring reproductive success and caregiving; second, midlife (40-70 years) is a critical window where optimal IR reduces mortality by 69%. After age 70, mortality rates converge between resilient and non-resilient groups, reflecting biological limits on longevity extension. TNFα-blockers restore salutogenesis pathways, indicating IR delays aging-related processes rather than altering aging rates. By reframing aging as a salutogenic-pathogenic balance, we establish TCF7-centered IR as central to healthy longevity. Targeted midlife interventions to enhance IR offer actionable strategies to maximize healthspan before biological constraints limit benefits.
Collapse
Affiliation(s)
- Muthu Saravanan Manoharan
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Grace C Lee
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Pharmacotherapy Education and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Nathan Harper
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Justin A Meunier
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Marcos I Restrepo
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Fabio Jimenez
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Sreenath Karekatt
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Anne P Branum
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Alvaro A Gaitan
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Kian Andampour
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Alisha M Smith
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Michael Mader
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Michelle Noronha
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Devjit Tripathy
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Nu Zhang
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Alvaro G Moreira
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Lavanya Pandranki
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Sandra Sanchez-Reilly
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Hanh D Trinh
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Clea Barnett
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Luis Angel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Leopoldo N Segal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Susannah Nicholson
- Department of Surgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Robert A Clark
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Weijing He
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | | | - Sunil K Ahuja
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| |
Collapse
|
16
|
Lyons H, Pradhan P, Prakasam G, Vashishtha S, Li X, Eppert M, Fornero C, Tcheuyap VT, McGlynn K, Yu Z, Raju DR, Koduru PR, Xing C, Kapur P, Brugarolas J, Sabari BR. RNA polymerase II partitioning is a shared feature of diverse oncofusion condensates. Cell 2025:S0092-8674(25)00404-0. [PMID: 40286793 DOI: 10.1016/j.cell.2025.04.002] [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: 05/17/2024] [Revised: 12/12/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
Condensates regulate transcription by selectively compartmentalizing biomolecules, yet the rules of specificity and their relationship to function remain enigmatic. To identify rules linked to function, we leverage the genetic selection bias of condensate-promoting oncofusions. Focusing on the three most frequent oncofusions driving translocation renal cell carcinoma, we find that they promote the formation of condensates that activate transcription by gain-of-function RNA polymerase II partitioning through a shared signature of elevated π and π-interacting residues and depletion of aliphatic residues. This signature is shared among a broad set of DNA-binding oncofusions associated with diverse cancers. We find that this signature is necessary and sufficient for RNA polymerase II partitioning, gene activation, and cancer cell phenotypes. Our results reveal that dysregulated condensate specificity is a shared molecular mechanism of diverse oncofusions, highlighting the functional role of condensate composition and the power of disease genetics in investigating relationships between condensate specificity and function.
Collapse
Affiliation(s)
- Heankel Lyons
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Prashant Pradhan
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gopinath Prakasam
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hematology-Oncology Division, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shubham Vashishtha
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiang Li
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mikayla Eppert
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christy Fornero
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vanina T Tcheuyap
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hematology-Oncology Division, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kathleen McGlynn
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ze Yu
- Eugene McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dinesh Ravindra Raju
- Eugene McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Prasad R Koduru
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Lyda Hill Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Peter O'Donnell School of Public Health, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Payal Kapur
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hematology-Oncology Division, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Benjamin R Sabari
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
17
|
Li WJ, Chen YC, Lin YA, Zou YQ, Hu GS, Yang JJ, Nie XY, Li MY, Wang YR, He YH, Zhao Y, Tan YH, Deng X, He WL, Cheng Y, Fu FM, Liu W. Hypoxia-induced PRMT1 methylates HIF2β to promote breast tumorigenesis via enhancing glycolytic gene transcription. Cell Rep 2025; 44:115487. [PMID: 40173041 DOI: 10.1016/j.celrep.2025.115487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 01/28/2025] [Accepted: 03/07/2025] [Indexed: 04/04/2025] Open
Abstract
Hypoxia-induced metabolic reprogramming is closely linked to breast cancer progression. Through transcriptomic analysis, we identified PRMT1 as a direct target of hypoxia-inducible factor 1α (HIF1α) under hypoxic conditions in breast cancer cells. In turn, PRMT1 enhances the expression of HIF1α-driven glycolytic genes. Mechanistically, PRMT1 methylates HIF2β at arginine 42, facilitating the formation, chromatin binding, and the transcriptional activity of the HIF1α/HIF2β heterodimer. Genetic and pharmacological inhibition of PRMT1 suppresses HIF2β methylation, HIF1α/HIF2β heterodimer formation, chromatin binding, glycolytic gene expression, lactate production, and the malignant behaviors of breast cancer cells. Moreover, combination treatment with iPRMT1, a PRMT1 inhibitor, and menadione, an HIF1α/P300 interaction inhibitor, demonstrates synergistic effects in suppressing breast tumor growth. Clinically, PRMT1 and PRMT1-mediated HIF2β methylation were significantly elevated in breast tumors compared with adjacent normal tissues. In conclusion, our findings reveal the critical role of PRMT1-mediated arginine methylation in glycolytic gene expression, metabolic reprogramming, and breast tumor growth.
Collapse
Affiliation(s)
- Wen-Juan Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yan-Chao Chen
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yi-An Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yi-Qin Zou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Guo-Sheng Hu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jing-Jing Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xin-Yu Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Mei-Yan Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yi-Ran Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yao-Hui He
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yan Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yu-Hua Tan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Wei-Ling He
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Fang-Meng Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| | - Wen Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
18
|
Monté D, Lens Z, Dewitte F, Fislage M, Aumercier M, Verger A, Villeret V. Structural basis of human Mediator recruitment by the phosphorylated transcription factor Elk-1. Nat Commun 2025; 16:3772. [PMID: 40263353 PMCID: PMC12015215 DOI: 10.1038/s41467-025-59014-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
One function of Mediator complex subunit MED23 is to mediate transcriptional activation by the phosphorylated transcription factor Elk-1, in response to the Ras-MAPK signaling pathway. Using cryogenic electron microscopy, we solve a 3.0 Å structure of human MED23 complexed with the phosphorylated activation domain of Elk-1. Elk-1 binds to MED23 via a hydrophobic sequence PSIHFWSTLSPP containing one phosphorylated residue (S383p), which forms a tight turn around the central Phenylalanine. Binding of Elk-1 induces allosteric changes in MED23 that propagate to the opposite face of the subunit, resulting in the dynamic behavior of a 19-residue segment, which alters the molecular surface of MED23. We design a specific MED23 mutation (G382F) that disrupts Elk--1 binding and consequently impairs Elk-1-dependent serum-induced activation of target genes in the Ras-Raf-MEK-ERK signaling pathway. The structure provides molecular details and insights into a Mediator subunit-transcription factor interface.
Collapse
Affiliation(s)
- Didier Monté
- CNRS EMR 9002 Integrative Structural Biology, Inserm U 1167 - RID-AGE, Univ. Lille, CHU Lille, Institut Pasteur de Lille, Lille, France.
| | - Zoé Lens
- CNRS EMR 9002 Integrative Structural Biology, Inserm U 1167 - RID-AGE, Univ. Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Frédérique Dewitte
- CNRS EMR 9002 Integrative Structural Biology, Inserm U 1167 - RID-AGE, Univ. Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Marcus Fislage
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, Brussels, Belgium
| | - Marc Aumercier
- CNRS EMR 9002 Integrative Structural Biology, Inserm U 1167 - RID-AGE, Univ. Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Alexis Verger
- CNRS EMR 9002 Integrative Structural Biology, Inserm U 1167 - RID-AGE, Univ. Lille, CHU Lille, Institut Pasteur de Lille, Lille, France.
| | - Vincent Villeret
- CNRS EMR 9002 Integrative Structural Biology, Inserm U 1167 - RID-AGE, Univ. Lille, CHU Lille, Institut Pasteur de Lille, Lille, France.
| |
Collapse
|
19
|
Xu S, Peng C, Ren R, Lu H, Zhao H, Xia S, Shen Y, Xu B, Zhang H, Cheng X, Blobel GA, Lan X. SWI/SNF complex-mediated ZNF410 cooperative binding maintains chromatin accessibility and enhancer activity. Cell Rep 2025; 44:115476. [PMID: 40158221 DOI: 10.1016/j.celrep.2025.115476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/21/2025] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
The clustering of multiple transcription factor binding sites (TFBSs) for the same TF has proved to be a pervasive feature of cis-regulatory elements in the eukaryotic genome. However, the contribution of binding sites within the homotypic clusters of TFBSs (HCTs) to TF binding and target gene expression remains to be understood. Here, we characterize the CHD4 enhancers that harbor unique functional ZNF410 HCTs genome wide. We uncover that ZNF410 controls chromatin accessibility and activity of the CHD4 enhancer regions. We demonstrate that ZNF410 binds to the HCTs in a collaborative fashion, further conferring transcriptional activation. In particular, three ZNF410 motifs (sub-HCTs) located at 3' end of the distal enhancer act as "switch motifs" to control chromatin accessibility and enhancer activity. Mechanistically, the SWI/SNF complex is selectively required to mediate cooperative ZNF410 binding for CHD4 expression. Together, our findings expose a complex functional hierarchy of homotypic clustered motifs, which cooperate to fine-tune target gene expression.
