1
|
Xiang X, Mao J, Tang D, Huang H, Tang H. The ZBTB family in cardiac development and diseases. Biochem Biophys Res Commun 2025; 771:152026. [PMID: 40398093 DOI: 10.1016/j.bbrc.2025.152026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/17/2025] [Accepted: 05/14/2025] [Indexed: 05/23/2025]
Abstract
ZBTB (zinc finger and BTB domain) proteins are a class of evolutionarily conserved transcriptional factors (TFs) with zinc finger (ZF) and BTB (Broad-complex, Tram-track, and Bric-à-brac) domains. The ZBTB protein family has a wide range of functions in numerous biological processes, including cell cycle regulation, DNA repair, organ development, and haematopoietic stem cell fate determination. The ZBTB proteins regulate gene expression through interactions with transcriptional regulators, influencing processes such as myocardial contractility, inflammation, fibrosis, and cellular metabolism. Given the critical role of the ZBTB family in cardiac biology, the present review endeavours to comprehensively summarize the regulatory roles of seven ZBTB family members (HIC2, BCL6, PLZF, ZBTB17, ZBTB20, ZBTB7a, and ZBTB11) in cardiac development and diseases, along with their potential molecular mechanisms. Elucidating the molecular mechanisms of ZBTB proteins opens avenues for developing targeted therapies for cardiovascular diseases, including hypertrophy, fibrosis, and inflammation. This review provides a comprehensive summary of recent research on the role of ZBTB proteins in regulating cardiac transcription. Particular emphasis is placed on elucidating their functions in both cardiac development and the pathogenesis of cardiac diseases.
Collapse
Affiliation(s)
- Xing Xiang
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, China; Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, China; Institute of Cardiovascular Disease, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jie Mao
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, China; Institute of Cardiovascular Disease, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China; School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan, 421001, China
| | - Dan Tang
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, China; Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, China; Institute of Cardiovascular Disease, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hong Huang
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, China; Institute of Cardiovascular Disease, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| | - Huifang Tang
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, China; Institute of Cardiovascular Disease, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
2
|
Nussinov R, Yavuz BR, Jang H. Tumors and their microenvironments: Learning from pediatric brain pathologies. Biochim Biophys Acta Rev Cancer 2025; 1880:189328. [PMID: 40254040 PMCID: PMC12124968 DOI: 10.1016/j.bbcan.2025.189328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Early clues to tumors and their microenvironments come from embryonic development. Here we review the literature and consider whether the embryonic brain and its pathologies can serve as a better model. Among embryonic organs, the brain is the most heterogenous and complex, with multiple lineages leading to wide spectrum of cell states and types. Its dysregulation promotes neurodevelopmental brain pathologies and pediatric tumors. Embryonic brain pathologies point to the crucial importance of spatial heterogeneity over time, akin to the tumor microenvironment. Tumors dedifferentiate through genetic mutations and epigenetic modulations; embryonic brains differentiate through epigenetic modulations. Our innovative review proposes learning developmental brain pathologies to target tumor evolution-and vice versa. We describe ways through which tumor pharmacology can learn from embryonic brains and their pathologies, and how learning tumor, and its microenvironment, can benefit targeting neurodevelopmental pathologies. Examples include pediatric low-grade versus high-grade brain tumors as in rhabdomyosarcomas and gliomas.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
3
|
Liu D, Jiang Y, Ma B, Li L. Structure-based artificial intelligence-aided design of MYC-targeting degradation drugs for cancer therapy. Biochem Biophys Res Commun 2025; 766:151870. [PMID: 40288261 DOI: 10.1016/j.bbrc.2025.151870] [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: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
The MYC protein is an oncoprotein that plays a crucial role in various cancers. Although its significance has been well recognized in research, the development of drugs targeting MYC remains relatively slow. In this study, we developed a novel MYC peptide inhibitor based on the MYC/MAX dimer structure, integrating artificial intelligence-assisted peptide drug design. Additionally, we introduced a chaperone-mediated autophagy signal to construct a MYC-targeted degradation drug, MYC-LYSO. By incorporating nano-selenium delivery, we further formulated an enhanced MYC degradation agent, Se-MYC-LYSO. Se-MYC-LYSO demonstrated potent efficacy in inducing MYC degradation, inhibiting tumor cell proliferation, and promoting apoptosis. Moreover, our findings indicate that the efficacy of Se-MYC-LYSO is dependent on the autophagy pathway. These results provide a novel strategy for targeting MYC in cancer therapy.
Collapse
Affiliation(s)
- Donghua Liu
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yize Jiang
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Bohan Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Lei Li
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
4
|
Gao J, Pan H, Guo X, Huang Y, Luo JY. Endothelial Krüppel-like factor 2/4: Regulation and function in cardiovascular diseases. Cell Signal 2025; 130:111699. [PMID: 40023301 DOI: 10.1016/j.cellsig.2025.111699] [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: 12/02/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
This review presents an overview of the regulation, function, disease-relevance and pharmacological regulation of the critical endothelial transcription factors KLF2/4 in vasculature. The regulatory mechanisms of KLF2/4 expression and activity in vascular endothelium in response to hemodynamic forces and biochemical stimuli are depicted. The functional effects mediated by direct or indirect target genes of KLF2/4 in endothelial cells are systematically summarized. The contributory roles that dysregulated KLF2/4 play in relevant cardiovascular pathologies, such as atherosclerotic vascular lesions, pulmonary arterial hypertension and vascular complications of diabetes were reviewed. Moreover, this review also discusses the pharmacological regulation of KLF2/4 by drugs used in clinics and therapeutic possibility by directly targeting these two transcription factors for treating atherosclerotic cardiovascular diseases. Finally, prospective opinions on the gaps in disclosing novel vascular function mediated by KLF2/4 and future research needs are expressed.
Collapse
Affiliation(s)
- Jing Gao
- Department of Cardiology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Hongjie Pan
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Jiang-Yun Luo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Egan G, Tasian SK. Precision medicine for high-risk gene fusions in pediatric AML: a focus on KMT2A, NUP98, and GLIS2 rearrangements. Blood 2025; 145:2574-2586. [PMID: 39808803 DOI: 10.1182/blood.2024026598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
ABSTRACT Robust genetic characterization of pediatric acute myeloid leukemia (AML) has demonstrated that fusion oncogenes are highly prevalent drivers of AML leukemogenesis in young children. Identification of fusion oncogenes associated with adverse outcomes has facilitated risk stratification of patients, although successful development of precision medicine approaches for most fusion-driven AML subtypes have been historically challenging. This knowledge gap has been in part due to difficulties in targeting structural alterations involving transcription factors and in identification of a therapeutic window for selective inhibition of the oncofusion without deleterious effects upon essential wild-type proteins. Herein, we discuss the current molecular landscape and functional characterization of 3 of the most lethal childhood AML fusion-oncogene driven subtypes harboring KMT2A, NUP98, or CBFA2T3::GLIS2 rearrangements. We further review early-phase clinical trial data of novel targeted inhibitors and immunotherapies that have demonstrated initial success specifically for children with these poor-prognosis genetic subtypes of AML and provide appreciable optimism to improve clinical outcomes in the future.
Collapse
Affiliation(s)
- Grace Egan
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Princess Máxima Center for Paediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
6
|
Sun Q, Wang H, Xie J, Wang L, Mu J, Li J, Ren Y, Lai L. Computer-Aided Drug Discovery for Undruggable Targets. Chem Rev 2025. [PMID: 40423592 DOI: 10.1021/acs.chemrev.4c00969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Undruggable targets are those of therapeutical significance but challenging for conventional drug design approaches. Such targets often exhibit unique features, including highly dynamic structures, a lack of well-defined ligand-binding pockets, the presence of highly conserved active sites, and functional modulation by protein-protein interactions. Recent advances in computational simulations and artificial intelligence have revolutionized the drug design landscape, giving rise to innovative strategies for overcoming these obstacles. In this review, we highlight the latest progress in computational approaches for drug design against undruggable targets, present several successful case studies, and discuss remaining challenges and future directions. Special emphasis is placed on four primary target categories: intrinsically disordered proteins, protein allosteric regulation, protein-protein interactions, and protein degradation, along with discussion of emerging target types. We also examine how AI-driven methodologies have transformed the field, from applications in protein-ligand complex structure prediction and virtual screening to de novo ligand generation for undruggable targets. Integration of computational methods with experimental techniques is expected to bring further breakthroughs to overcome the hurdles of undruggable targets. As the field continues to evolve, these advancements hold great promise to expand the druggable space, offering new therapeutic opportunities for previously untreatable diseases.
Collapse
Affiliation(s)
- Qi Sun
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
| | - Hanping Wang
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Juan Xie
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Liying Wang
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Junxi Mu
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Junren Li
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yuhao Ren
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Luhua Lai
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
- Research Unit of Drug Design Method, Chinese Academy of Medical Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
7
|
Lee Y, Yoo S, Cho S, Kim I, Kim IS. Advances in transcription factor delivery: Target selection, engineering strategies, and delivery platforms. J Control Release 2025; 384:113885. [PMID: 40425091 DOI: 10.1016/j.jconrel.2025.113885] [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: 03/11/2025] [Revised: 05/03/2025] [Accepted: 05/22/2025] [Indexed: 05/29/2025]
Abstract
Recent advances in delivery systems for transcription factors (TFs) have opened new therapeutic opportunities in regenerative medicine, cancer therapy, and genetic disorders. However, effective TF delivery still faces substantial obstacles, including limited cellular uptake, inefficient nuclear translocation, low cargo stability, and insufficient target specificity. Furthermore, artificial TFs have enabled targeted modulation of gene expression, further expanding their therapeutic potential. This review comprehensively discusses current progress in TF delivery methodologies, including direct TF protein delivery using cell-penetrating peptides, and extracellular vesicles, as well as TF gene delivery approaches utilizing both lipid-based nanoparticles and viral strategies. Notably, engineered nanoparticles have emerged as promising platforms due to their precise control over TF delivery, improved specificity, and minimized off-target effects. Despite these significant advancements, major hurdles in delivery efficiency, cargo stability, and overall safety persist. Overcoming these obstacles will be essential to accelerate the clinical translation of TF-based therapeutics for a broad spectrum of diseases.
Collapse
Affiliation(s)
- Yeji Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea; Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute Science and Technology, Seoul 02792, South Korea
| | - Seongkyeong Yoo
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea
| | - Seongeon Cho
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea; Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute Science and Technology, Seoul 02792, South Korea
| | - Iljin Kim
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea; Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute Science and Technology, Seoul 02792, South Korea.
| |
Collapse
|
8
|
Ni CX, Xu JJ, Pang Y, Xu JJ. Treatment strategies targeting the phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin pathway against triple-negative breast cancer. World J Clin Oncol 2025; 16:104623. [DOI: 10.5306/wjco.v16.i5.104623] [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: 12/27/2024] [Revised: 02/25/2025] [Accepted: 03/25/2025] [Indexed: 05/19/2025] Open
Abstract
Triple negative breast cancer (TNBC) is an exceptionally aggressive subtype of breast cancer with a poor prognosis. TNBC patients have limited treatment options beyond conventional chemotherapy, and they face significant challenges associated with disease recurrence and resistance to chemotherapy. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) signaling pathway plays a pivotal role in cell proliferation, growth, metabolism, and survival. Its aberrant activation is closely linked to the development and progression of TNBC, as well as treatment response and drug resistance. Currently, numerous targeted drugs specifically inhibiting this signaling pathway are being developed and undergoing clinical trials. These include inhibitors targeting PI3K, AKT, or mTOR individually, as well as dual-target or multi-target inhibitors simultaneously targeting different components of this pathway. Encouragingly, some inhibitors have demonstrated promising potential in clinical trials. This review delves into the therapeutic potential of the PI3K/AKT/mTOR signaling pathway for TNBC and explores prospects for drug discovery.