Collapse
Affiliation(s)
- Siyuan Xu
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chuxuan Peng
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ren Ren
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Haowen Lu
- Department of Urology, School of Medicine, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Han Zhao
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Sijian Xia
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China; Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China
| | - Yijie Shen
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bin Xu
- Department of Urology, School of Medicine, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Haoyue Zhang
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Xiaodong Cheng
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gerd A Blobel
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xianjiang Lan
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
20
|
Xiao L, Jin H, Dang Y, Zhao P, Li S, Shi Y, Wang S, Zhang K. DUX-mediated configuration of p300/CBP drives minor zygotic genome activation independent of its catalytic activity. Cell Rep 2025; 44:115544. [PMID: 40202846 DOI: 10.1016/j.celrep.2025.115544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/18/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
Maternal-deposited factors initiate zygotic genome activation (ZGA), driving the maternal-to-zygotic transition; however, the coordination between maternal coactivators and transcription factors (TFs) in this process remains unclear. In this study, by profiling the dynamic landscape of p300 during mouse ZGA, we reveal its role in promoting RNA polymerase II (Pol II) pre-configuration at ZGA gene regions and sequentially establishing enhancer activity and regulatory networks. Moreover, p300/CBP-catalyzed acetylation drives Pol II elongation and minor ZGA gene expression by inducing pivotal TFs such as Dux. Remarkably, the supplementation of exogenous Dux rescues ZGA failure and developmental defects caused by the loss of p300/CBP acetylation. DUX functions as a pioneer factor, guiding p300 and Pol II to minor ZGA gene regions and activating them in a manner dependent on the non-catalytic functions of p300/CBP. Together, our findings reveal a mutual dependency between p300/CBP and DUX, highlighting their coordinated role in regulating minor ZGA activation.
Collapse
Affiliation(s)
- Lieying Xiao
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hao Jin
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yanna Dang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Panpan Zhao
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shuang Li
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yan Shi
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shaohua Wang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Kun Zhang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
21
|
Norrie JL, Lupo MS, Little DR, Shirinifard A, Mishra A, Zhang Q, Geiger N, Putnam D, Djekidel N, Ramirez C, Xu B, Dundee JM, Yu J, Chen X, Dyer MA. Latent epigenetic programs in Müller glia contribute to stress and disease response in the retina. Dev Cell 2025; 60:1199-1216.e7. [PMID: 39753128 PMCID: PMC12014377 DOI: 10.1016/j.devcel.2024.12.014] [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: 10/31/2023] [Revised: 07/09/2024] [Accepted: 12/06/2024] [Indexed: 04/24/2025]
Abstract
Previous studies have demonstrated the dynamic changes in chromatin structure during retinal development correlate with changes in gene expression. However, those studies lack cellular resolution. Here, we integrate single-cell RNA sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) with bulk data to identify cell-type-specific changes in chromatin structure during human and murine development. Although promoter activity is correlated with chromatin accessibility, we discovered several hundred genes that were transcriptionally silent but had accessible chromatin at their promoters. Most of those silent/accessible gene promoters were in Müller glial cells, which function to maintain retinal homeostasis and respond to stress, injury, or disease. We refer to these as "pliancy genes" because they allow the Müller glia to rapidly change their gene expression and cellular state in response to retinal insults. The Müller glial cell pliancy program is established during development, and we demonstrate that pliancy genes are important for regulating inflammation in the murine retina in vivo.
Collapse
Affiliation(s)
- Jackie L Norrie
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Marybeth S Lupo
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Danielle R Little
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Akhilesh Mishra
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Qiong Zhang
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Natalie Geiger
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Daniel Putnam
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Nadhir Djekidel
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Cody Ramirez
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jacob M Dundee
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jiang Yu
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiang Chen
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael A Dyer
- Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
22
|
Liang P, Chen JJ, Yang X, Long R, Li Y, Wang ZL, Yang PL, Liang YD. Association and functional study of ATP6V1D and GPHN gene polymorphisms with depression in Chinese population. World J Psychiatry 2025; 15:102182. [PMID: 40309610 PMCID: PMC12038669 DOI: 10.5498/wjp.v15.i4.102182] [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: 10/11/2024] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Depression is a disease with a significant global social burden. Single nucleotide polymorphisms (SNPs) are correlated with the development of depression. This study investigates the relationship between polymorphisms in the GPHN and ATP6V1D gene promoter regions and susceptibility to depression in the Chinese population. AIM To provide new insights into identifying SNPs for predicting depression in the Chinese population. METHODS We conducted a case-control study involving 555 individuals with depression and 509 healthy controls. GPHN rs8020095 and ATP6V1D rs3759755, rs10144417, rs2031564, and rs8016024 in the promoter region were genotyped using next-generation sequencing. Dual luciferase reporter genes were employed to assess the transcriptional activity of promoter regions for each SNP genotype, with transcription factors binding to each site predicted using the JASPAR database. RESULTS Compared to healthy controls, the ATP6V1D promoter rs10144417 AG genotype (P = 0.015), rs3759755 AC/CC genotype (P = 0.036), and GPHN gene rs8020095 GA and AA genotypes (GA: P = 0.028, GG: P = 0.025) were significantly associated with a lower prevalence of depression. Linked disequilibria were present in five SNPs, with the AGATA haplotype frequency in patients significantly lower than in healthy subjects (P = 0.023). Luciferase activity of the rs3759755-A recombinant plasmid was significantly higher than that of the rs3759755-C recombinant plasmid (P = 0.026), and the rs8020095-A recombinant plasmid activity was significantly higher than that of the rs8020095-G recombinant plasmid (P = 0.001). Transcription factors orthodenticle homeobox 2, orthodenticle homeobox 1, forkhead box L1, NK homeobox 3-1, and nuclear factor, interleukin 3 regulated demonstrated binding affinity with rs3759755A > C and rs8020095A > G. CONCLUSION This study demonstrates that SNPs (rs3759755 and rs10144417) in the promoter region of the ATP6V1D and SNP (rs8020095) of GPHN are indeed associated with susceptibility to depression.
Collapse
Affiliation(s)
- Peng Liang
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Jing-Jie Chen
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Xue Yang
- Department of Geriatric Psychiatry, The First Psychiatric Hospital of Harbin, Harbin 150001, Heilongjiang Province, China
| | - Rui Long
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Yue Li
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Zi-Ling Wang
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Ping-Liang Yang
- Department of Anesthesiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Yun-Dan Liang
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| |
Collapse
|
23
|
Guo W, Wang S, Yang Z, Dong Y, Xia Z, Xue W, Zhang C. SAP30 promotes clear cell renal cell carcinoma proliferation and inhibits apoptosis through the MT1G axis. Eur J Med Res 2025; 30:306. [PMID: 40247376 PMCID: PMC12007153 DOI: 10.1186/s40001-025-02440-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Sin3A-associated protein 30 (SAP30) is a crucial component of the SIN/HDAC histone deacetylase complex and acts as a scaffold that facilitates target gene binding. SAP30 is highly expressed in various tumours; however, its role in renal cell carcinoma (RCC) remains unclear. In our study, we observed the upregulation of SAP30 in clear cell renal cell carcinoma (ccRCC) tissues, and its elevated expression was correlated with a poor prognosis. Previous research has suggested that SAP30 may influence the growth, proliferation, and apoptosis of RCC cells. Gene Ontology (GO) analysis of the downstream regulatory targets of SAP30 revealed that SAP30 suppressed the expression of MT1G, a protein that binds to p53. Mechanistically, SAP30 inhibited MT1G transcription, thereby impairing the function of MT1G in delivering zinc ions to p53, which diminished p53 activity. Moreover, reduced MT1G levels attenuated the inhibitory effect of MT1G on MDM2, further destabilizing p53. Consequently, this cascade promoted RCC progression. In conclusion, our findings indicate that SAP30 inhibits the p53 pathway through MT1G suppression, suggesting that SAP30 and MT1G are potential prognostic markers and therapeutic targets for RCC.
Collapse
Affiliation(s)
- Wei Guo
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Shuwen Wang
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Zitong Yang
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yu Dong
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Zhinan Xia
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wei Xue
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Cheng Zhang
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
24
|
Kock KH, Tan LM, Han KY, Ando Y, Jevapatarakul D, Chatterjee A, Lin QXX, Buyamin EV, Sonthalia R, Rajagopalan D, Tomofuji Y, Sankaran S, Park MS, Abe M, Chantaraamporn J, Furukawa S, Ghosh S, Inoue G, Kojima M, Kouno T, Lim J, Myouzen K, Nguantad S, Oh JM, Rayan NA, Sarkar S, Suzuki A, Thungsatianpun N, Venkatesh PN, Moody J, Nakano M, Chen Z, Tian C, Zhang Y, Tong Y, Tan CTY, Tizazu AM, Loh M, Hwang YY, Ho RC, Larbi A, Ng TP, Won HH, Wright FA, Villani AC, Park JE, Choi M, Liu B, Maitra A, Pithukpakorn M, Suktitipat B, Ishigaki K, Okada Y, Yamamoto K, Carninci P, Chambers JC, Hon CC, Matangkasombut P, Charoensawan V, Majumder PP, Shin JW, Park WY, Prabhakar S. Asian diversity in human immune cells. Cell 2025; 188:2288-2306.e24. [PMID: 40112801 DOI: 10.1016/j.cell.2025.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/03/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
The relationships of human diversity with biomedical phenotypes are pervasive yet remain understudied, particularly in a single-cell genomics context. Here, we present the Asian Immune Diversity Atlas (AIDA), a multi-national single-cell RNA sequencing (scRNA-seq) healthy reference atlas of human immune cells. AIDA comprises 1,265,624 circulating immune cells from 619 donors, spanning 7 population groups across 5 Asian countries, and 6 controls. Though population groups are frequently compared at the continental level, we found that sub-continental diversity, age, and sex pervasively impacted cellular and molecular properties of immune cells. These included differential abundance of cell neighborhoods as well as cell populations and genes relevant to disease risk, pathogenesis, and diagnostics. We discovered functional genetic variants influencing cell-type-specific gene expression, which were under-represented in non-Asian populations, and helped contextualize disease-associated variants. AIDA enables analyses of multi-ancestry disease datasets and facilitates the development of precision medicine efforts in Asia and beyond.