Collapse
Affiliation(s)
- Chun-Xiao Ni
- Department of Minimally Invasive Oncology, Tai’an Central Hospital Affiliated to Qingdao University, Tai’an 271000, Shandong Province, China
| | - Jia-Ju Xu
- Department of Pediatrics, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai 264000, Shandong Province, China
| | - Yu Pang
- Department of Pathology, Tai’an Central Hospital Affiliated to Qingdao University, Tai’an 271000, Shandong Province, China
| | - Jia-Ju Xu
- Department of Medical Oncology, Tai’an Central Hospital Affiliated to Qingdao University, Tai’an 271000, Shandong Province, China
| |
Collapse
|
9
|
Dixit A, Paegel BM. Solid-phase DNA-encoded library synthesis: a master builder's instructions. Nat Protoc 2025:10.1038/s41596-025-01190-4. [PMID: 40404924 DOI: 10.1038/s41596-025-01190-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 04/01/2025] [Indexed: 05/24/2025]
Abstract
Solid-phase DNA-encoded library (DEL) synthesis is a next-generation drug discovery technology with powerful activity-based and cellular lead identification capabilities. Solid-phase DELs combine the one-bead-one-compound approach with DNA encoding to furnish beads that display multiple copies of photocleavable library members and DNA encoding tags. Sequential chemical synthesis and enzymatic DNA ligation reactions yield an encoded library in which individual library members are physically isolable, enabling various high-throughput screening modalities. This advancement from on-DNA synthesis, in which small molecules are directly attached to their DNA-encoding tags, decouples the library member from the steric bulk of the DNA tag, which prevents biased binding to a target. Here we provide step-by-step instructions for solid-phase DEL synthesis, incorporating all of our most recent quality control innovations to ensure robust library production. The protocol begins with on-bead synthesis of a linker containing a spectroscopic handle for chromatographic analysis, an ionization enhancer for mass spectrometry and an alkyne for installation of DNA encoding sites via copper-catalyzed azide-alkyne cycloaddition click chemistry. Coupling of a photocleavable linker before library synthesis enables compound liberation from the bead for activity-based screening. Powerful combinatorial split-and-pool parallel synthesis tactics transform modest collections of small-molecule building blocks into large DELs of all possible building block combinations. Post synthesis, decoding and mass analysis of single DEL beads as well as whole-library deep sequencing provides rigorous chemical and bioinformatic quality control and establishes suitability for screening. The solid-phase chemistry is highly accessible: expertise in chemical synthesis is not necessary and solid-phase synthesis apparatus is routinely available in molecular biology laboratories. This procedure requires ~1 month to complete.
Collapse
Affiliation(s)
- Anjali Dixit
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Brian M Paegel
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA.
- Department of Chemistry, University of California, Irvine, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
10
|
Liu BH, Liu M, Radhakrishnan S, Dai MY, Jaladanki CK, Gao C, Tang JP, Kumari K, Go ML, Vu KAL, Kwon J, Seo HS, Song K, Tian X, Feng L, Tan JL, Melkonian AV, Liu Z, Wulf G, Arthanari H, Qi J, Dhe-Paganon S, Clohessy JG, Choong YK, Sivaraman J, Fan H, Tenen DG, Chai L. Targeting transcription factors through an IMiD independent zinc finger domain. EMBO Mol Med 2025:10.1038/s44321-025-00241-3. [PMID: 40369156 DOI: 10.1038/s44321-025-00241-3] [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: 01/07/2024] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Immunomodulatory imide drugs (IMiDs) degrade specific C2H2 zinc finger degrons in transcription factors, making them effective against certain cancers. SALL4, a cancer driver, contains seven C2H2 zinc fingers in three clusters, including an IMiD degron in zinc finger cluster one (ZFC1). Surprisingly, IMiDs do not inhibit the growth of SALL4-expressing cancer cells. To overcome this limit, we focused on a non-IMiD domain, SALL4 zinc finger cluster four (ZFC4). By combining ZFC4-DNA crystal structure and an in silico docking algorithm, in conjunction with cell viability assays, we screened several chemical libraries against a potentially druggable binding pocket, leading to the discovery of SH6, a compound that selectively targets SALL4-expressing cancer cells. Mechanistic studies revealed that SH6 degrades SALL4 protein through the CUL4A/CRBN pathway, while deletion of ZFC4 abolished this activity. Moreover, SH6 treatment led to a significant 87% tumor growth inhibition of SALL4+ patient-derived xenografts and demonstrated good bioavailability in pharmacokinetic studies. In summary, these studies represent a new approach for IMiD independent drug discovery targeting C2H2 transcription factors such as SALL4 in cancer.
Collapse
Affiliation(s)
- Bee Hui Liu
- Cancer Science Institute of Singapore, Singapore, 117599, Singapore
| | - Miao Liu
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Meng-Yuan Dai
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chaitanya Kumar Jaladanki
- Bioinformatics Institute, Agency for Science, Technology and Research, Matrix, 138671, Singapore, Singapore
| | - Chong Gao
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jing Ping Tang
- Cancer Science Institute of Singapore, Singapore, 117599, Singapore
| | - Kalpana Kumari
- Cancer Science Institute of Singapore, Singapore, 117599, Singapore
| | - Mei Lin Go
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore, Singapore
| | - Kim Anh L Vu
- Cancer Science Institute of Singapore, Singapore, 117599, Singapore
| | - Junsu Kwon
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Kijun Song
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Xi Tian
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Li Feng
- Cancer Science Institute of Singapore, Singapore, 117599, Singapore
| | - Justin L Tan
- Cancer Science Institute of Singapore, Singapore, 117599, Singapore
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Arek V Melkonian
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, MA, USA
| | - Zhaoji Liu
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA
| | - Gerburg Wulf
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA
| | - Haribabu Arthanari
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - John G Clohessy
- Preclinical Murine Pharmacogenetics Facility and Mouse Hospital, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yeu Khai Choong
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - J Sivaraman
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Hao Fan
- Bioinformatics Institute, Agency for Science, Technology and Research, Matrix, 138671, Singapore, Singapore
- Cancer and Stem Cell Program, Duke-NUS Medical School, Singapore, Singapore
| | - Daniel G Tenen
- Cancer Science Institute of Singapore, Singapore, 117599, Singapore.
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Harvard Medical School, MA, USA.
| | - Li Chai
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Karatzas P, Brotzakis ZF, Sarimveis H. Small Molecules Targeting the Structural Dynamics of AR-V7 Partially Disordered Proteins Using Deep Ensemble Docking. J Chem Theory Comput 2025; 21:4898-4909. [PMID: 40231860 PMCID: PMC12080126 DOI: 10.1021/acs.jctc.5c00171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
The extensive conformational dynamics of partially disordered proteins hinders the efficiency of traditional in-silico structure-based drug discovery approaches due to the challenge of screening large chemical spaces of compounds, albeit with an excessive number of transient binding sites, quickly making this problem intractable. In this study, using the monomer of the AR-V7 transcription factor splicing variant related to prostate cancer as a test case, we present a deep ensemble docking pipeline that accelerates the screening of small molecule binders targeting partially disordered proteins at functional regions. By swiftly identifying the conformational ensemble of AR-V7 and reducing the dimension of binding sites by a factor of 90, we identify functionally relevant binding sites along the AR-V7 structural ensemble at phase separation-prone regions that have been experimentally shown to contribute to enhanced transcription activity and the onset of tumor growth. Following this, we combine physics-based molecular docking and multiobjective classification machine learning models to speed up the screening for binders in a larger chemical space able to target these functional multiple binding sites of AR-V7. This step increases the multibinding site hit rate of small molecules by a factor of 17 compared to naive molecular docking. Finally, assessing in atomistic molecular dynamics the effect of a selected binder on AR-V7 dynamics, we find that in the presence of the ChEMBL22003 compound, AR-V7 exhibits less conformational entropy, smaller solvent exposure of phase separation-prone regions, and higher solvent exposure of other protein regions, promoting this compound as a potential AR-V7 phase separation modulator.
Collapse
Affiliation(s)
- Pantelis Karatzas
- School
of Chemical Engineering, National Technical
University of Athens, 9 Heroon Polytechniou Street, Athens 15780, Greece
| | - Z. Faidon Brotzakis
- Institute
of Bioinnovation (IBI), Biomedical Science Research Center Alexander
Fleming, 34 Fleming Street, Vari 16672, Greece
- Centre
for Misfolding Diseases, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Haralambos Sarimveis
- School
of Chemical Engineering, National Technical
University of Athens, 9 Heroon Polytechniou Street, Athens 15780, Greece
| |
Collapse
|
12
|
Cui Z, Huang F, Fang K, Yan J, Zhang Y, Kang DD, Zhou Y, Zhao Y, Everitt JI, Hankey W, Armstrong AJ, Huang J, Wang H, Jin VX, Dong Y, Wang Q. SCORT-Cas13d Nanotherapy Precisely Targets the 'Undruggable' Transcription Factor HoxB13 in Metastatic Prostate Cancer In Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417605. [PMID: 40349174 DOI: 10.1002/advs.202417605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/01/2025] [Indexed: 05/14/2025]
Abstract
Metastatic cancer, the primary cause of cancer mortality, frequently exhibits heightened dependence on certain transcription factors (TFs), which serve as master regulators of oncogenic signaling yet are often untargetable by small molecules. Selective Cell in ORgan Targeting (SCORT) nanoparticles are developed for precise CRISPR/Cas13d mRNA and gRNA delivery to metastatic cancer cells in vivo, aiming to knock down the undruggable oncogenic TF HoxB13. In prostate cancer liver metastasis models driven by HoxB13, repeated systemic SCORT-Cas13d-gHoxB13 treatment significantly decreases HoxB13 expression, reduces metastasis, and extends mouse survival. Prolonged treatment shows no significant impact on major organ function, histology or immune markers. Mechanistically, SCORT-Cas13d-gHoxB13 treatment suppresses metastatic tumor proliferation and angiogenesis while promoting apoptosis by regulating multiple gene pathways. Unexpectedly, it inhibits the non-canonical, EMT-independent oncogenic function of Snail. These findings suggest that SCORT-Cas13d-gHoxB13 can effectively and safely target the undruggable HoxB13 in metastatic prostate cancer, positioning CRISPR/Cas13d as a potential treatment.