Collapse
Affiliation(s)
- Kian Hong Kock
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Le Min Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Kyung Yeon Han
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Yoshinari Ando
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Laboratory for Transcriptome Technology, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Damita Jevapatarakul
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Ankita Chatterjee
- John C. Martin Centre for Liver Research and Innovations, Sonarpur, Kolkata 700150, India
| | - Quy Xiao Xuan Lin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Eliora Violain Buyamin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Radhika Sonthalia
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Deepa Rajagopalan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Yoshihiko Tomofuji
- Laboratory for Systems Genetics, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Statistical Genetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shvetha Sankaran
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Mi-So Park
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Mai Abe
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Juthamard Chantaraamporn
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Seiko Furukawa
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Supratim Ghosh
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India
| | - Gyo Inoue
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Miki Kojima
- Laboratory for Transcriptome Technology, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Tsukasa Kouno
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Jinyeong Lim
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Keiko Myouzen
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Sarintip Nguantad
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Jin-Mi Oh
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Nirmala Arul Rayan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Sumanta Sarkar
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Narita Thungsatianpun
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Prasanna Nori Venkatesh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Jonathan Moody
- Laboratory for Genome Information Analysis, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Masahiro Nakano
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Ziyue Chen
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Chi Tian
- Department of Pharmacy, Faculty of Science, National University of Singapore (NUS), Singapore 117543, Singapore
| | - Yuntian Zhang
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine (YLLSoM), NUS, Singapore 119228, Singapore
| | - Yihan Tong
- Department of Pharmacy, Faculty of Science, National University of Singapore (NUS), Singapore 117543, Singapore
| | - Crystal T Y Tan
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Anteneh Mehari Tizazu
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Marie Loh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Nanyang Technological University (NTU), Lee Kong Chian School of Medicine (LKCMedicine), 11 Mandalay Road, Singapore 308232, Singapore
| | - You Yi Hwang
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Roger C Ho
- Department of Psychological Medicine, YLLSoM, NUS, 1E Kent Ridge Road, Singapore 119228, Singapore; Institute for Health Innovation & Technology, NUS, 14 Medical Drive, Singapore 117599, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Tze Pin Ng
- Department of Geriatric Medicine, Khoo Teck Puat Hospital, Singapore 768828, Singapore; St Luke's Hospital, Singapore 659674, Singapore; Geriatric Education and Research Institute, Singapore 768024, Singapore
| | - Hong-Hee Won
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Fred A Wright
- Department of Biological Sciences, Bioinformatics Research Center, and Department of Statistics, North Carolina State University, Raleigh, NC 27695, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, and Mass General Cancer Center, Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon 34051, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Boxiang Liu
- Department of Pharmacy, Faculty of Science, National University of Singapore (NUS), Singapore 117543, Singapore; Department of Biomedical Informatics, Yong Loo Lin School of Medicine (YLLSoM), NUS, Singapore 119228, Singapore; Precision Medicine Translational Research Programme, NUS Centre for Cancer Research, and Cardiovascular-Metabolic Disease Translational Research Programme, YLLSoM, NUS, Singapore 119228, Singapore
| | - Arindam Maitra
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India
| | - Manop Pithukpakorn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Bhoom Suktitipat
- Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand; Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yukinori Okada
- Laboratory for Systems Genetics, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Statistical Genetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan; Laboratory of Statistical Immunology, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Premium Research Institute for Human Metaverse Medicine, Osaka University, Suita 565-0871, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Piero Carninci
- Laboratory for Transcriptome Technology, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Genomics Research Center, Fondazione Human Technopole, Viale Rita Levi-Montalcini, 1 - Area MIND, Milano, Lombardy 20157, Italy
| | - John C Chambers
- Nanyang Technological University (NTU), Lee Kong Chian School of Medicine (LKCMedicine), 11 Mandalay Road, Singapore 308232, Singapore
| | - Chung-Chau Hon
- Laboratory for Genome Information Analysis, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-3-2 Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan
| | - Ponpan Matangkasombut
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Varodom Charoensawan
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand; Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Partha P Majumder
- John C. Martin Centre for Liver Research and Innovations, Sonarpur, Kolkata 700150, India; Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India
| | - Jay W Shin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea.
| | - Shyam Prabhakar
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Nanyang Technological University (NTU), Lee Kong Chian School of Medicine (LKCMedicine), 11 Mandalay Road, Singapore 308232, Singapore; Cancer Science Institute of Singapore, NUS, 14 Medical Drive, Singapore 117599, Singapore.
| |
Collapse
|
25
|
Horton JR, Yu M, Zhou J, Tran M, Anakal RR, Lu Y, Blumenthal RM, Zhang X, Huang Y, Zhang X, Cheng X. Multimeric transcription factor BCL11A utilizes two zinc-finger tandem arrays to bind clustered short sequence motifs. Nat Commun 2025; 16:3672. [PMID: 40246927 PMCID: PMC12006351 DOI: 10.1038/s41467-025-58998-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BCL11A, a transcription factor, is vital for hematopoiesis, including B and T cell maturation and the fetal-to-adult hemoglobin switch. Mutations in BCL11A are linked to neurodevelopmental disorders. BCL11A contains two DNA-binding zinc-finger arrays, low-affinity ZF2-3 and high-affinity ZF4-6, separated by a 300-amino-acid linker. ZF2-3 and ZF4-5 share 73% identity, including five out of six DNA base-interacting residues. These arrays bind similar short sequence motifs in clusters, with the linker enabling a broader binding span. Crystallographic structures of ZF4-6, in complex with oligonucleotides from the β-globin locus region, reveal DNA sequence recognition by residues Asn756 (ZF4), Lys784 and Arg787 (ZF5). A Lys784-to-Thr mutation, linked to a neurodevelopmental disorder with persistent fetal globin expression, reduces DNA binding over 10-fold but gains interaction with a variable base pair. BCL11A isoforms may form oligomers, enhancing chromatin occupancy and repressor functions by allowing multiple copies of both low- and high-affinity ZF arrays to bind DNA. These distinctive properties, apparently conserved among vertebrates, provide essential functional flexibility to this crucial regulator.
Collapse
Affiliation(s)
- John R Horton
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Meigen Yu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jujun Zhou
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Melody Tran
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rithvi R Anakal
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Robert M Blumenthal
- Department of Medical Microbiology and Immunology, and Program in Bioinformatics, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Xiaotian Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
| | - Xing Zhang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaodong Cheng
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
26
|
Zhang G, Song C, Yin M, Liu L, Zhang Y, Li Y, Zhang J, Guo M, Li C. TRAPT: a multi-stage fused deep learning framework for predicting transcriptional regulators based on large-scale epigenomic data. Nat Commun 2025; 16:3611. [PMID: 40240358 PMCID: PMC12003887 DOI: 10.1038/s41467-025-58921-0] [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: 06/17/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
It is challenging to identify regulatory transcriptional regulators (TRs), which control gene expression via regulatory elements and epigenomic signals, in context-specific studies on the onset and progression of diseases. The use of large-scale multi-omics epigenomic data enables the representation of the complex epigenomic patterns of control of the regulatory elements and the regulators. Herein, we propose Transcription Regulator Activity Prediction Tool (TRAPT), a multi-modality deep learning framework, which infers regulator activity by learning and integrating the regulatory potentials of target gene cis-regulatory elements and genome-wide binding sites. The results of experiments on 570 TR-related datasets show that TRAPT outperformed state-of-the-art methods in predicting the TRs, especially in terms of forecasting transcription co-factors and chromatin regulators. Moreover, we successfully identify key TRs associated with diseases, genetic variations, cell-fate decisions, and tissues. Our method provides an innovative perspective on identifying TRs by using epigenomic data.
Collapse
Affiliation(s)
- Guorui Zhang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Chao Song
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
- School of Computer, University of South China, Hengyang, Hunan, 421001, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan, 421001, China
| | - Mingxue Yin
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Liyuan Liu
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yuexin Zhang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
| | - Ye Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jianing Zhang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
| | - Maozu Guo
- School of Intelligence Science and Technology, Beijing University of Civil Engineering and Architecture, Beijing, 100044, China.
| | - Chunquan Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China.
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- School of Computer, University of South China, Hengyang, Hunan, 421001, China.
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
27
|
Poudel P, Miteva MA, Alexov E. Strategies for in Silico Drug Discovery to Modulate Macromolecular Interactions Altered by Mutations. FRONT BIOSCI-LANDMRK 2025; 30:26339. [PMID: 40302318 DOI: 10.31083/fbl26339] [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: 08/29/2024] [Revised: 09/22/2024] [Accepted: 10/09/2024] [Indexed: 05/02/2025]
Abstract
Most human diseases have genetic components, frequently single nucleotide variants (SNVs), which alter the wild type characteristics of macromolecules and their interactions. A straightforward approach for correcting such SNVs-related alterations is to seek small molecules, potential drugs, that can eliminate disease-causing effects. Certain disorders are caused by altered protein-protein interactions, for example, Snyder-Robinson syndrome, the therapy for which focuses on the development of small molecules that restore the wild type homodimerization of spermine synthase. Other disorders originate from altered protein-nucleic acid interactions, as in the case of cancer; in these cases, the elimination of disease-causing effects requires small molecules that eliminate the effect of mutation and restore wild type p53-DNA affinity. Overall, especially for complex diseases, pathogenic mutations frequently alter macromolecular interactions. This effect can be direct, i.e., the alteration of wild type affinity and specificity, or indirect via alterations in the concentration of the binding partners. Here, we outline progress made in methods and strategies to computationally identify small molecules capable of altering macromolecular interactions in a desired manner, reducing or increasing the binding affinity, and eliminating the disease-causing effect. When applicable, we provide examples of the outlined general strategy. Successful cases are presented at the end of the work.
Collapse
Affiliation(s)
- Pitambar Poudel
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Maria A Miteva
- Université Paris Cité, CNRS UMR 8038 CiTCoM, Inserm, U1268 MCTR Paris, France
| | - Emil Alexov
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
28
|
Mahendrawada L, Warfield L, Donczew R, Hahn S. Low overlap of transcription factor DNA binding and regulatory targets. Nature 2025:10.1038/s41586-025-08916-0. [PMID: 40240607 DOI: 10.1038/s41586-025-08916-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 03/19/2025] [Indexed: 04/18/2025]
Abstract
DNA sequence-specific transcription factors (TFs) modulate transcription and chromatin architecture, acting from regulatory sites in enhancers and promoters of eukaryotic genes1,2. How multiple TFs cooperate to regulate individual genes is still unclear. In yeast, most TFs are thought to regulate transcription via binding to upstream activating sequences, which are situated within a few hundred base pairs upstream of the regulated gene3. Although this model has been validated for individual TFs and specific genes, it has not been tested in a systematic way. Here we integrated information on the binding and expression targets for the near-complete set of yeast TFs and show that, contrary to expectations, there are few TFs with dedicated activator or repressor roles, and that most TFs have a dual function. Although nearly all protein-coding genes are regulated by one or more TFs, our analysis revealed limited overlap between TF binding and gene regulation. Rapid depletion of many TFs also revealed many regulatory targets that were distant from detectable TF binding sites, suggesting unexpected regulatory mechanisms. Our study provides a comprehensive survey of TF functions and offers insights into interactions between the set of TFs expressed in a single cell type and how they contribute to the complex programme of gene regulation.
Collapse
Affiliation(s)
| | | | - Rafal Donczew
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Steven Hahn
- Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
29
|
Bernhard C, Geles K, Pawlak G, Dhifli W, Dispot A, Dusol J, Kondratova M, Martin S, Messé M, Reita D, Tulasne D, Van Seuningen I, Entz-Werle N, Ciafrè SA, Dontenwill M, Elati M. A coregulatory influence map of glioblastoma heterogeneity and plasticity. NPJ Precis Oncol 2025; 9:110. [PMID: 40234567 PMCID: PMC12000621 DOI: 10.1038/s41698-025-00890-0] [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: 09/22/2024] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
We present GBM-cRegMap, an online resource providing a comprehensive coregulatory influence network perspective on glioblastoma (GBM) heterogeneity and plasticity. Using representation learning algorithms, we derived two components of this resource: GBM-CoRegNet, a highly specific coregulatory network of tumor cells, and GBM-CoRegMap, a unified network influence map based on 1612 tumors from 16 studies. As a widely applicable closed-loop system connecting cellular models and tumors, GBM-cRegMap will provide the GBM research community with an easy-to-use web tool ( https://gbm.cregmap.com ) that maps any existing or newly generated transcriptomic "query" data to a reference coregulatory network and a large-scale manifold of disease heterogeneity. Using GBM-cRegMap, we demonstrated the synergy between the two components by refining the molecular classification of GBM, identifying potential key regulators, and aligning the transcriptional profiles of tumors and in vitro models. Through the amalgamation of a vast dataset, we validated the proneural (PN)-mesenchymal (MES) axis and identified three subclasses of classical (CL) tumors: astrocyte-like (CL-A), epithelial basal-like (CL-B), and cilium-rich (CL-C). We revealed the CL-C subclass, an intermediate state demonstrating the plasticity of GBM cells along the PN-MES axis under chemotherapy. We identified key regulators, such as PAX8, and NKX2.5, potentially involved in temozolomide (TMZ) resistance. Notably, NKX2.5, more expressed in higher-grade gliomas, negatively impacts patient survival, and regulates genes involved in glucose metabolism.
Collapse
Affiliation(s)
- Chloé Bernhard
- UMR7021 CNRS, University of Strasbourg, Illkirch, France
| | - Konstantinos Geles
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, F-59000, France
| | - Geoffrey Pawlak
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, F-59000, France
| | - Wajdi Dhifli
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, F-59000, France
| | - Aurélien Dispot
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, F-59000, France
| | - Jules Dusol
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, F-59000, France
| | - Maria Kondratova
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, F-59000, France
| | - Sophie Martin
- UMR7021 CNRS, University of Strasbourg, Illkirch, France
| | - Mélissa Messé
- UMR7021 CNRS, University of Strasbourg, Illkirch, France
| | - Damien Reita
- UMR7021 CNRS, University of Strasbourg, Illkirch, France
- Department of Cancer Molecular Genetics, Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, 67200, Strasbourg, France
| | - David Tulasne
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, F-59000, France
| | - Isabelle Van Seuningen
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, F-59000, France
| | - Natacha Entz-Werle
- UMR7021 CNRS, University of Strasbourg, Illkirch, France
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 67098, Strasbourg, France
| | - Silvia Anna Ciafrè
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | | | - Mohamed Elati
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, F-59000, France.
| |
Collapse
|
30
|
Martyn GE, Montgomery MT, Jones H, Guo K, Doughty BR, Linder J, Bisht D, Xia F, Cai XS, Chen Z, Cochran K, Lawrence KA, Munson G, Pampari A, Fulco CP, Sahni N, Kelley DR, Lander ES, Kundaje A, Engreitz JM. Rewriting regulatory DNA to dissect and reprogram gene expression. Cell 2025:S0092-8674(25)00352-6. [PMID: 40245860 DOI: 10.1016/j.cell.2025.03.034] [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: 12/20/2023] [Revised: 12/16/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025]
Abstract
Regulatory DNA provides a platform for transcription factor binding to encode cell-type-specific patterns of gene expression. However, the effects and programmability of regulatory DNA sequences remain difficult to map or predict. Here, we develop variant effects from flow-sorting experiments with CRISPR targeting screens (Variant-EFFECTS) to introduce hundreds of designed edits to endogenous regulatory DNA and quantify their effects on gene expression. We systematically dissect and reprogram 3 regulatory elements for 2 genes in 2 cell types. These data reveal endogenous binding sites with effects specific to genomic context, transcription factor motifs with cell-type-specific activities, and limitations of computational models for predicting the effect sizes of variants. We identify small edits that can tune gene expression over a large dynamic range, suggesting new possibilities for prime-editing-based therapeutics targeting regulatory DNA. Variant-EFFECTS provides a generalizable tool to dissect regulatory DNA and to identify genome editing reagents that tune gene expression in an endogenous context.
Collapse
Affiliation(s)
- Gabriella E Martyn
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA 94305, USA
| | - Michael T Montgomery
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA 94305, USA
| | - Hank Jones
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA 94305, USA
| | - Katherine Guo
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA 94305, USA
| | - Benjamin R Doughty
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Johannes Linder
- Calico Life Sciences LLC, South San Francisco, CA 94080, USA
| | - Deepa Bisht
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Fan Xia
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA 94305, USA
| | - Xiangmeng S Cai
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Ziwei Chen
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Kelly Cochran
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Kathryn A Lawrence
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Glen Munson
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anusri Pampari
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Charles P Fulco
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA; Quantitative and Computational Biosciences Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - David R Kelley
- Calico Life Sciences LLC, South San Francisco, CA 94080, USA
| | - Eric S Lander
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, MIT, Cambridge, MA 02139, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Jesse M Engreitz
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA 94305, USA; Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
31
|
Cadefau-Fabregat M, Martínez-Cebrián G, Lorenzi L, Weiss FD, Frank AK, Castelló-García JM, Julià-Vilella E, Gámez-García A, Yera L, de Castro CPM, Wang YF, Meissner F, Vaquero A, Merkenschlager M, Porse BT, Cuartero S. Mutant CEBPA promotes tolerance to inflammatory stress through deficient AP-1 activation. Nat Commun 2025; 16:3492. [PMID: 40221437 PMCID: PMC11993602 DOI: 10.1038/s41467-025-58712-7] [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: 07/12/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
The CEBPA transcription factor is frequently mutated in acute myeloid leukemia (AML). Mutations in the CEBPA gene, which are typically biallelic, result in the production of a shorter isoform known as p30. Both the canonical 42-kDa isoform (p42) and the AML-associated p30 isoform bind chromatin and activate transcription, but the specific transcriptional programs controlled by each protein and how they are linked to a selective advantage in AML is not well understood. Here, we show that cells expressing the AML-associated p30 have reduced baseline inflammatory gene expression and display altered dynamics of transcriptional induction in response to LPS, consequently impacting cytokine secretion. This confers p30-expressing cells an increased resistance to the adverse effects of prolonged exposure to inflammatory signals. Mechanistically, we show that these differences primarily arise from the differential regulation of AP-1 family proteins. In addition, we find that the impaired function of the AP-1 member ATF4 in p30-expressing cells alters their response to ER stress. Collectively, these findings uncover a link between mutant CEBPA, inflammation and the stress response, potentially revealing a vulnerability in AML.
Collapse
Affiliation(s)
- Maria Cadefau-Fabregat
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Doctoral Program in Biomedicine, Universitat de Barcelona (UB), Barcelona, Spain
| | | | - Lucía Lorenzi
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Felix D Weiss
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Anne-Katrine Frank
- The Finsen Laboratory, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Eric Julià-Vilella
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
- Doctoral Program in Biomedicine, Universitat de Barcelona (UB), Barcelona, Spain
| | - Andrés Gámez-García
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Laura Yera
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Carini Picardi Morais de Castro
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
- Doctoral Program in Biomedicine, Universitat de Barcelona (UB), Barcelona, Spain
| | - Yi-Fang Wang
- MRC London Institute of Medical Sciences, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Felix Meissner
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Alejandro Vaquero
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Matthias Merkenschlager
- MRC London Institute of Medical Sciences, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Bo T Porse
- The Finsen Laboratory, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sergi Cuartero
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain.