Collapse
Affiliation(s)
- Zhifen Cui
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Furong Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kun Fang
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Jingyue Yan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Yuebao Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Diana D Kang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Icahn Genomics Institute, Precision Immunology Institute, Department of Immunology and Immunotherapy, Department of Oncological Sciences, Tisch Cancer Institute, Biomedical Engineering and Imaging Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yufan Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Yue Zhao
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jeffrey I Everitt
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - William Hankey
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Andrew J Armstrong
- Department of Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute Center for Prostate and Urologic Cancer, Durham, NC, 27710, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Hongyan Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Victor X Jin
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yizhou Dong
- Icahn Genomics Institute, Precision Immunology Institute, Department of Immunology and Immunotherapy, Department of Oncological Sciences, Tisch Cancer Institute, Biomedical Engineering and Imaging Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Qianben Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute Center for Prostate and Urologic Cancer, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| |
Collapse
|
13
|
Li BY, Li HL, Zeng FE, Luan XY, Liu BQ, Wang ZZ, Zhang L, Dong XZ. Identification of PD-L1-related biomarkers for selecting gastric adenocarcinoma patients for PD-1/PD-L1 inhibitor therapy. Discov Oncol 2025; 16:689. [PMID: 40338384 PMCID: PMC12061829 DOI: 10.1007/s12672-025-02515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
PD-1/PD-L1 inhibitors have been used to treat gastric cancer, and PD-L1 expression has been identified as a biomarker for predicting the effectiveness of immunotherapy in the treatment of gastric cancer. However, PD-L1 expression prediction for immunotherapy response is inaccurate, and improved response biomarkers are required. Thus, it is important to identify additional biomarkers that can predict the responses to PD-1/PD-L1 monoclonal antibodies in gastric cancer. In this study, GO and KEGG enrichment analysis of 142 DEGs co-expressed with PD-L1 were performed, and 41 genes were identified based on the intersection of the mRNA-significant GO term network and the mRNA-significant signalling pathway network. Further intersection analysis of the 41 candidate genes and 137 positive immunotherapy response genes indicated that BATF2 significantly affects the overall survival of GC patients. The transcription factor prediction for BATF2 identified additional potential predictors and therapeutic targets for GC. STAT and IRF family members were predicted to be transcription factors for BATF2. In addition, BATF2 knockdown significantly promoted GC cell growth, and PD-L1 expression was upregulated in si-BATF2-treated MKN-45 cells. Thus, BATF2 may serve as a biomarker for predicting the efficacy of PD-L1 blockade therapy in GC. BATF2 acts as a tumour suppressor gene during the development of GC. BATF2 is closely related to PD-L1 expression in GC, and high BATF2 expression positively correlates with low PD-L1 expression. BATF2 can be used as a potential biomarker and therapeutic target for responding to anti-PD-1 and anti-PD-L1 immunotherapies in GC.
Collapse
Affiliation(s)
- Bo-Ya Li
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China
| | - Hui-Ling Li
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China
| | - Fei-Er Zeng
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Xuan-Yu Luan
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bi-Qing Liu
- Department of Pharmacy, Children's Hospital Affiliated to Capital Institute of Paediatrics, Beijing, China
| | - Zhi-Zhou Wang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China.
| | - Xian-Zhe Dong
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China.
| |
Collapse
|
14
|
Qi X, Zhou J, Wang P, Li Y, Li H, Miao Y, Ma X, Luo X, Zhang Z, He Y, Shen W, Zhao W, Cui R, Li C, Zhu H, Lyu J. KLF7-regulated ITGA2 as a therapeutic target for inhibiting oral cancer stem cells. Cell Death Dis 2025; 16:354. [PMID: 40316546 PMCID: PMC12048542 DOI: 10.1038/s41419-025-07689-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
Cancer stem cells (CSCs) play crucial roles in tumor metastasis, therapy resistance, and immune evasion. Identifying and understanding the factors that regulate the stemness of tumor cells presents promising opportunities for developing effective therapeutic strategies. In this study on oral squamous cell carcinoma (OSCC), we confirmed the key role of KLF7 in maintaining the stemness of OSCC. Using chromatin immunoprecipitation sequencing and dual-luciferase assays, we identified ITGA2, a membrane receptor, as a key downstream gene regulated by KLF7 in the maintenance of stemness. Tumor sphere formation assays, flow cytometry analyses, and in vivo limiting dilution tumorigenicity evaluations demonstrated that knocking down ITGA2 significantly impaired stemness. Upon binding to its extracellular matrix (ECM) ligand, type I collagen, ITGA2 activates stemness-associated signaling pathways, including PI3K-AKT, MAPK, and Hippo. TC-I 15, a small-molecule inhibitor of the ITGA2-collagen interaction, significantly sensitizes oral squamous cell carcinoma (OSCC) to cisplatin in xenograft models. In summary, we reveal that the KLF7/ITGA2 axis is a crucial modulator of stemness in OSCC. Our findings suggest that ITGA2 is a promising therapeutic target, offering a novel anti-CSC strategy.
Collapse
Affiliation(s)
- Xin Qi
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Jiang Zhou
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Pan Wang
- Department of Stomatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yunyan Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Haoran Li
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Yuwen Miao
- Zhejiang University, School of Medicine, Affiliated Stomatology Hospital, Hangzhou, Zhejiang, P. R. China
| | - XiaoQing Ma
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiayan Luo
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Zhiling Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Yanling He
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Wenyi Shen
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Wenquan Zhao
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China
| | - Rutao Cui
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China
| | - Cang Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Clinical Research Center for CANCER; Cancer Center of Zhejiang University, Hangzhou, China.
| | - Huiyong Zhu
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China.
| | - Jiong Lyu
- Zhejiang University, School of Medicine, First Affiliated Hospital, Hangzhou, Zhejiang, P. R. China.
| |
Collapse
|
15
|
Chen Z, Dhruv H, Zhang X, Rej RK, Bai L, McEachern D, Kirchhoff P, Nagilla R, Jolivette LJ, Rice CT, Orth P, Strickland CO, Priestley ES, Mohammad HP, Wang M, Wen B, Sun D, Sui Z, Wang S. Development of PVTX-405 as a potent and highly selective molecular glue degrader of IKZF2 for cancer immunotherapy. Nat Commun 2025; 16:4095. [PMID: 40312344 PMCID: PMC12046021 DOI: 10.1038/s41467-025-58431-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/24/2025] [Indexed: 05/03/2025] Open
Abstract
IKZF2 (Helios) is a transcription factor that is selectively expressed by Tregs and is essential for preserving the function and stability of Tregs in the tumor microenvironment (TME), where it suppresses the anti-tumor immune response. Targeted IKZF2 degradation by small molecules represents a promising strategy for the development of a new class of cancer immunotherapy. Herein, we describe the discovery of PVTX-405, a potent, effective, highly selective, and orally efficacious IKZF2 molecular glue degrader. PVTX-405 degrades IKZF2 (DC50 = 0.7 nM and Dmax = 91%) while sparing other CRBN neo-substrates. Degradation of IKZF2 by PVTX-405 increases production of inflammatory cytokine IL-2 and reduces the suppressive activity of Tregs, leading to an increase in Teff cell proliferation. Once-daily oral administration of PVTX-405 as single agent significantly delays the growth of MC38 tumors in a syngeneic tumor model using humanized CRBN mice. PVTX-405 in combination with anti-PD1 or anti-LAG3 significantly increases animal survival compared to anti-PD1 or anti-LAG3 alone. Together, these results demonstrate that PVTX-405 is a promising IKZF2 degrader for clinical development for the treatment of human cancers.
Collapse
Affiliation(s)
- Zhixiang Chen
- Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | | | | | - Rohan Kalyan Rej
- Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Longchuan Bai
- Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Donna McEachern
- Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Paul Kirchhoff
- Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Cory T Rice
- SK Life Sciences Labs, King of Prussia, PA, USA
| | - Peter Orth
- SK Life Sciences Labs, King of Prussia, PA, USA
| | | | | | | | - Meilin Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Zhihua Sui
- SK Life Sciences Labs, King of Prussia, PA, USA
| | - Shaomeng Wang
- Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
Chen L, Li T, Xu J, Liao S, Xu H, Li K, Ivanova D, Si Y, Zhang X, Zhang J, Li F, Chen X, Wang Y, Xie B, Shi D, Wang M. Screening of Oxidative Stress-related Biomarkers and Antioxidant Drugs for Polycystic Ovary Syndrome Based on Bioinformatics and Molecular Docking. Reprod Sci 2025; 32:1644-1660. [PMID: 39875693 DOI: 10.1007/s43032-025-01790-1] [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: 07/21/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder affecting women of reproductive age. Oxidative stress (OS) is suggested to play a significant role in the development of PCOS. Using antioxidants to reduce OS and maintain a healthy balance in the body could be a novel treatment approach for PCOS. This study analyzed transcriptome data from the Gene Expression Omnibus database, focusing on genes associated with OS. By implementing two machine learning algorithms, three OS-related biomarkers-HMOX1, MMP9, and KLF2-were successfully identified. To evaluate the diagnostic potential of these biomarkers, a Logistic regression model was employed. Additionally, granulosa cells were collected from healthy individuals and infertile women with PCOS, and the reliability of HMOX1, MMP9, and KLF2 was verified by quantitative real-time PCR experiments. Furthermore, small molecule drugs targeting proteins encoded by genes HMOX1 and MMP9 were predicted through the Drug Signature Database. Molecular docking of drugs to proteins identified two antioxidants, butein and demethoxycurcumin, as potential candidates for PCOS therapy.
Collapse
Affiliation(s)
- Lanhui Chen
- Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Tian Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jialiang Xu
- Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Siqi Liao
- Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Haocheng Xu
- Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Kun Li
- Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Deyana Ivanova
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuewen Si
- Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xin Zhang
- Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jianning Zhang
- Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Xuemei Chen
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Biao Xie
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Dan Shi
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, China.
- Nutrition Innovation Platform-Sichuan and Chongqing, School of Public Health, Chongqing Medical University, Chongqing, China.
| | - Meijiao Wang
- Department of Physiology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China.
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
17
|
Sun Y, Wei H, Yu W, Gao H, Li J, Li X, Zhang H, Zhang H, Miao S, Zhao L, Yang R, Xu J, Lu Y, Wei F, Zhou H, Gao D, Jin Y, Zhang L. The actin-binding protein drebrin disrupts NF2-LATS kinases complex assembly to facilitate liver tumorigenesis. Hepatology 2025; 81:1433-1451. [PMID: 39325963 DOI: 10.1097/hep.0000000000001063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/17/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND AIMS The Hippo signaling has emerged as a crucial regulator of tissue homeostasis, regeneration, and tumorigenesis, representing a promising therapeutic target. Neurofibromin 2 (NF2), a component of Hippo signaling, is directly linked to human cancers but has been overlooked as a target for cancer therapy. APPROACH AND RESULTS Through a high-content RNA interference genome-wide screen, the actin-binding protein Drebrin (DBN1) has been identified as a novel modulator of YAP localization. Further investigations have revealed that DBN1 directly interacts with NF2, disrupting the activation of large tumor suppressor kinases (LATS1/2) by competing with LATS kinases for NF2 binding. Consequently, DBN1 knockout considerably promotes YAP nuclear exclusion and repression of target gene expression, thereby preventing cell proliferation and liver tumorigenesis. We identified three lysine residues (K238, K248, and K252) essential for DBN1-NF2 interaction and developed a mutant DBN1 (DBN1-3K mut ) that is defective in NF2 binding and incompetent to trigger NF2-dependent YAP activation and tumorigenesis both in vitro and in vivo. Furthermore, BTP2, a DBN1 inhibitor, successfully restored NF2-LATS kinase binding and elicited potent antitumor activity. The combination of sorafenib and BTP2 exerted synergistic inhibitory effects against HCC. CONCLUSIONS Our study identifies a novel DBN1-NF2-LATS axis, and pharmacological inhibition of DBN1 represents a promising alternative intervention targeting the Hippo pathway in cancer treatment.