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain.
| |
Collapse
|
32
|
Saad S, Swigut T, Tabatabaee S, Lalgudi P, Jarosz DF, Wysocka J. DNA binding and mitotic phosphorylation protect polyglutamine proteins from assembly formation. Cell 2025:S0092-8674(25)00349-6. [PMID: 40239647 DOI: 10.1016/j.cell.2025.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/20/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025]
Abstract
Polyglutamine (polyQ) expansion is associated with pathogenic protein aggregation in neurodegenerative disorders. However, long polyQ tracts are also found in many transcription factors (TFs), such as FOXP2, a TF implicated in human speech. Here, we explore how FOXP2 and other glutamine-rich TFs avoid unscheduled assembly. Throughout interphase, DNA binding, irrespective of sequence specificity, has a solubilizing effect. During mitosis, multiple phosphorylation events promote FOXP2's eviction from chromatin and supplant the solubilizing function of DNA. Further, human-specific amino acid substitutions linked to the evolution of speech map to a mitotic phospho-patch, the "EVO patch," and reduce the propensity of the human FOXP2 to assemble. Fusing the pathogenic form of Huntingtin to either a DNA-binding domain, a phosphomimetic variant of this EVO patch, or a negatively charged peptide is sufficient to diminish assembly formation, suggesting that hijacking mechanisms governing solubility of glutamine-rich TFs may offer new strategies for treatment of polyQ expansion diseases.
Collapse
Affiliation(s)
- Shady Saad
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tomek Swigut
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Saman Tabatabaee
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pranav Lalgudi
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daniel F Jarosz
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
33
|
Okubo C, Nakamura M, Sato M, Shichino Y, Mito M, Takashima Y, Iwasaki S, Takahashi K. EIF3D safeguards the homeostasis of key signaling pathways in human primed pluripotency. SCIENCE ADVANCES 2025; 11:eadq5484. [PMID: 40203091 PMCID: PMC11980838 DOI: 10.1126/sciadv.adq5484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Although pluripotent stem cell (PSC) properties, such as differentiation and infinite proliferation, have been well documented within the frameworks of transcription factor networks, epigenomes, and signal transduction, they remain unclear and fragmented. Directing attention toward translational regulation as a bridge between these events can yield additional insights into previously unexplained mechanisms. Our functional CRISPR interference screen-based approach revealed that EIF3D, a translation initiation factor, is crucial for maintaining primed pluripotency. Loss of EIF3D disrupted the balance of pluripotency-associated signaling pathways, thereby compromising primed pluripotency. Moreover, EIF3D ensured robust proliferation by controlling the translation of various p53 regulators, which maintain low p53 activity in the undifferentiated state. In this way, EIF3D-mediated translation contributes to tuning the homeostasis of the primed pluripotency networks, ensuring the maintenance of an undifferentiated state with high proliferative potential. This study provides further insights into the translation network in maintaining pluripotency.
Collapse
Affiliation(s)
- Chikako Okubo
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Michiko Nakamura
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Masae Sato
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Yuichi Shichino
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
| | - Mari Mito
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
| | - Yasuhiro Takashima
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Shintaro Iwasaki
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8561, Japan
| | - Kazutoshi Takahashi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
34
|
Wang F, Zhou H, Tian Y, Wang X, Huang Y, Tu Y, Li L, Zhen H. ELK4 induced upregulation of HOMER3 promotes the proliferation and metastasis in glioma via Wnt/β-catenin/EMT signaling pathway. Biol Direct 2025; 20:48. [PMID: 40205485 PMCID: PMC11980352 DOI: 10.1186/s13062-025-00643-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
Glioma is an aggressive brain tumor characterized by its high invasiveness, which complicates prognosis and contributes to patient resistance against various treatment options. The HOMER family, consisting of HOMER1, HOMER2, and HOMER3, has been implicated in various cancers, yet their specific roles in glioma remain inadequately understood. This study conducted a comprehensive pan-cancer analysis to evaluate the expression profiles of HOMER family members across different tumor types, utilizing data from public databases such as TCGA and GTEx. Our findings indicate significant dysregulation of HOMER1, HOMER2, and HOMER3 in multiple cancers, with HOMER3 emerging as a potential prognostic biomarker, particularly for lower-grade glioma. Elevated expression levels of HOMER3 were associated with shorter overall survival and disease-specific survival in LGG patients, supported by Cox regression analysis that confirmed HOMER3 as an independent prognostic factor. Furthermore, HOMER3 expression correlated positively with advanced clinical stages and key tumor markers. To elucidate the mechanisms behind HOMER3 dysregulation, we identified ELK4 as a transcription factor that binds to the HOMER3 promoter, promoting its expression in glioma cells. Functional assays demonstrated that silencing HOMER3 significantly reduced glioma cell proliferation and metastatic potential in vitro and in vivo, highlighting its oncogenic role. Additionally, HOMER3 was found to influence the Wnt/β-catenin/EMT signaling pathway, with knockdown resulting in altered expression of critical EMT markers. Collectively, our results indicated that HOMER3 plays a crucial role in glioma progression and metastasis, underscoring its potential as a therapeutic target and prognostic biomarker in glioma management.
Collapse
Affiliation(s)
- Furong Wang
- Department of Pathology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
- Department of Pathology, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Hui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Yu Tian
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Xiaoling Wang
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Youcai Huang
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Yanyang Tu
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China.
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China.
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China.
| | - Liwen Li
- Department of Bioscience, College of Life Sciences, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| | - Haining Zhen
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, No. 127 Changle West Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
35
|
Xie Z, Sokolov I, Osmala M, Yue X, Bower G, Pett JP, Chen Y, Wang K, Cavga AD, Popov A, Teichmann SA, Morgunova E, Kvon EZ, Yin Y, Taipale J. DNA-guided transcription factor interactions extend human gene regulatory code. Nature 2025:10.1038/s41586-025-08844-z. [PMID: 40205063 DOI: 10.1038/s41586-025-08844-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 02/26/2025] [Indexed: 04/11/2025]
Abstract
In the same way that the mRNA-binding specificities of transfer RNAs define the genetic code, the DNA-binding specificities of transcription factors (TFs) form the molecular basis of the gene regulatory code1,2. The human gene regulatory code is much more complex than the genetic code, in particular because there are more than 1,600 TFs that commonly interact with each other. TF-TF interactions are required for specifying cell fate and executing cell-type-specific transcriptional programs. Despite this, the landscape of interactions between DNA-bound TFs is poorly defined. Here we map the biochemical interactions between DNA-bound TFs using CAP-SELEX, a method that can simultaneously identify individual TF binding preferences, TF-TF interactions and the DNA sequences that are bound by the interacting complexes. A screen of more than 58,000 TF-TF pairs identified 2,198 interacting TF pairs, 1,329 of which preferentially bound to their motifs arranged in a distinct spacing and/or orientation. We also discovered 1,131 TF-TF composite motifs that were markedly different from the motifs of the individual TFs. In total, we estimate that the screen identified between 18% and 47% of all human TF-TF motifs. The novel composite motifs we found were enriched in cell-type-specific elements, active in vivo and more likely to be formed between developmentally co-expressed TFs. Furthermore, TFs that define embryonic axes commonly interacted with different TFs and bound to distinct motifs, explaining how TFs with a similar specificity can define distinct cell types along developmental axes.
Collapse
Affiliation(s)
- Zhiyuan Xie
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ilya Sokolov
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Generative and Synthetic Genomics Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Maria Osmala
- Applied Tumor Genomics Program, Biomedicum, University of Helsinki, Helsinki, Finland
| | - Xue Yue
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Grace Bower
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - J Patrick Pett
- Cellular Genetics Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Yinan Chen
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Generative and Synthetic Genomics Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Kai Wang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ayse Derya Cavga
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Alexander Popov
- European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Sarah A Teichmann
- Department of Medicine and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Ekaterina Morgunova
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Evgeny Z Kvon
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Yimeng Yin
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China.
| | - Jussi Taipale
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
- Generative and Synthetic Genomics Programme, Wellcome Sanger Institute, Hinxton, UK.
- Applied Tumor Genomics Program, Biomedicum, University of Helsinki, Helsinki, Finland.
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
36
|
Jones T, Sigauke RF, Sanford L, Taatjes DJ, Allen MA, Dowell RD. TF Profiler: a transcription factor inference method that broadly measures transcription factor activity and identifies mechanistically distinct networks. Genome Biol 2025; 26:92. [PMID: 40205447 PMCID: PMC11983743 DOI: 10.1186/s13059-025-03545-2] [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: 01/08/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
TF Profiler is a method of inferring transcription factor (TF) regulatory activity, i.e., when a TF is present and actively participating in the regulation of transcription, directly from nascent sequencing assays such as PRO-seq and GRO-seq. While ChIP assays have measured DNA localization, they fall short of identifying when and where the effector domain of a transcription factor is active. Our method uses RNA polymerase activity to infer TF effector domain activity across hundreds of data sets and transcription factors. TF Profiler is broadly applicable, providing regulatory insights on any PRO-seq sample for any transcription factor with a known binding motif.
Collapse
Affiliation(s)
- Taylor Jones
- BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB 596, Boulder, CO, 80309, USA
- Biochemistry, University of Colorado Boulder, 3415 Colorado Ave., UCB 596, Boulder, CO, 80309, USA
| | - Rutendo F Sigauke
- BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB 596, Boulder, CO, 80309, USA
| | - Lynn Sanford
- BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB 596, Boulder, CO, 80309, USA
| | - Dylan J Taatjes
- Biochemistry, University of Colorado Boulder, 3415 Colorado Ave., UCB 596, Boulder, CO, 80309, USA
| | - Mary A Allen
- BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB 596, Boulder, CO, 80309, USA.
| | - Robin D Dowell
- BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB 596, Boulder, CO, 80309, USA.