Collapse
Affiliation(s)
- Yang Sun
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Henan Wei
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wentao Yu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Haoran Gao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jinhui Li
- HuidaGene Therapeutics Co., Ltd., Shanghai, China
| | - Xiaoyu Li
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Haijiao Zhang
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Haoen Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Sen Miao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Lihua Zhao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Ruizeng Yang
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Jinjin Xu
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Yi Lu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fang Wei
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Hu Zhou
- State Key Laboratory of Drug Research, Department of Analytical Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Daming Gao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yunyun Jin
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Lei Zhang
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
18
|
Li Y, Bao K, Sun J, Ge R, Zhang Q, Zhang B, Yan X, Li J, Shi F, Zhang M, Zang J, Liu M, Zhou J, Mi W, Xie S, Chen D, Shi L, Dong C. Design of PROTACs utilizing the E3 ligase GID4 for targeted protein degradation. Nat Struct Mol Biol 2025:10.1038/s41594-025-01537-1. [PMID: 40295770 DOI: 10.1038/s41594-025-01537-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/18/2025] [Indexed: 04/30/2025]
Abstract
Proteolysis targeting chimeras (PROTACs) hijack E3 ligases and the ubiquitin-proteasome system to achieve selective degradation of neo-substrates. Their ability to target otherwise intractable substrates has rendered them a valuable modality in drug discovery. However, only a handful of over 600 human E3 ligases have been functionalized for PROTAC applications. Here we show that the E3 ligase GID4 (glucose-induced degradation deficient complex 4) can be leveraged for targeted protein degradation using a noncovalent small molecule. We design and synthesize GID4-based PROTACs, exemplified by NEP162, which can eliminate endogenous BRD4 in a GID4- and ubiquitin-proteasome system-dependent manner. NEP162 exhibits antiproliferative activity and inhibits tumor growth in a xenograft model, hinting toward potential anticancer applications. We further present the crystal structures of GID4-PROTAC-BRD4 ternary complexes in three distinct states, unveiling plastic interactions between GID4 and BRD4. These structural insights, combined with in vitro and in vivo data, decipher the molecular basis by which the hereby developed PROTACs recruit BRD4 to GID4 for targeted degradation and expand our arsenal of PROTAC-exploitable E3 ligases.
Collapse
Affiliation(s)
- Yanran Li
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Kaiwen Bao
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jiyue Sun
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ruixin Ge
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Qiqing Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Bing Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaojie Yan
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Junlin Li
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Fengying Shi
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Meiling Zhang
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Jinzhi Zang
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Min Liu
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Wenyi Mi
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Songbo Xie
- Department of Ophthalmology, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK 'Belt and Road' Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin, China.
| | - Dongxing Chen
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Lei Shi
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| | - Cheng Dong
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
19
|
Yang G, Liu Y, Gong Z, Chen S, Wang J, Song L, Liu S. Genome wide identification of LcC2DPs gene family in Lotus corniculatus provides insights into regulatory network in response to abiotic stresses. Sci Rep 2025; 15:13380. [PMID: 40251318 PMCID: PMC12008259 DOI: 10.1038/s41598-025-97896-2] [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: 01/04/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
Low temperatures and drought reduce forage yield and quality, with protein kinases crucial for plant stress response. This study examines the LcC2DPs protein kinase family in Lotus corniculatus, identifying 90 members, with some tandemly distributed on chromosomes 2-6, and grouped into 5 subfamilies (I-V). 34 homologous gene pairs were found in Arabidopsis thaliana. LcC2DP genes promoters contain hormone and stress response elements. GO analysis highlights enrichment in hormone response and kinase activity. Transcriptomic analysis links 78 genes to environmental response and stress growth, with 10 validated by qRT-PCR after treatment with 100 μM ABA and IAA, 20% PEG6000, and 4 °C. Protein interaction analysis identifies 5 core proteins (LcC2DP5, 11, 15, 38, and 58) activated by drought and cold stress. Gene analysis revealed that only LcC2DP5 and LcC2DP15 share co-expression transcription factors, with bZIP, bHLH, WRKY, NAC, MYB-related, MYB, C3H, and C2H2 being prominent. These proteins are expressed under drought and cold conditions, highlighting LcC2DP5 and LcC2DP15 activity. NAC and C2H2 are vital for drought response, while bZIP and MYB-related are important for cold response. This suggests that various LcC2DPs in Lotus corniculatus respond to hormones and stress via a TF regulatory network.
Collapse
Affiliation(s)
- Guangfen Yang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, 550025, Guizhou Province, China
- National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, 550025, Guizhou Province, China
| | - Yujie Liu
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Zouxian Gong
- Clinical Medical College, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Siya Chen
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Juanying Wang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, 550025, Guizhou Province, China
- National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, 550025, Guizhou Province, China
| | - Li Song
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, 550025, Guizhou Province, China.
- National-Local Joint Engineering Research Center of Karst Region Plant Resources Utilization & Breeding (Guizhou), Guiyang, 550025, Guizhou Province, China.
| | - Shihui Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, 550025, Guizhou Province, China.
| |
Collapse
|
20
|
Stankey CT, Lee JC. The Role of ETS2 in Macrophage Inflammation. DNA Cell Biol 2025. [PMID: 40227609 DOI: 10.1089/dna.2025.0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025] Open
Abstract
Autoimmune and inflammatory diseases are rising globally yet widely effective therapies remain elusive. Most treatments have limited efficacy, significant potential side effects, or eventually lose response, underscoring the urgent need for new therapeutic approaches. We recently discovered that ETS2, a transcription factor, functions as a master regulator of macrophage-driven inflammation-and is causally linked to the pathogenesis of multiple inflammatory diseases via human genetics. The pleotropic inflammatory effects of ETS2 included upregulation of many cytokines that are individually targeted by current disease therapies, including TNFα, IL-23, IL1β, and TNF-like ligand 1A signaling. With the move toward combination treatment-to maximize efficacy-targeting ETS2 presents a unique opportunity to potentially induce a broad therapeutic effect. However, there will be multiple challenges to overcome since direct ETS2 inhibition is unlikely to be feasible. Here, we discuss these challenges and other unanswered questions about the central role that ETS2 plays in macrophage inflammation.
Collapse
Affiliation(s)
- Christina T Stankey
- Genetic Mechanisms of Disease Lab, The Francis Crick Institute, London, United Kingdom
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Washington University School of Medicine, Saint Louis, Missouri, USA
| | - James Christopher Lee
- Genetic Mechanisms of Disease Lab, The Francis Crick Institute, London, United Kingdom
- Department of Gastroenterology, Royal Free Hospital, London, United Kingdom
- Division of Medicine, Institute for Liver and Digestive Health, University College London, London, United Kingdom
| |
Collapse
|
21
|
Ma Z, Zhou M, Chen H, Shen Q, Zhou J. Deubiquitinase-Targeting Chimeras (DUBTACs) as a Potential Paradigm-Shifting Drug Discovery Approach. J Med Chem 2025; 68:6897-6915. [PMID: 40135978 DOI: 10.1021/acs.jmedchem.4c02975] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Developing proteolysis-targeting chimeras (PROTACs) is well recognized through target protein degradation (TPD) toward promising therapeutics. While a variety of diseases are driven by aberrant ubiquitination and degradation of critical proteins with protective functions, target protein stabilization (TPS) rather than TPD is emerging as a unique therapeutic modality. Deubiquitinase-targeting chimeras (DUBTACs), a class of heterobifunctional protein stabilizers consisting of deubiquitinase (DUB) and protein-of-interest (POI) targeting ligands conjugated with a linker, can rescue such proteins from aberrant elimination. DUBTACs stabilize the levels of POIs in a DUB-dependent manner, removing ubiquitin from polyubiquitylated and degraded proteins. DUBTACs can induce a new interaction between POI and DUB by forming a POI-DUBTAC-DUB ternary complex. Herein, therapeutic benefits of TPS approaches for human diseases are introduced, and recent advances in developing DUBTACs are summarized. Relevant challenges, opportunities, and future perspectives are also discussed.
Collapse
Affiliation(s)
- Zonghui Ma
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Mingxiang Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Qiang Shen
- Department of Interdisciplinary Oncology, School of Medicine, LSU LCMC Health Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| |
Collapse
|
22
|
Zhang X, Hong Y, Hu C, Zhai Y, Pan N, Ding L, Han W, Cui W. Chryxanthone A, an extracted substance from endophytic fungal Aspergillus versicolor, produces anti-oxidant neuroprotection possibly via the action on mTOR/CREB axis. Gene 2025; 944:149298. [PMID: 39884402 DOI: 10.1016/j.gene.2025.149298] [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: 11/07/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Neurons are susceptible to oxidative stress due to the elevated reactive oxygen species (ROS) production and the limited antioxidant defense mechanisms. Therefore, it is possible to treat oxidative stress-related neurological disorders via the inhibition of oxidative stress. Chryxanthone A is an extracted substance derived from the endophytic fungal Aspergillus versicolor, with an atypical dihydropyran ring. However, it is unknown whether and how chryxanthone A could produce anti-oxidant protection. PURPOSES The activity and mechanisms underlying the anti-oxidant protection of chryxanthone A were explored in the study. STUDY DESIGN AND METHODS HT22 neuronal cells were used to evaluate the anti-oxidant protection of chryxanthone A. Comprehensive bioinformatic methods, including RNA-seq analysis, transcription factor prediction, CMap prediction and molecular docking analysis, were utilized to explore the molecular mechanisms how chryxanthone A prevented oxidative stress, which was confirmed by Western blotting analysis. RESULTS Chryxanthone A concentration-dependently prevented H2O2-induced cell death and increase in intracellular ROS in HT22 cells. Results from RNA-seq and bioinformatic analysis indicated that chryxanthone A might act on mTOR/CREB axis, possibly via binding to the Val2227 site within ATP binding pocket of mTOR. The action of chryxanthone A on H2O2-induced alteration of mTOR/CREB axis were further confirmed in HT22 cells. CONCLUSION These results suggested that chryxanthone A produced anti-oxidant protection via the action on mTOR/CREB axis, providing a support that chryxanthone A might be developed as a novel drug candidate for the treatment of oxidative stress-related disorders.
Collapse
Affiliation(s)
- Xinyu Zhang
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China; Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China; Department of Marine Pharmacy, Ningbo University, Zhejiang 315211, China
| | - Yirui Hong
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China
| | - Chenwei Hu
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China
| | - Yijie Zhai
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100 Shaanxi, China
| | - Nanyi Pan
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China
| | - Lijian Ding
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China; Department of Marine Pharmacy, Ningbo University, Zhejiang 315211, China
| | - Wenbo Han
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100 Shaanxi, China.
| | - Wei Cui
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China.
| |
Collapse
|
23
|
Snyder LB, Neklesa TK, Willard RR, Gordon DA, Pizzano J, Vitale N, Robling K, Dorso MA, Moghrabi W, Landrette S, Gedrich R, Lee SH, Taylor IC, Houston JG. Preclinical Evaluation of Bavdegalutamide (ARV-110), a Novel PROteolysis TArgeting Chimera Androgen Receptor Degrader. Mol Cancer Ther 2025; 24:511-522. [PMID: 39670468 PMCID: PMC11962395 DOI: 10.1158/1535-7163.mct-23-0655] [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: 10/20/2023] [Revised: 10/08/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
Androgen receptor (AR) signaling is the principal driver of prostate cancer, and drugs that target this pathway (e.g., abiraterone and enzalutamide) are standard treatments for metastatic hormone-sensitive prostate cancer and metastatic castration-resistant prostate cancer. However, continual evolution during prostate cancer progression can result in AR alterations (e.g., mutation, amplification, and splicing) that can cause tumors to become resistant to these therapies. Bavdegalutamide (ARV-110) is a PROteolysis TArgeting Chimera (PROTAC) protein degrader that recruits the cereblon-containing E3 ubiquitin ligase to direct the polyubiquitination and subsequent proteasomal degradation of AR. Bavdegalutamide selectively degrades wild-type AR and most clinically relevant mutants with low nanomolar potency. The advantages of the degradation mechanism of action are demonstrated by the higher activity of bavdegalutamide relative to the AR antagonist enzalutamide in cell-based systems that assess effects on PSA synthesis, proliferation of prostate cancer cells, and induction of apoptosis. In an AR-expressing patient-derived xenograft mouse model, bavdegalutamide showed substantial AR degradation and greater tumor growth inhibition compared with enzalutamide. Bavdegalutamide also showed robust tumor growth inhibition in enzalutamide- and abiraterone-resistant prostate cancer animal models and enhanced activity in combination with abiraterone. These promising preclinical data supported the clinical development of bavdegalutamide as a potential treatment for patients with prostate cancer. Bavdegalutamide was the first PROTAC protein degrader to enter human clinical trials, specifically in patients with metastatic castration-resistant prostate cancer in a phase I/II study (NCT03888612).
Collapse
Affiliation(s)
- Lawrence B. Snyder
- Department of Chemistry, Arvinas Operations, Inc., New Haven, Connecticut
| | - Taavi K. Neklesa
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Ryan R. Willard
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Deborah A. Gordon
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Jennifer Pizzano
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Nicholas Vitale
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Kaitlynn Robling
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Madeline A. Dorso
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Walid Moghrabi
- Department of General Administration, Arvinas Operations, Inc., New Haven, Connecticut
| | - Sean Landrette
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Richard Gedrich
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Sang Hyun Lee
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - Ian C.A. Taylor
- Department of Biology, Arvinas Operations, Inc., New Haven, Connecticut
| | - John G. Houston
- Department of General Administration, Arvinas Operations, Inc., New Haven, Connecticut
| |
Collapse
|
24
|
Zhang W, Wang J, Li H, Zhang X, Yao D, Zhang H, Zhou X, Nie J, Lai T, Zhu H, Gong Y, Tanaka Y, Li X, Liao X, Su L. TAF7 directly targets SAA1 to enhance triple-negative breast cancer metastasis via phosphorylating E-cadherin and N-cadherin. iScience 2025; 28:111989. [PMID: 40083715 PMCID: PMC11903838 DOI: 10.1016/j.isci.2025.111989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/10/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Identification of metastasis drivers of triple-negative breast cancer (TNBC) is a multifaceted challenge. Here, we identified TATA-box binding protein associated factor 7 (TAF7) as a candidate to modulate TNBC metastasis. TAF7 exhibited high expression in metastatic TNBC patients, and its elevated expression showed a negative correlation with overall survival in TNBC patients. The knockdown of TAF7 suppressed the migration and invasion of TNBC, suggesting TAF7 plays a role in the metastatic processes. Further, TAF7 was enhancing serum amyloid A1 (SAA1) transcription by binding to a specific motif in the SAA1 gene promoter. The elevated SAA1 in TNBC cells directly increased E-cadherin and N-cadherin phosphorylation thereby regulating cell adhesion. Mechanistically, TAF7 modulated cell invasion, migration, and lung metastasis through an SAA1-dependent manner in vitro and in vivo experiments. Taken together, it is likely that TAF7 could directly act on the SAA1 gene promoter, upregulating SAA1 and consequently promoting TNBC metastasis.
Collapse
Affiliation(s)
- Wanjun Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jun Wang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Hanning Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue Zhang
- Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Dunjie Yao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Huimin Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Xinhong Zhou
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiaqi Nie
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tongxing Lai
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Haichuan Zhu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Yiping Gong
- Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Yoshimasa Tanaka
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinghua Liao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Li Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
25
|
Yeh HW, Lang YD, Lee HY, Jou YS. PSPC1 bridges cancer stemness and malignancy in acute myeloid leukemia. Cell Stem Cell 2025; 32:335-337. [PMID: 40054451 DOI: 10.1016/j.stem.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 05/13/2025]
Abstract
In a recent publication in Cell Stem Cell, Hong et al.1 uncovered paraspeckle component 1 (PSPC1) as a determinant of leukemogenic characteristics in acute myeloid leukemia (AML). PSPC1 is not essential for normal hematopoiesis but it is upregulated to interact with PU.1 to promote AML progression and is also emerging as a therapeutic target.
Collapse
Affiliation(s)
- Hsi-Wen Yeh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yaw-Dong Lang
- Department of Medical Research, Precision Medicine Research Center, Center for Precision Health and Quantitative Sciences, Taipei Medical University Hospital, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Yi Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuh-Shan Jou
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
26
|
Li X, Hu H, Wang H, Liu J, Jiang W, Zhou F, Zhang J. DNA nanotechnology-based strategies for minimising hybridisation-dependent off-target effects in oligonucleotide therapies. MATERIALS HORIZONS 2025; 12:1388-1412. [PMID: 39692461 DOI: 10.1039/d4mh01158a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Targeted therapy has emerged as a transformative breakthrough in modern medicine. Oligonucleotide drugs, such as antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs), have made significant advancements in targeted therapy. Other oligonucleotide-based therapeutics like clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein (Cas) systems are also leading a revolution in targeted gene therapy. However, hybridisation-dependent off-target effects, arising from imperfect base pairing, remain a significant and growing concern for the clinical translation of oligonucleotide-based therapeutics. These mismatches in base pairing can lead to unintended steric blocking or cleavage events in non-pathological genes, affecting the efficacy and safety of the oligonucleotide drugs. In this review, we examine recent developments in oligonucleotide-based targeted therapeutics, explore the factors influencing sequence-dependent targeting specificity, and discuss the current approaches employed to reduce the off-target side effects. The existing strategies, such as chemical modifications and oligonucleotide length optimisation, often require a trade-off between specificity and binding affinity. To further address the challenge of hybridisation-dependent off-target effects, we discuss DNA nanotechnology-based strategies that leverage the collaborative effects of nucleic acid assembly in the design of oligonucleotide-based therapies. In DNA nanotechnology, collaborative effects refer to the cooperative interactions between individual strands or nanostructures, where multiple bindings result in more stable and specific hybridisation behaviour. By requiring multiple complementary interactions to occur simultaneously, the likelihood of unintended partially complementary binding events in nucleic acid hybridisation should be reduced. And thus, with the aid of collaborative effects, DNA nanotechnology has great promise in achieving both high binding affinity and high specificity to minimise the hybridisation-dependent off-target effects of oligonucleotide-based therapeutics.
Collapse
Affiliation(s)
- Xiaoyu Li
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China.
- Ningbo Cixi Institute of Biomedical Engineering, Ningbo, China
| | - Huanhuan Hu
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China.
- Ningbo Cixi Institute of Biomedical Engineering, Ningbo, China
| | - Hailong Wang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China.
- Ningbo Cixi Institute of Biomedical Engineering, Ningbo, China
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, China
| | - Jia Liu
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China.
- Ningbo Cixi Institute of Biomedical Engineering, Ningbo, China
| | - Wenting Jiang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China.
- Ningbo Cixi Institute of Biomedical Engineering, Ningbo, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Feng Zhou
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China.
- Ningbo Cixi Institute of Biomedical Engineering, Ningbo, China
| | - Jiantao Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China.
- Ningbo Cixi Institute of Biomedical Engineering, Ningbo, China
| |
Collapse
|
27
|
Yang J, Zhou F, Luo X, Fang Y, Wang X, Liu X, Xiao R, Jiang D, Tang Y, Yang G, You L, Zhao Y. Enhancer reprogramming: critical roles in cancer and promising therapeutic strategies. Cell Death Discov 2025; 11:84. [PMID: 40032852 DOI: 10.1038/s41420-025-02366-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 01/24/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
Transcriptional dysregulation is a hallmark of cancer initiation and progression, driven by genetic and epigenetic alterations. Enhancer reprogramming has emerged as a pivotal driver of carcinogenesis, with cancer cells often relying on aberrant transcriptional programs. The advent of high-throughput sequencing technologies has provided critical insights into enhancer reprogramming events and their role in malignancy. While targeting enhancers presents a promising therapeutic strategy, significant challenges remain. These include the off-target effects of enhancer-targeting technologies, the complexity and redundancy of enhancer networks, and the dynamic nature of enhancer reprogramming, which may contribute to therapeutic resistance. This review comprehensively encapsulates the structural attributes of enhancers, delineates the mechanisms underlying their dysregulation in malignant transformation, and evaluates the therapeutic opportunities and limitations associated with targeting enhancers in cancer.
Collapse
Affiliation(s)
- Jinshou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Feihan Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xiyuan Luo
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Yuan Fang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Decheng Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Yuemeng Tang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
28
|
Katz LS, Visser EJ, Plitzko KF, Pennings MAM, Cossar PJ, Tse IL, Kaiser M, Brunsveld L, Ottmann C, Scott DK. Molecular glues of the regulatory ChREBP/14-3-3 complex protect beta cells from glucolipotoxicity. Nat Commun 2025; 16:2110. [PMID: 40025013 PMCID: PMC11873037 DOI: 10.1038/s41467-025-57241-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025] Open
Abstract
The Carbohydrate Response Element Binding Protein (ChREBP) is a glucose-responsive transcription factor (TF) with two major splice isoforms (α and β). In chronic hyperglycemia and glucolipotoxicity, ChREBPα-mediated ChREBPβ expression surges, leading to insulin-secreting β-cell dedifferentiation and death. 14-3-3 binding to ChREBPα results in cytoplasmic retention and suppression of transcriptional activity. Thus, small molecule-mediated stabilization of this protein-protein interaction (PPI) may be of therapeutic value. Here, we show that structure-based optimizations of a 'molecular glue' compound led to potent ChREBPα/14-3-3 PPI stabilizers with cellular activity. In primary human β-cells, the most active compound retained ChREBPα in the cytoplasm, and efficiently protected β-cells from glucolipotoxicity while maintaining β-cell identity. This study may thus not only provide the basis for the development of a unique class of compounds for the treatment of Type 2 Diabetes but also showcases an alternative 'molecular glue' approach for achieving small molecule control of notoriously difficult to target TFs.
Collapse
Affiliation(s)
- Liora S Katz
- Diabetes, Obesity and Metabolism Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emira J Visser
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Kathrin F Plitzko
- Chemical Biology, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Marloes A M Pennings
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Peter J Cossar
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Isabelle L Tse
- Diabetes, Obesity and Metabolism Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Markus Kaiser
- Chemical Biology, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany.
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
29
|
Huang F, Li K, Chen Z, Cui Z, Hankey W, Fang K, Yan J, Wang H, Jin VX, Dong Y, Wang Q. Integrative analysis identifies the atypical repressor E2F8 as a targetable transcriptional activator driving lethal prostate cancer. Oncogene 2025; 44:481-493. [PMID: 39613933 DOI: 10.1038/s41388-024-03239-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024]
Abstract
Acquired resistance to androgen receptor (AR)-targeted therapies underscores the need to identify alternative therapeutic targets for treating lethal prostate cancer. In this study, we evaluated the prognostic significance of 1635 human transcription factors (TFs) by analyzing castration-resistant prostate cancer (CRPC) datasets from the West and East Stand Up to Cancer (SU2C) cohorts. Through this screening approach, we identified E2F8, a putative transcriptional repressor, as a TF consistently associated with poorer patient outcomes in both cohorts. Notably, E2F8 is highly expressed and active in AR-negative CRPC compared to AR-positive CRPC. Integrative profiling of E2F8 cistromes and transcriptomes in AR-negative CRPC cells revealed that E2F8 directly and non-canonically activates target oncogenes involved in cancer-associated pathways. To target E2F8 in CRPC, we employed the CRISPR/CasRx system to knockdown E2F8 mRNA, resulting in effective and specific downregulation of E2F8 and its target oncogenes, as well as significant growth inhibition in AR-negative CRPC in both cultured cells and xenograft models. Our findings identify and characterize E2F8 as a targetable transcriptional activator driving CRPC, particularly the growth of AR-negative CRPC.