- Computer Science, University of Colorado Boulder, 1111 Engineering Drive, UCB 430, Boulder, CO, 80309, USA.
- Molecular, Cellular and Developmental Biology, University of Colorado Boulder, 1945 Colorado Ave, UCB 347, Boulder, CO, 80309, USA.
| |
Collapse
|
37
|
Zhou X, Zhou X, Li J, He Y, Qiu S, Xu Y, Liu Z, Yao Y, Liu J, Wen Y, Xie S, Chen J, Liu L, Ou Z, Cai C, Lin J, Lei B, Zou F. Bclaf1 mediates super-enhancer-driven activation of POLR2A to enhance chromatin accessibility in nitrosamine-induced esophageal carcinogenesis. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138218. [PMID: 40220379 DOI: 10.1016/j.jhazmat.2025.138218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Gene-environment interactions are pivotal contributors to nitrosamine-induced esophageal carcinogenesis. While genetic mechanisms in esophageal carcinoma (ESCA) are well-defined, epigenetic drivers remain elusive. This study identifies a novel mechanism of epigenetic regulation centered on B-cell lymphoma-2-associated transcription factor 1 (Bclaf1) in nitrosamine-induced (Methylnitronitrosoguanidine, MNNG) esophageal carcinogenesis. In nitrosamine-induced malignant transformation cells (MNNG-M), Bclaf1 expression is progressively increased with malignancy, and elevated Bclaf1 levels are correlated with poor prognosis in ESCA patients. Functionally, Bclaf1 significantly promotes the abnormal proliferation of MNNG-M and ESCA cells in vitro and in vivo. Mechanistically, transposase-accessible chromatin sequencing (ATAC-seq) results suggest that Bclaf1 silencing markedly reduces chromatin accessibility, thereby impairing the synthesis of newly transcribed RNA. Bclaf1 activates RNA polymerase II subunit POLR2A to promote chromatin accessibility through distinct transcriptional and splicing mechanisms. More specifically, cleavage under targets and tagmentation (CUT&Tag) assays revealed Bclaf1/P300/H3K27ac co-recruitment at the POLR2A promoter, driving transcription via the E2/E3 elements of the POLR2A super-enhancer. Additionally, RNA-binding protein immunoprecipitation (RIP) assays demonstrated that the Bclaf1 cofactor, small nuclear ribonucleoprotein polypeptide A (SNRPA), interacts with pre-POLR2A to regulate its splicing. Collectively, our study reveals that Bclaf1 facilitates nitrosamine-induced ESCA by controlling POLR2A transcriptional and splicing activities, providing novel insight for early detection and intervention.
Collapse
Affiliation(s)
- Xiangjun Zhou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Xueqiong Zhou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China.
| | - Jun Li
- Department of thoracic surgery, The third affiliated hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Yingzheng He
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Shizhen Qiu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Ye Xu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Zeyu Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Yina Yao
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Jia Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Ying Wen
- Guangzhou Women and Children's Medical Centre, Guangzhou Medical University Institute of Pediatrics, 9 Jinsui Road, Guangzhou 510623, China
| | - Sitong Xie
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Jialong Chen
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Linhua Liu
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Zejin Ou
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | - Chunqing Cai
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Junyuan Lin
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Bingxi Lei
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Fei Zou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China.
| |
Collapse
|
38
|
Al Masri C, Vilseck JZ, Yu J, Hayes RL. Multisite λ-Dynamics for Protein-DNA Binding Affinity Prediction. J Chem Theory Comput 2025; 21:3536-3544. [PMID: 40123340 PMCID: PMC11983716 DOI: 10.1021/acs.jctc.4c01408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Transcription factors (TFs) regulate gene expression by binding to specific DNA sequences, playing critical roles in cellular processes and disease pathways. Computational methods, particularly λ-Dynamics, offer a promising approach for predicting TF relative binding affinities. This study evaluates the effectiveness of different λ-Dynamics perturbation schemes in determining binding free energy changes (ΔΔGb) of the WRKY transcription factor upon mutating its W-box binding site (GGTCAA) to a nonspecific sequence (GATAAA). Among the schemes tested, the single λ per base pair protocol demonstrated the fastest convergence and highest precision. Extending this protocol to additional mutants (GGTCCG and GGACAA) yielded ΔΔGb values that successfully ranked binding affinities, showcasing its strong potential for high-throughput screening of DNA binding sites.
Collapse
Affiliation(s)
- Carmen Al Masri
- Department
of Physics and Astronomy, Uninversity of
California, Irvine, California 92697, United States
| | - Jonah Z. Vilseck
- Department
of Biochemistry and Molecular Biology, Center for Computational Biology
and Bioinformatics, Indiana University School
of Medicine, Indianapolis, Indiana 46202, United States
| | - Jin Yu
- Department
of Physics and Astronomy, Department of Chemistry, University of California, Irvine, California 92697, United States
| | - Ryan L. Hayes
- Department
of Chemical and Biomolecular Engineering, Department of Pharmaceutical
Sciences, University of California, Irvine, California 92697, United States
| |
Collapse
|
39
|
Gabaev I, Rowland A, Jovanovic E, Gawden-Bone CM, Crozier TWM, Teixeira-Silva A, Greenwood EJD, Gerber PP, Wit N, Nathan JA, Matheson NJ, Lehner PJ. CRISPR-Cas9 genetic screens reveal regulation of TMPRSS2 by the Elongin BC-VHL complex. Sci Rep 2025; 15:11907. [PMID: 40195420 PMCID: PMC11976923 DOI: 10.1038/s41598-025-95644-0] [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: 07/16/2024] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
The TMPRSS2 cell surface protease is used by a broad range of respiratory viruses to facilitate entry into target cells. Together with ACE2, TMPRSS2 represents a key factor for SARS-CoV-2 infection, as TMPRSS2 mediates cleavage of viral spike protein, enabling direct fusion of the viral envelope with the host cell membrane. Since the start of the COVID-19 pandemic, TMPRSS2 has gained attention as a therapeutic target for protease inhibitors which would inhibit SARS-CoV-2 infection, but little is known about TMPRSS2 regulation, particularly in cell types physiologically relevant for SARS-CoV-2 infection. Here, we performed an unbiased genome-wide CRISPR-Cas9 library screen, together with a library targeted at epigenetic modifiers and transcriptional regulators, to identify cellular factors that modulate cell surface expression of TMPRSS2 in human colon epithelial cells. We find that endogenous TMPRSS2 is regulated by the Elongin BC-VHL complex and HIF transcription factors. Depletion of Elongin B or treatment of cells with PHD inhibitors resulted in downregulation of TMPRSS2 and inhibition of SARS-CoV-2 infection. We show that TMPRSS2 is still utilised by SARS-CoV-2 Omicron variants for entry into colonic epithelial cells. Our study enhances our understanding of the regulation of endogenous surface TMPRSS2 in cells physiologically relevant to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ildar Gabaev
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Alexandra Rowland
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Emilija Jovanovic
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Christian M Gawden-Bone
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Thomas W M Crozier
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Ana Teixeira-Silva
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Edward J D Greenwood
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Pehuén Pereyra Gerber
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Niek Wit
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - James A Nathan
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Nicholas J Matheson
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Paul J Lehner
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK.
| |
Collapse
|
40
|
Wu Z, Li Y, Dong J, Qin JJ. An updated review on the role of small molecules in mediating protein degradation. Eur J Med Chem 2025; 287:117370. [PMID: 39933402 DOI: 10.1016/j.ejmech.2025.117370] [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: 11/18/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Targeted protein degradation (TPD) technologies, inspired by physiological processes, have recently provided new directions for drug development. Unlike conventional drug development focusing on targeting the active sites of disease-related proteins, TPD can utilize any nook or cranny of a protein to drive degradation through the cell's inherent destruction mechanism. It offers various advantages such as stronger pharmacological effects, an expanded range of drug targets, and higher selectivity. Based on the ubiquitin-proteasome system and the lysosomal degradation pathway, a variety of TPD strategies have been developed including PROTAC, PROTAB, and AUTOTAC. These TPD strategies have continuously enriched the toolbox for targeted protein degradation and expanded the scope of application, providing new ideas for biological research and drug discovery. This review attempts to introduce up-to-date research progress in the TPD strategies, focusing mainly on their design concepts, advantages, potential applications, and challenges, which may provide some inspiration for drug design, drug discovery, and clinical application for biologists and chemists.
Collapse
Affiliation(s)
- Zumei Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yulong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jinyun Dong
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| | - Jiang-Jiang Qin
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| |
Collapse
|
41
|
Tang R, Sun T, Xing Z, Fan X, Jiang P, Le B, Jia K, Cai Y, Bi X, Zhang D, Lin R, He X. Glucocorticoid Receptor Inhibits the Progression of Schistosomiasis Hepatic Fibrosis Through Inducing Circadian Clock Gene Per1. J Infect Dis 2025:jiaf104. [PMID: 40183568 DOI: 10.1093/infdis/jiaf104] [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: 10/18/2024] [Indexed: 04/05/2025] Open
Abstract
Hepatic fibrosis is the leading cause of morbidity and mortality in schistosomiasis, and transcription factors (TF) may become potential therapeutic targets for this disease. Here, we found that a TF, NR3C1, was significantly downregulated in hepatic stellate cells (HSC), the effector cell of hepatic fibrosis, from mice infected with Schistosoma japonicum using RNA sequencing. Activation of NR3C1 using dexamethasone blocked HSC activation and hepatic fibrosis progression, while these effects were completely abolished upon specific deletion of NR3C1 in HSCs. Genome-wide binding site and transcriptome analyses suggested that Per1, a circadian clock gene, was under the direct control of NR3C1 through binding the glucocorticoid response elements, and it was responsible for the inhibitory effect of NR3C1 on HSC activation. Therefore, NR3C1 is a key TF in the activation of HSCs and a potential therapeutic target for hepatic schistosomiasis.
Collapse
Affiliation(s)
- Rui Tang
- Department of Tropical Infectious Diseases, Naval Medical University, Shanghai, China
| | - Tao Sun
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zhou Xing
- Department of Tropical Infectious Diseases, Naval Medical University, Shanghai, China
| | - XiaoBin Fan
- Department of Tropical Infectious Diseases, Naval Medical University, Shanghai, China
| | - PengYue Jiang
- Department of Tropical Infectious Diseases, Naval Medical University, Shanghai, China
| | - Bin Le
- Department of Tropical Infectious Diseases, Naval Medical University, Shanghai, China
| | - KaiWei Jia
- Department of Tropical Infectious Diseases, Naval Medical University, Shanghai, China
| | - YiLi Cai
- Department of Gastroenterology, Naval Medical Center, Naval Medical University, Shanghai, China
| | - XiaoJuan Bi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - DongMei Zhang
- Department of Tropical Infectious Diseases, Naval Medical University, Shanghai, China
| | - RenYong Lin
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xing He
- Department of Tropical Infectious Diseases, Naval Medical University, Shanghai, China
| |
Collapse
|
42
|
Skribbe M, Soneson C, Stadler MB, Schwaiger M, Suma Sreechakram VN, Iesmantavicius V, Hess D, Moreno EPF, Braun S, Seebacher J, Smallwood SA, Bühler M. A comprehensive Schizosaccharomyces pombe atlas of physical transcription factor interactions with proteins and chromatin. Mol Cell 2025; 85:1426-1444.e8. [PMID: 40015273 DOI: 10.1016/j.molcel.2025.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/16/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025]
Abstract
Transcription factors (TFs) are key regulators of gene expression, yet many of their targets and modes of action remain unknown. In Schizosaccharomyces pombe, one-third of TFs are solely homology predicted, with few experimentally validated. We created a comprehensive library of 89 endogenously tagged S. pombe TFs, mapping their protein and chromatin interactions using immunoprecipitation-mass spectrometry and chromatin immunoprecipitation sequencing. Our study identified protein interactors for half the TFs, with over a quarter potentially forming stable complexes. We discovered DNA-binding sites for most TFs across 2,027 unique genomic regions, revealing motifs for 38 TFs and uncovering a complex network of extensive TF cross- and autoregulation. Characterization of the largest TF family revealed conserved DNA sequence preferences but diverse binding patterns and identified a repressive heterodimer, Ntu1/Ntu2, linked to perinuclear gene localization. Our TFexplorer webtool makes all data interactively accessible, offering insights into TF interactions and regulatory mechanisms with broad biological relevance.
Collapse
Affiliation(s)
- Merle Skribbe
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, Basel, Switzerland; University of Basel, Petersplatz 10, Basel, Switzerland.
| | - Charlotte Soneson
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, Basel, Switzerland; SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Michael B Stadler
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, Basel, Switzerland; University of Basel, Petersplatz 10, Basel, Switzerland; SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Michaela Schwaiger
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, Basel, Switzerland; SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | | | | | - Daniel Hess
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, Basel, Switzerland
| | | | - Sigurd Braun
- Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jan Seebacher
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, Basel, Switzerland
| | - Sebastien A Smallwood
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, Basel, Switzerland
| | - Marc Bühler
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, Basel, Switzerland; University of Basel, Petersplatz 10, Basel, Switzerland.
| |
Collapse
|
43
|
Lambourne L, Mattioli K, Santoso C, Sheynkman G, Inukai S, Kaundal B, Berenson A, Spirohn-Fitzgerald K, Bhattacharjee A, Rothman E, Shrestha S, Laval F, Carroll BS, Plassmeyer SP, Emenecker RJ, Yang Z, Bisht D, Sewell JA, Li G, Prasad A, Phanor S, Lane R, Moyer DC, Hunt T, Balcha D, Gebbia M, Twizere JC, Hao T, Holehouse AS, Frankish A, Riback JA, Salomonis N, Calderwood MA, Hill DE, Sahni N, Vidal M, Bulyk ML, Fuxman Bass JI. Widespread variation in molecular interactions and regulatory properties among transcription factor isoforms. Mol Cell 2025; 85:1445-1466.e13. [PMID: 40147441 DOI: 10.1016/j.molcel.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 12/06/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
Most human transcription factor (TF) genes encode multiple protein isoforms differing in DNA-binding domains, effector domains, or other protein regions. The global extent to which this results in functional differences between isoforms remains unknown. Here, we systematically compared 693 isoforms of 246 TF genes, assessing DNA binding, protein binding, transcriptional activation, subcellular localization, and condensate formation. Relative to reference isoforms, two-thirds of alternative TF isoforms exhibit differences in one or more molecular activities, which often could not be predicted from sequence. We observed two primary categories of alternative TF isoforms: "rewirers" and "negative regulators," both of which were associated with differentiation and cancer. Our results support a model wherein the relative expression levels of, and interactions involving, TF isoforms add an understudied layer of complexity to gene regulatory networks, demonstrating the importance of isoform-aware characterization of TF functions and providing a rich resource for further studies.
Collapse
Affiliation(s)
- Luke Lambourne
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kaia Mattioli
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Clarissa Santoso
- Department of Biology, Boston University, Boston, MA 02215, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Gloria Sheynkman
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sachi Inukai
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Babita Kaundal
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anna Berenson
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA 02215, USA
| | - Kerstin Spirohn-Fitzgerald
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Anukana Bhattacharjee
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Elisabeth Rothman
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | - Florent Laval
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; TERRA Teaching and Research Centre, University of Liège, Gembloux 5030, Belgium; Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Brent S Carroll
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stephen P Plassmeyer
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA; Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Ryan J Emenecker
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA; Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Zhipeng Yang
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Deepa Bisht
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jared A Sewell
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Guangyuan Li
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Anisa Prasad
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard College, Cambridge, MA 02138, USA
| | - Sabrina Phanor
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ryan Lane
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Devlin C Moyer
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Toby Hunt
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CD10 1SD, UK
| | - Dawit Balcha
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Marinella Gebbia
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada; Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Jean-Claude Twizere
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; TERRA Teaching and Research Centre, University of Liège, Gembloux 5030, Belgium; Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Tong Hao
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA; Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Adam Frankish
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CD10 1SD, UK
| | - Josh A Riback
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael A Calderwood
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - David E Hill
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Martha L Bulyk
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Juan I Fuxman Bass
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biology, Boston University, Boston, MA 02215, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA; Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
44
|
Danaeifar M, Najafi A. Artificial Intelligence and Computational Biology in Gene Therapy: A Review. Biochem Genet 2025; 63:960-983. [PMID: 38635012 DOI: 10.1007/s10528-024-10799-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
One of the trending fields in almost all areas of science and technology is artificial intelligence. Computational biology and artificial intelligence can help gene therapy in many steps including: gene identification, gene editing, vector design, development of new macromolecules and modeling of gene delivery. There are various tools used by computational biology and artificial intelligence in this field, such as genomics, transcriptomic and proteomics data analysis, machine learning algorithms and molecular interaction studies. These tools can introduce new gene targets, novel vectors, optimized experiment conditions, predict the outcomes and suggest the best solutions to avoid undesired immune responses following gene therapy treatment.
Collapse
Affiliation(s)
- Mohsen Danaeifar
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Science, P.O. Box 19395-5487, Tehran, Iran
| | - Ali Najafi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Science, P.O. Box 19395-5487, Tehran, Iran.
| |
Collapse
|
45
|
Tremblay F, Xiong Q, Shah SS, Ko CW, Kelly K, Morrison MS, Giancarlo C, Ramirez RN, Hildebrand EM, Voytek SB, El Sebae GK, Wright SH, Lofgren L, Clarkson S, Waters C, Linder SJ, Liu S, Eom T, Parikh S, Weber Y, Martinez S, Malyala P, Abubucker S, Friedland AE, Maeder ML, Lombardo A, Myer VE, Jaffe AB. A potent epigenetic editor targeting human PCSK9 for durable reduction of low-density lipoprotein cholesterol levels. Nat Med 2025; 31:1329-1338. [PMID: 39930141 PMCID: PMC12003160 DOI: 10.1038/s41591-025-03508-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/13/2025] [Indexed: 02/27/2025]
Abstract
Epigenetic editing holds the promise of durable therapeutic effects by silencing disease-causing genes without changing the underlying DNA sequence. In this study, we designed an epigenetic editor to target human PCSK9 and thereby induce DNA methylation at this locus. A single administration of lipid nanoparticles encapsulating mRNA encoding this epigenetic editor was sufficient to drive near-complete silencing of human PCSK9 in transgenic mice. Silencing was durable for at least 1 year and was fully maintained after partial hepatectomy-induced liver regeneration. In addition, we showed reversibility of epigenetic editing in mice with previously silenced PCSK9 upon treatment with a targeted epigenetic activator designed to demethylate the PCSK9 locus. Notably, in cynomolgus monkeys, a single administration of the epigenetic editor potently and durably decreased circulating PCSK9 protein levels by approximately 90% with concomitant reduction in low-density lipoprotein cholesterol levels by approximately 70%. These findings demonstrate the therapeutic potential of durable and reversible epigenetic editing in vivo and support the development of epigenetic editor-based treatment for hypercholesterolemia.