Collapse
Affiliation(s)
- Furong Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Kexin Li
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Zhong Chen
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Zhifen Cui
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - William Hankey
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Kun Fang
- Data Science Institute, MCW Cancer Center and Mellowes Center for Genome Science and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jingyue Yan
- Icahn Genomics Institute, Precision Immunology Institute, Department of Immunology and Immunotherapy, Department of Oncological Sciences, Tisch Cancer Institute, Biomedical Engineering and Imaging Institute, Friedman Brain Institute , Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hongyan Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Victor X Jin
- Data Science Institute, MCW Cancer Center and Mellowes Center for Genome Science and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yizhou Dong
- Icahn Genomics Institute, Precision Immunology Institute, Department of Immunology and Immunotherapy, Department of Oncological Sciences, Tisch Cancer Institute, Biomedical Engineering and Imaging Institute, Friedman Brain Institute , Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qianben Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
30
|
Neil CR, Schaening-Burgos C, Alexis MS, Reynolds DJ, Smith PG, Seiler MW, Vaillancourt FH, Agrawal AA. Poison exons: tuning RNA splicing for targeted gene regulation. Trends Pharmacol Sci 2025; 46:264-278. [PMID: 39915130 DOI: 10.1016/j.tips.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 03/09/2025]
Abstract
Poison exons (PEs) are a class of alternatively spliced exons whose inclusion targets mRNA transcripts for degradation via the nonsense-mediated decay (NMD) pathway. Although a role for NMD as an essential mRNA quality control pathway has long been appreciated, recent advances in RNA sequencing (RNA-seq) strategies and analyses have revealed that its coupling to RNA splicing is broadly used to regulate mRNA stability and abundance. Regulation of PE splicing affects patterns of targeted degradation across the transcriptome and influences gene expression in both healthy and disease states. Importantly, PEs represent a novel therapeutic opportunity to modulate the expression of disease-relevant genes with sequence-specific resolution. We review the emergence of PE splicing in endogenous gene regulation, its misregulation in disease, and the ways in which it can be leveraged for therapeutic benefit.
Collapse
|
31
|
Wang W, Du Y, Datta S, Fowler JF, Sang HT, Albadari N, Li W, Foster J, Zhang R. Targeting the MYCN-MDM2 pathways for cancer therapy: Are they druggable? Genes Dis 2025; 12:101156. [PMID: 39802403 PMCID: PMC11719324 DOI: 10.1016/j.gendis.2023.101156] [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: 05/15/2023] [Revised: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 01/16/2025] Open
Abstract
Targeting oncogenes and their interactive partners is an effective approach to developing novel targeted therapies for cancer and other chronic diseases. We and others have long suggested the MDM2 oncogene being an excellent target for cancer therapy, based on its p53-dependent and -independent oncogenic activities in a variety of cancers. The MYC family proteins are transcription factors that also regulate diverse biological functions. Dysregulation of MYC, such as amplification of MYCN, is associated with tumorigenesis, especially for neuroblastoma. Although the general survival rate of neuroblastoma patients has significantly improved over the past few decades, high-risk neuroblastoma still presents a poor prognosis. Therefore, innovative and more potent therapeutic strategies are needed to eradicate these aggressive neoplasms. This review focuses on the oncogenic properties of MYCN and its molecular regulation and summarizes the major therapeutic strategies being developed based on preclinical findings. We also highlight the potential benefits of targeting both the MYCN and MDM2 oncogenes, providing preclinical evidence of the efficacy and safety of this approach. In conclusion, the development of effective small molecules that inhibit both MYCN and MDM2 represents a promising new strategy for the treatment of neuroblastoma and other cancers.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
- Drug Discovery Institute, University of Houston, Houston, TX 77204, USA
| | - Yi Du
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Josef F. Fowler
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Hannah T. Sang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Najah Albadari
- College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wei Li
- College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jennifer Foster
- Texas Children's Hospital, Department of Pediatrics, Section of Hematology-Oncology Baylor College of Medicine, Houston, TX 77030, USA
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
- Drug Discovery Institute, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
32
|
Rosen HT, Li K, Li EL, Currier B, Brittain SM, Garcia FJ, Beard DC, Haenni-Holzinger S, Dovala D, McKenna JM, Schirle M, Maimone TJ, Nomura DK. Sulfinyl Aziridines as Stereoselective Covalent Destabilizing Degraders of the Oncogenic Transcription Factor MYC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639755. [PMID: 40060528 PMCID: PMC11888305 DOI: 10.1101/2025.02.24.639755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
While MYC is a significant oncogenic transcription factor driver of cancer, directly targeting MYC has remained challenging due to its intrinsic disorder and poorly defined structure, deeming it "undruggable." Whether transient pockets formed within intrinsically disordered and unstructured regions of proteins can be selectively targeted with small molecules remains an outstanding challenge. Here, we developed a bespoke stereochemically-paired spirocyclic oxindole aziridine covalent library and screened this library for degradation of MYC. Through this screen, we identified a hit covalent ligand KL2-236, bearing a unique sulfinyl aziridine warhead, that engaged MYC in vitro as pure MYC/MAX protein complex and in situ in cancer cells to destabilize MYC, inhibit MYC transcriptional activity and degrade MYC in a proteasome-dependent manner through targeting intrinsically disordered C203 and D205 residues. Notably, this reactivity was most pronounced for specific stereoisomers of KL2-236 with a diastereomer KL4-019 that was largely inactive. Mutagenesis of both C203 and D205 completely attenuated KL2-236-mediated MYC degradation. We have also optimized our initial KL2-236 hit compound to generate a more durable MYC degrader KL4-219A in cancer cells. Our results reveal a novel ligandable site within MYC and indicate that certain intrinsically disordered regions within high-value protein targets, such as MYC, can be interrogated by isomerically unique chiral small molecules, leading to destabilization and degradation.
Collapse
Affiliation(s)
- Hannah T. Rosen
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Kelvin Li
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Erin L. Li
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Brynne Currier
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Scott M. Brittain
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Francisco J. Garcia
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Diana C. Beard
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Sandra Haenni-Holzinger
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Dustin Dovala
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Jeffrey M. McKenna
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Markus Schirle
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
- Novartis Biomedical Research, Emeryville, CA USA; Cambridge, MA USA; Basel, Switzerland
| | - Thomas J. Maimone
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| | - Daniel K. Nomura
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
- Novartis-Berkeley Translational Chemical Biology Institute, Berkeley, CA 94720 USA
| |
Collapse
|
33
|
Steinhauser S, Estoppey D, Buehler DP, Xiong Y, Pizzato N, Rietsch A, Wu F, Leroy N, Wunderlin T, Claerr I, Tropberger P, Müller M, Vissieres A, Davison LM, Farber-Eger E, Wells QS, Sheng Q, Bergling S, Wild S, Moulin P, Liang J, English WJ, Williams B, Knehr J, Altorfer M, Reyes A, Voshol J, Mickanin C, Hoepfner D, Nigsch F, Frederiksen M, Flynn CR, Fodor BD, Brown JD, Kolter C. The transcription factor ZNF469 regulates collagen production in liver fibrosis. JCI Insight 2025; 10:e182232. [PMID: 39998893 PMCID: PMC11981625 DOI: 10.1172/jci.insight.182232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) - characterized by excess accumulation of fat in the liver - now affects one-third of the world's population. As MASLD progresses, extracellular matrix components including collagen accumulate in the liver, causing tissue fibrosis, a major determinant of disease severity and mortality. To identify transcriptional regulators of fibrosis, we computationally inferred the activity of transcription factors (TFs) relevant to fibrosis by profiling the matched transcriptomes and epigenomes of 108 human liver biopsies from a deeply characterized cohort of patients spanning the full histopathologic spectrum of MASLD. CRISPR-based genetic KO of the top 100 TFs identified ZNF469 as a regulator of collagen expression in primary human hepatic stellate cells (HSCs). Gain- and loss-of-function studies established that ZNF469 regulates collagen genes and genes involved in matrix homeostasis through direct binding to gene bodies and regulatory elements. By integrating multiomic large-scale profiling of human biopsies with extensive experimental validation, we demonstrate that ZNF469 is a transcriptional regulator of collagen in HSCs. Overall, these data nominate ZNF469 as a previously unrecognized determinant of MASLD-associated liver fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fabian Wu
- Novartis Biomedical Research, Basel, Switzerland
| | - Nelly Leroy
- Novartis Biomedical Research, Basel, Switzerland
| | | | | | | | | | | | | | | | | | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | | | - Sophia Wild
- Novartis Biomedical Research, Basel, Switzerland
| | - Pierre Moulin
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University, Lausanne, Switzerland
- Hospital and Lausanne University, Lausanne, Switzerland
- Chief Scientific Officer, Deciphex Ltd., Dublin, Ireland
| | - Jiancong Liang
- Department of Pathology, Microbiology, and Immunology, VUMC, Nashville, Tennessee, USA
| | | | | | - Judith Knehr
- Novartis Biomedical Research, Basel, Switzerland
| | | | | | | | - Craig Mickanin
- Novartis Biomedical Research, Cambridge, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Paul A, Terrell JR, Farahat AA, Ogbonna EN, Kumar A, Boykin DW, Neidle S, Wilson WD. Alternative Approach to Sequence-Specific Recognition of DNA: Cooperative Stacking of Dication Dimers─Sensitivity to Compound Curvature, Aromatic Structure, and DNA Sequence. ACS Chem Biol 2025; 20:489-506. [PMID: 39920086 PMCID: PMC11851451 DOI: 10.1021/acschembio.4c00800] [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: 11/23/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/09/2025]
Abstract
With the growing number and diversity of known genome sequences, there is an increasing opportunity to regulate gene expression through synthetic, cell-permeable small molecules. Enhancing the DNA sequence recognition abilities of minor groove compounds has the potential to broaden their therapeutic applications with significant implications for areas such as modulating transcription factor activity. While various classes of minor groove binding agents can selectively identify pure AT and mixed AT and GC base pair(s) containing sequences, there remains a lack of compounds capable of distinguishing between different AT sequences. In this work, we report on the design compounds that exhibit selective binding to -TTAA- or -TATA- containing DNA minor groove sequences compared with other AT ones. Several studies have shown that the -AATT- and -TTAA- sequences have distinct physical and interaction properties, especially in terms of their different requirements for recognition in the minor groove. Achieving strong, selective minor groove binding at -TTAA- sequences has been challenging, but DB1003, a benzimidazole-furan-furan diamidine, has demonstrated cooperative dimeric binding activity at -TTAA-. It has significantly less binding preference for AATT. To better understand and modify the selectivity, we synthesized a set of rationally designed analogs of DB1003 by altering the position of the five-membered heterocyclic structure. Binding affinities and stoichiometries obtained from biosensor-surface plasmon resonance experiments show that DB1992, a benzimidazolefuran-thiophene diamidine, binds strongly to -TTAA- as a positive cooperative dimer with high cooperativity. The high-resolution crystal structure of the TTAA-DNA-DB1992 complex reveals that DB1992 binds as an antiparallel π-stacked dimer with numerous diverse contacts to the DNA minor groove. This distinctive binding arrangement and the properties of diamidines at the -TTAA- minor groove demonstrate that benzimidazole-furan-thiophene is a unique DNA binding pharmacophore. Competition mass spectroscopy and circular dichroism studies confirmed the binding stoichiometry and selectivity preference of the compounds for the -TTAA- sequence.
Collapse
Affiliation(s)
- Ananya Paul
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - J. Ross Terrell
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - Abdelbasset A. Farahat
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
- Department
of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- Master
of Pharmaceutical Sciences Program, California
North State University, Elk Grove, California 95757, United States
| | - Edwin N. Ogbonna
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - Arvind Kumar
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - David W. Boykin
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - Stephen Neidle
- School
of Pharmacy, University College London, London WC1N 1AX, U.K.
| | - W. David Wilson
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| |
Collapse
|
35
|
Newman JA, Gavard AE, Imprachim N, Aitkenhead H, Sheppard HE, Te Poele R, Clarke PA, Hossain MA, Temme L, Oh HJ, Wells CI, Davis-Gilbert ZW, Workman P, Gileadi O, Drewry DH. Structural insights into human brachyury DNA recognition and discovery of progressible binders for cancer therapy. Nat Commun 2025; 16:1596. [PMID: 39952925 PMCID: PMC11828899 DOI: 10.1038/s41467-025-56213-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 01/10/2025] [Indexed: 02/17/2025] Open
Abstract
Brachyury is a transcription factor that plays an essential role in tumour growth of the rare bone cancer chordoma and is implicated in other solid tumours. Brachyury is minimally expressed in healthy tissues, making it a potential therapeutic target. Unfortunately, as a ligandless transcription factor, brachyury has historically been considered undruggable. To investigate direct targeting of brachyury by small molecules, we determine the structure of human brachyury both alone and in complex with DNA. The structures provide insights into DNA binding and the context of the chordoma associated G177D variant. We use crystallographic fragment screening to identify hotspots on numerous pockets on the brachyury surface. Finally, we perform follow-up chemistry on fragment hits and describe the progression of a thiazole chemical series into binders with low µM potency. Thus we show that brachyury is ligandable and provide an example of how crystallographic fragment screening may be used to target protein classes that are difficult to address using other approaches.
Collapse
Affiliation(s)
- Joseph A Newman
- Centre for Medicines Discovery, University of Oxford, Oxford, UK.
| | - Angeline E Gavard
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Exscientia, Oxford, UK
| | - Nergis Imprachim
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Hazel Aitkenhead
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Diamond Light Source Ltd, Didcot, UK
| | - Hadley E Sheppard
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK
- Sano Genetics Ltd, Cambridge, UK
| | - Robert Te Poele
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK
| | - Paul A Clarke
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK
| | - Mohammad Anwar Hossain
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Louisa Temme
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Hans J Oh
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carrow I Wells
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- GlaxoSmithKline, Collegeville, PA, USA
| | - Zachary W Davis-Gilbert
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paul Workman
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK.
| | - Opher Gileadi
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- SGC Karolinska, Centre for Molecular Medicine, Stockholm, Sweden
| | - David H Drewry
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
36
|
Gao P, Yuan S, Wang Y, Wang Y, Li X, Liu T, Zheng Y, Wang J, Liu D, Xu L, Jiang Y, Zeng K, Tu P. Corydalis decumbens and tetrahydropalmatrubin inhibit macrophages inflammation to relieve rheumatoid arthritis by targeting Fosl2. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119348. [PMID: 39805480 DOI: 10.1016/j.jep.2025.119348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Corydalisdecumbens (Thunb.) (CD) is a traditional Chinese medicine and as a single herb or formula has been used to treat RA for decades. Rheumatoid arthritis (RA) is a persistent, systemic autoimmune inflammatory disease. However, the anti-inflammatory target, effective constituents and mechanism was unclear. AIM OF THE STUDY The purpose of this study was to identify anti-RA and anti-inflammatory targets of CD, elucidate effective constituents and molecular pharmacological mechanism. MATERIALS AND METHODS Anti-RA and anti-inflammatory effect of CD were evaluated on CIA-rats and in LPS-induced RAW264.7 cells respectively. The anti-inflammatory target of CD was identified using thermal proteome profiling (TPP). The recombinant Fosl2 protein was expressed and purified and the target-based effective constituents was screened with bio-layer interferometry (BLI) analysis. Combining photoaffinity probe, LC-MS/MS analysis, docking and point mutation, the binding site was confirmed between Fosl2 and THP. Furtherly, immunofluorescence (IF), co-immunoprecipitation (co-IP) were used to research the pharmacological mechanism of THP and the THP-influenced downstream pathways were elucidated by transcriptomics analysis. RESULTS CD had therapeutic effect on CIA-rats and a significant anti-inflammation on macrophages. Fosl2 was identified as a target of CD and we elucidated the target-based effective constituents was protoberberine-type alkaloids. THP can inhibit inflammation and transcription of AP-1 via targeting Fosl2 on LPS-induced RAW264.7 cells. For mechanism, THP promoted Fosl2 nuclear translocating and interacting with c-Jun. CONCLUSIONS These findings firstly elucidated the target and effective constituents of CD treating RA, and found that "undruggable target" Fosl2 can be used as therapeutic targets for RA. Meanwhile, our research suggested that THP has a significant potential for the treatment of RA.
Collapse
Affiliation(s)
- Peng Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Shuo Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yanhang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yuqi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiaoshuang Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Tingting Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yongzhe Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Dan Liu
- Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing, 100191, China
| | - Luzheng Xu
- Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing, 100191, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
37
|
Chen H, Xu Y, Chen D, Xiao D, Yang B, Wang W, Han H. The Hippo pathway promotes platinum-based chemotherapy by inhibiting MTF1-dependent heavy metal response. BMC Cancer 2025; 25:223. [PMID: 39920630 PMCID: PMC11806854 DOI: 10.1186/s12885-025-13661-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/05/2025] [Indexed: 02/09/2025] Open
Abstract
The platinum-based compounds are widely used in treating various types of cancer through their heavy metal component platinum. However, the development of chemoresistance often limits their clinical effectiveness. In this study, we report the roles of heavy metal response and its associated Hippo pathway in regulating platinum-based chemotherapy. Our data show that the MTF1-dependent heavy metal response induces cancer cell resistance to platinum-based compounds both in vitro and in vivo. This resistance is mitigated by Hippo pathway-mediated phosphorylation of MTF1. Moreover, pharmacological activation of the Hippo pathway sensitizes cancer cells to platinum-based compounds. Clinically, lung adenocarcinoma (LUAD) patients with high MTF1 activity exhibit poor overall survival rates, and Hippo pathway inactivation is positively correlated with elevated MTF1 transcriptional activity in platinum-treated LUAD patients. Collectively, our findings not only unveil a critical role of the Hippo-MTF1 pathway in regulating the response to platinum-based chemotherapy, but also suggest new strategies to enhance its efficacy by targeting the heavy metal response.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China
| | - Yue Xu
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China
| | - Dingshan Chen
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China
| | - Di Xiao
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China
| | - Bing Yang
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA.
| | - Han Han
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
38
|
Lyu H, Chen X, Cheng Y, Zhang T, Wang P, Wong JHY, Wang J, Stasiak L, Sun L, Yang G, Wang L, Yue F. Pioneer factor GATA6 promotes colorectal cancer through 3D genome regulation. SCIENCE ADVANCES 2025; 11:eads4985. [PMID: 39919174 PMCID: PMC11804904 DOI: 10.1126/sciadv.ads4985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025]
Abstract
Colorectal cancer (CRC) is one of the most lethal and prevalent malignancies. While the overexpression of pioneer factor GATA6 in CRC has been linked with metastasis, its role in genome-wide gene expression dysregulation remains unclear. Through studies of primary human CRC tissues and analysis of the TCGA data, we found that GATA6 preferentially binds at CRC-specific active enhancers, with enrichment at enhancer-promoter loop anchors. GATA6 protein also physically interacts with CTCF, suggesting its critical role in 3D genome organization. The ablation of GATA6 through AID and CRISPR systems severely impaired cancer cell clonogenicity and proliferation. Mechanistically, GATA6 knockout induced global loss of CRC-specific open chromatins and extensive alterations of critical enhancer-promoter interactions for CRC oncogenes. Last, we showed that GATA6 knockout greatly reduced tumor growth and improved survival in mice. Together, we revealed a previously unidentified mechanism by which GATA6 contributes to the pathogenesis of colorectal cancer.
Collapse
Affiliation(s)
- Huijue Lyu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xintong Chen
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yang Cheng
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Te Zhang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ping Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Josiah Hiu-yuen Wong
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Juan Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lena Stasiak
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leyu Sun
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Guangyu Yang
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Lu Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
39
|
Zhou M, Li S, Tan Y, Huang W, Li Y, Yuan X, Li Z. Global Profiling Lysine Reactivity and Ligandability with Oxidant-Triggered Bioconjugation Chemistry. Angew Chem Int Ed Engl 2025; 64:e202418473. [PMID: 39543955 DOI: 10.1002/anie.202418473] [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: 09/25/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/17/2024]
Abstract
Due to the high abundance and diverse functions of lysine residues, both in the interior and on the surface of proteins, the development of new methods to characterize their reactivity and ligandability could significantly expand the pool of druggable targets. To date, only a limited number of aminophilic electrophiles have been assessed for interactions with the lysine proteome, resulting in a substantial fraction remaining inaccessible to current probes. Here, to the best of our knowledge, we report the first oxidant-triggered bioconjugation platform for in-depth profiling of lysines. We quantified over 7000 covalently modifiable lysine residues, which significantly expands the coverage of ligandable lysines in the whole proteome. Chemical proteomics enabled the mapping of more than 100 endogenous kinases, thus providing a comprehensive landscape of ligandable catalytic lysines within the kinome. Moreover, we identified a suite of new ligandable lysines such as K60 of ENO1 and K31 of PPIA, offering insights for exploring new functional and targetable residues. These findings could provide valuable clues for the development of targeted covalent inhibitors (TCIs).
Collapse
Affiliation(s)
- Mengya Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632, Guangzhou, China
| | - Shengrong Li
- Guangdong Second Provincial General Hospital, Postdoctoral Station of Traditional Chinese Medicine, Jinan University, 510632, Guangzhou, China
| | - Yi Tan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632, Guangzhou, China
| | - Weizhen Huang
- The First Huizhou Affiliated Hospital of Guangdong Medical University, 516001, Huizhou, China
| | - Yifang Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632, Guangzhou, China
| | - Xia Yuan
- The First Huizhou Affiliated Hospital of Guangdong Medical University, 516001, Huizhou, China
| | - Zhengqiu Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632, Guangzhou, China
| |
Collapse
|
40
|
García-Díaz N, Solli E, Hajjar E, Cornillot-Clément S, Landskron J, Ahmad R, Wei Q, Taskén K. MAPK and STAT3 Inhibitors Modulate FoxP3 Expression and Regulatory T Cell Function. Eur J Immunol 2025; 55:e202451225. [PMID: 39955647 DOI: 10.1002/eji.202451225] [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: 04/26/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
Regulatory T cells (Tregs) are a subset of T cells defined by the expression of Forkhead box protein P3 (FoxP3) playing a crucial role in regulating effector T cell activity. Tregs accumulate in the tumor microenvironment facilitating tumor growth. Thus, targeting FoxP3+ Tregs could improve cancer immunotherapies. Here, we conducted a high-throughput, phenotypic screening of a drug repurposing library to identify compounds downregulating FoxP3 expression in human primary T cells. We identified the tyrosine kinase inhibitor bosutinib and the STAT3 inhibitor nifuroxazide effectively downregulating FoxP3 expression. To identify more potent compounds, structural analogs of these two compounds were searched and validated. These analogs were found to reduce FoxP3 expression in a similar- or more potent manner than the original hits. All compounds inhibited Treg suppressive functions and reduced the expression of Treg activation markers. Importantly, bosutinib disrupted FAK and CaMKII signaling more potently in Tregs, whilst nifuroxazide and its analog NA16 targeted STAT3 protein levels more effectively in Tregs. Additionally, bosutinib and NA16 targeted effector Tregs more effectively than other Treg subsets. In summary, bosutinib, nifuroxazide, and their analogs inhibited FoxP3 expression, Treg suppressive abilities, and Treg activation effectively, which could serve as tools for the improvement of current cancer immunotherapies.