Collapse
Affiliation(s)
| | - Qiang Xiong
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Shrijal S Shah
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Chih-Wei Ko
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | | | | | | | | | - Sarah B Voytek
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | - Shane H Wright
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | | | | | | | - Songlei Liu
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Taesun Eom
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Shefal Parikh
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Yuki Weber
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | - Padma Malyala
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | | | | | - Angelo Lombardo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | | | - Aron B Jaffe
- Chroma Medicine, Boston, MA, USA
- Curie.Bio, Cambridge, MA, USA
| |
Collapse
|
46
|
Zappia L, Richter S, Ramírez-Suástegui C, Kfuri-Rubens R, Vornholz L, Wang W, Dietrich O, Frishberg A, Luecken MD, Theis FJ. Feature selection methods affect the performance of scRNA-seq data integration and querying. Nat Methods 2025; 22:834-844. [PMID: 40082610 PMCID: PMC11978513 DOI: 10.1038/s41592-025-02624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 02/08/2025] [Indexed: 03/16/2025]
Abstract
The availability of single-cell transcriptomics has allowed the construction of reference cell atlases, but their usefulness depends on the quality of dataset integration and the ability to map new samples. Previous benchmarks have compared integration methods and suggest that feature selection improves performance but have not explored how best to select features. Here, we benchmark feature selection methods for single-cell RNA sequencing integration using metrics beyond batch correction and preservation of biological variation to assess query mapping, label transfer and the detection of unseen populations. We reinforce common practice by showing that highly variable feature selection is effective for producing high-quality integrations and provide further guidance on the effect of the number of features selected, batch-aware feature selection, lineage-specific feature selection and integration and the interaction between feature selection and integration models. These results are informative for analysts working on large-scale tissue atlases, using atlases or integrating their own data to tackle specific biological questions.
Collapse
Affiliation(s)
- Luke Zappia
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- School of Computing, Information and Technology, Technical University of Munich, Munich, Germany
| | - Sabrina Richter
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Ciro Ramírez-Suástegui
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Raphael Kfuri-Rubens
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
- Klinikum rechts der Isar, IIIrd Medical Department, Munich, Germany
| | - Larsen Vornholz
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Weixu Wang
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Oliver Dietrich
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Amit Frishberg
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Malte D Luecken
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Institute of Lung Health & Immunity, Helmholtz Munich; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany.
- School of Computing, Information and Technology, Technical University of Munich, Munich, Germany.
- School of Life Sciences Weihenstephan, Technical University of Munich, Friesing, Germany.
| |
Collapse
|
47
|
Chen S, Dedkova LM, Hecht SM. Biological Regulation Studied in Vitro and in Cellulo with Modified Proteins. Acc Chem Res 2025; 58:1109-1119. [PMID: 40072328 PMCID: PMC11964198 DOI: 10.1021/acs.accounts.5c00023] [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] [Indexed: 03/21/2025]
Abstract
ConspectusProteins and peptides occur ubiquitously in organisms and play key functional roles, as structural elements and catalysts. Their major natural source is ribosomal synthesis, which produces polypeptides from 20 amino acid building blocks. Peptides containing noncanonical amino acids have long been prepared by chemical synthesis, which has provided a wealth of physiologically active compounds. Comparatively, preparing modified proteins has been more challenging. Site-directed mutagenesis provided an important advance but was initially limited to canonical amino acids. New techniques for tRNA activation with noncanonical amino acids subsequently increased the scope of site-directed mutagenesis.Our report in 2012 demonstrated that modification of bacterial ribosomes at key positions enabled the selection of ribosomes capable of introducing β-amino acids into proteins in vitro. The generality of the selection procedure was tested further. Ribosomes capable of incorporating dipeptides, conformationally constrained dipeptides, dipeptidometics with embedded fluorophores, contiguous nucleobase amino acids, and phosphorylated amino acids were successfully identified.In this Account, we focus on the application of the new technology to dramatically alter protein structure in ways that enable new strategies for understanding and altering protein function. To illustrate the robustness of the technology we have provided examples studied in vitro and in cellulo. The first category involves the introduction of nucleobase amino acids into proteins in support of specific interactions with RNA and DNA. The energetic differences between potential protein-nucleic acid complexes formed from two binding partners are often quite small. It seems logical to think that selective binding can be achieved by using a nucleobase moiety in each of the binding partners by utilizing known interactions between nucleic acid bases (located in the protein and nucleic acid) to achieve energetically favorable interactions. We do so both in vitro and in cellulo. A second focus has involved the design of small fluorescent probes not much larger than amino acids that are genetically encodable and which can be incorporated during protein biosynthesis, serving as detectable probes of protein trafficking and interaction with other macromolecules. We provide an in vitro example of strongly fluorescent tryptophan analogues positioned at single sites within dihydrofolate reductase, permitting selective communication with a FRET acceptor at a known position, even in the presence of several tryptophans. An oxazole amino acid, weakly fluorescent in aqueous solution, fluoresced more strongly following incorporation into MreB, a scaffold protein produced in cellulo. Finally, we describe the introduction of a single phosphorylated tyrosine into the p50 subunit of NF-κB. When present at either of two key positions, the resulting NF-κB significantly enhanced binding in vitro to the promoter DNA as well as subsequent mRNA transcription of its client protein CD40 in cellulo. In a separate expression in activated Jurkat cells, an increased production of CD40 protein was observed.
Collapse
Affiliation(s)
- Shengxi Chen
- Center for BioEnergetics, Biodesign Institute and School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Larisa M. Dedkova
- Center for BioEnergetics, Biodesign Institute and School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Sidney M. Hecht
- Center for BioEnergetics, Biodesign Institute and School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
48
|
Darieva Z, Zarrineh P, Phillips N, Mallen J, Garcia Mora A, Donaldson I, Bridoux L, Douglas M, Dias Henriques SF, Schulte D, Birket MJ, Bobola N. Ubiquitous MEIS transcription factors actuate lineage-specific transcription to establish cell fate. EMBO J 2025; 44:2232-2262. [PMID: 40021842 PMCID: PMC12000411 DOI: 10.1038/s44318-025-00385-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 03/03/2025] Open
Abstract
Control of gene expression is commonly mediated by distinct combinations of transcription factors (TFs). This cooperative action allows the integration of multiple biological signals at regulatory elements, resulting in highly specific gene expression patterns. It is unclear whether combinatorial binding is also necessary to bring together TFs with distinct biochemical functions, which collaborate to effectively recruit and activate RNA polymerase II. Using a cardiac differentiation model, we find that the largely ubiquitous homeodomain proteins MEIS act as actuators, fully activating transcriptional programs selected by lineage-restricted TFs. Combinatorial binding of MEIS with lineage-enriched TFs, GATA, and HOX, provides selectivity, guiding MEIS to function at cardiac-specific enhancers. In turn, MEIS TFs promote the accumulation of the methyltransferase KMT2D to initiate lineage-specific enhancer commissioning. MEIS combinatorial binding dynamics, dictated by the changing dosage of its partners, drive cells into progressive stages of differentiation. Our results uncover tissue-specific transcriptional activation as the result of ubiquitous actuator TFs harnessing general transcriptional activator at tissue-specific enhancers, to which they are directed by binding with lineage- and domain-specific TFs.
Collapse
Affiliation(s)
- Zoulfia Darieva
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Peyman Zarrineh
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Naomi Phillips
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Joshua Mallen
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Araceli Garcia Mora
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ian Donaldson
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Laure Bridoux
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Megan Douglas
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Dorothea Schulte
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), Frankfurt am Main, Germany
| | - Matthew J Birket
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Nicoletta Bobola
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
49
|
Wang R, Zhang F, Li J, Yang D, Zhao H, Yuan J, Jia Y, Yu W, Guo W, Zou L, Zou K. GATA2 promotes cervical cancer progression under the transcriptional activation of TRIP4. Cell Signal 2025; 132:111778. [PMID: 40180167 DOI: 10.1016/j.cellsig.2025.111778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/09/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
The continued rise in recurrence and mortality rates of cervical cancer suggests the need to find novel therapeutic targets. Previous studies suggest that TRIP4 acts as a transcription factor to regulate cervical carcinogenesis and progression. Our aim was to explore whether the key downstream genes of TRIP4 functions same as TRIP4 in promoting cervical cancer development. We analyzed and confirmed the downstream targets of TRIP4 by RNA sequencing in cervical cancer cells with TRIP4 knockdown. The expression correlation between TRIP4 and GATA2 and the effect of GATA2 on cervical cancer cell growth were determined respectively by Western Blot, Scratch, Spheroid, and MTT analyses. Pulldown and ChIP experiments were performed to analyze the binding of TRIP4 to the promoter of GATA2. The clinical significance of GATA2 and TRIP4 expression in cervical cancer patients was analyzed by tissue microarray staining. GATA2 was highly expressed in cervical cancer tissues. Knockdown of GATA2 inhibited the growth, metastasis and stemness of cervical cancer cells and sensitized cervical cancer cells to radiation therapy. The inhibitory effect of TRIP4 knockdown on cervical cancer cells was rescued by GATA2 overexpression. Furthermore, TRIP4 could bind to the specific GATA2 promoter region, thereby activating its transcription. Clinical tissue microarray analysis indicated that the expression of TRIP4 and GATA2 was positively correlated, and high expression of both predicted a poor prognosis in cervical cancer patients. Our study demonstrated that GATA2 functions as the key downstream target of TRIP4 to promote cervical cancer progression and effective intervention of TRIP4/GATA2 signaling is expected to be developed as potential cervical cancer therapeutic strategy.
Collapse
Affiliation(s)
- Ruonan Wang
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Feng Zhang
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiazhi Li
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dian Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Hongmei Zhao
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jie Yuan
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuhan Jia
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Wei Guo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Lijuan Zou
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Kun Zou
- The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
50
|
Morin A, Chu CP, Pavlidis P. Identifying reproducible transcription regulator coexpression patterns with single cell transcriptomics. PLoS Comput Biol 2025; 21:e1012962. [PMID: 40257984 PMCID: PMC12011263 DOI: 10.1371/journal.pcbi.1012962] [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: 11/06/2024] [Accepted: 03/13/2025] [Indexed: 04/23/2025] Open
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, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ching Pan Chu
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Paul Pavlidis
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|