Collapse
Affiliation(s)
- Nuria García-Díaz
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Elise Solli
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ehsan Hajjar
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Selma Cornillot-Clément
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Johannes Landskron
- Centre for Molecular Medicine, Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | - Rafi Ahmad
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Biotechnology, University of Inland Norway, Hamar, Norway
| | - Qian Wei
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for B-cell Malignancies, University of Oslo, Oslo, Norway
| |
Collapse
|
41
|
Zhang G, Yan S, Liu Y, Du Z, Min Q, Qin S. PROTACs coupled with oligonucleotides to tackle the undruggable. Bioanalysis 2025; 17:261-276. [PMID: 39895280 PMCID: PMC11864318 DOI: 10.1080/17576180.2025.2459528] [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: 12/31/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
Undruggable targets account for roughly 85% of human disease-related targets and represent a category of therapeutic targets that are difficult to tackle with traditional methods, but their considerable clinical importance. These targets are generally defined by planar functional interfaces and the absence of efficient ligand-binding pockets, making them unattainable for conventional pharmaceutical strategies. The advent of oligonucleotide-based proteolysis-targeting chimeras (PROTACs) has instilled renewed optimism in addressing these challenges. These PROTACs facilitate the targeted degradation of undruggable entities, including transcription factors (TFs) and RNA-binding proteins (RBPs), via proteasome-dependent mechanisms, thereby presenting novel therapeutic approaches for diseases linked to these targets. This review offers an in-depth examination of recent progress in the integration of PROTAC technology with oligonucleotides to target traditionally undruggable proteins, emphasizing the design principles and mechanisms of action of these innovative PROTACs.
Collapse
Affiliation(s)
- Guangshuai Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Si Yan
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Yan Liu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Ziwei Du
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Qin Min
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Shuanglin Qin
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, P.R. China
| |
Collapse
|
42
|
Patalano SD, Fuxman Bass P, Fuxman Bass JI. Transcription factors in the development and treatment of immune disorders. Transcription 2025; 16:118-140. [PMID: 38100543 PMCID: PMC11970766 DOI: 10.1080/21541264.2023.2294623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Immune function is highly controlled at the transcriptional level by the binding of transcription factors (TFs) to promoter and enhancer elements. Several TF families play major roles in immune gene expression, including NF-κB, STAT, IRF, AP-1, NRs, and NFAT, which trigger anti-pathogen responses, promote cell differentiation, and maintain immune system homeostasis. Aberrant expression, activation, or sequence of isoforms and variants of these TFs can result in autoimmune and inflammatory diseases as well as hematological and solid tumor cancers. For this reason, TFs have become attractive drug targets, even though most were previously deemed "undruggable" due to their lack of small molecule binding pockets and the presence of intrinsically disordered regions. However, several aspects of TF structure and function can be targeted for therapeutic intervention, such as ligand-binding domains, protein-protein interactions between TFs and with cofactors, TF-DNA binding, TF stability, upstream signaling pathways, and TF expression. In this review, we provide an overview of each of the important TF families, how they function in immunity, and some related diseases they are involved in. Additionally, we discuss the ways of targeting TFs with drugs along with recent research developments in these areas and their clinical applications, followed by the advantages and disadvantages of targeting TFs for the treatment of immune disorders.
Collapse
Affiliation(s)
- Samantha D. Patalano
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Paula Fuxman Bass
- Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I. Fuxman Bass
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
43
|
Yu P, Ye S, Zhou M, Zhang L, Zhang Z, Sun X, Li S, Hu C. PWWP domain-containing protein Crf4-3 specifically modulates fungal azole susceptibility by regulating sterol C-14 demethylase ERG11. mSphere 2025; 10:e0070324. [PMID: 39670730 PMCID: PMC11774033 DOI: 10.1128/msphere.00703-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/22/2024] [Indexed: 12/14/2024] Open
Abstract
The widespread use of azole antifungals in agriculture and clinical settings has led to serious drug resistance. Overexpression of the azole drug target 14α-demethylase ERG11 (CYP51) is the most common fungal resistance mechanism. However, the presence of additional regulatory proteins in the transcriptional response of erg11 is not yet fully elucidated. In this study, leveraging the identified key promoter region of erg11 that controls its response to azoles in Neurospora crassa, we pinpointed a protein, Crf4-3, which harbors a PWWP domain and exerts a positive regulatory influence on azole resistance, as determined by DNA pulldown assays. The removal of Crf4-3 results in heightened sensitivity to azoles while remaining unaffected by other stressors tested. Additionally, the deletion leads to the abolition of transcriptional responses of genes such as erg11 and erg6 to ketoconazole. Interestingly, the basal expression of erg1, erg11, erg25, and erg3A is also affected by the deletion of crf4-3, indicating its role in sterol homeostasis. Crf4-3 homologs are broadly distributed across the Pezizomycotina fungi. The gene deletion for its homologous protein in Aspergillus fumigatus also significantly improves sensitivity to azoles such as voriconazole, primarily through the attenuation of the transcriptional response of erg11. Our data, for the first time, identified Crf4-3 as a novel regulatory protein in the azole stress response of filamentous fungi, offering fresh insights into the mechanisms of azole resistance.IMPORTANCETranscriptional control of pivotal genes, such as erg11, stands as the primary driver of azole resistance. Although considerable effort has been dedicated to identifying transcription factors involved, our knowledge regarding the use of transcriptional regulation strategies to combat azole resistance is currently limited. In this study, we reveal that a PWWP domain-containing protein Crf4-3, which is conserved in Pezizomycotina fungi, modulates fungal azole sensitivity by transcriptionally regulating sterol biosynthetic genes, including erg11. These results also broaden the understanding of fungal PWWP domain-containing proteins regarding their roles in regulating resistance against azole antifungals. Considering research on small molecules targeting the PWWP domain in humans, Crf4-3 homolog emerges as a promising target for designing fungal-specific drugs to combat azole resistance.
Collapse
Affiliation(s)
- Pengju Yu
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuting Ye
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Mi Zhou
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Long Zhang
- Shandong Jinniu Group Co., Ltd., Jinan, China
| | | | - Xianyun Sun
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shaojie Li
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chengcheng Hu
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
44
|
Yu S, Jiang C, Yang Y, Cheng F, Liu F, Liu C, Gong X. Purine-rich element binding protein alpha: a DNA/RNA binding protein with multiple roles in cancers. Mol Med 2025; 31:20. [PMID: 39844051 PMCID: PMC11755881 DOI: 10.1186/s10020-025-01087-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
Proteins that bind to DNA/RNA are typically evolutionarily conserved with multiple regulatory functions in transcription initiation, mRNA translation, stability of RNAs, and RNA splicing. Therefore, dysregulation of DNA/RNA binding proteins such as purine-rich element binding protein alpha (PURα) disrupts signaling transduction and often leads to human diseases including cancer. PURα was initially recognized as a tumor suppressor in acute myeloid leukemia (AML) and prostate cancer (PC). Most recently, several studies have revealed that PURα is dysregulated in multiple cancers, such as breast cancer (BC) and esophageal squamous cell carcinoma (ESCC). The oncogenic or tumor-suppressive functions of PURα are realized via regulating RNA/protein interaction, mRNA translation, formation of stress granules (SGs), and transcriptional regulation of several oncogenes and tumor suppressors. Although DNA/RNA binding proteins are hardly targeted, novel strategies have been applied to identify compounds targeting PURα and have demonstrated promising anti-tumor efficacy in the preclinical study. The present review summarizes the most recently discovered critical roles of PURα in various cancer types, providing an overview of the biomarker and therapeutic target potential of PURα for patients with cancer.
Collapse
Affiliation(s)
- Shiyi Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Chengyang Jiang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yawen Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fei Cheng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fangchen Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Chang Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xue Gong
- Nanjing Women and Children's Healthcare Hospital, Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University, 123 Tianfei Alley, Mochou Road, Nanjing, China.
| |
Collapse
|
45
|
Zhang X, Zhang M, Sun H, Wang X, Wang X, Sheng W, Xu M. The role of transcription factors in the crosstalk between cancer-associated fibroblasts and tumor cells. J Adv Res 2025; 67:121-132. [PMID: 38309692 PMCID: PMC11725164 DOI: 10.1016/j.jare.2024.01.033] [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: 11/29/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Transcription factors (TFs) fulfill a critical role in the formation and maintenance of different cell types during the developmental process as well as disease. It is believed that cancer-associated fibroblasts (CAFs) are activation status of tissue-resident fibroblasts or derived from form other cell types via transdifferentiation or dedifferentiation. Despite a subgroup of CAFs exhibit anti-cancer effects, most of them are reported to exert effects on tumor progression, further indicating their heterogeneous origin. AIM OF REVIEW This review aimed to summarize and review the roles of TFs in the reciprocal crosstalk between CAFs and tumor cells, discuss the emerging mechanisms, and their roles in cell-fate decision, cellular reprogramming and advancing our understanding of the gene regulatory networks over the period of cancer initiation and progression. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript delves into the key contributory factors of TFs that are involved in activating CAFs and maintaining their unique states. Additionally, it explores how TFs play a pivotal and multifaceted role in the reciprocal crosstalk between CAFs and tumor cells. This includes their involvement in processes such as epithelial-mesenchymal transition (EMT), proliferation, invasion, and metastasis, as well as metabolic reprogramming. TFs also have a role in constructing an immunosuppressive microenvironment, inducing resistance to radiation and chemotherapy, facilitating angiogenesis, and even 'educating' CAFs to support the malignancies of tumor cells. Furthermore, this manuscript delves into the current status of TF-targeted therapy and considers the future directions of TFs in conjunction with anti-CAFs therapies to address the challenges in clinical cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xu Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xin Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| |
Collapse
|
46
|
Wang WJ, Xin ZY, Liu D, Liu Q, Liu Y, Qiu Z, Zhang J, Alam P, Cai XM, Zhao Z, Tang BZ. Intracellularly manipulable aggregation of the aggregation-induced emission luminogens. Biosens Bioelectron 2025; 267:116800. [PMID: 39341072 DOI: 10.1016/j.bios.2024.116800] [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: 04/29/2024] [Revised: 08/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
Biophotonics has seen significant advancements with the development of optical imaging techniques facilitating the noninvasive detection of biologically relevant species. Aggregation-induced emission (AIE) materials have emerged as a novel class of luminogens exhibiting enhanced luminescence or photodynamic efficiency in the aggregated state, making them ideal for biomedical applications. The intracellularly controlled aggregation of aggregate-induced emission luminogens (AIEgens) enables high-resolution imaging of intracellular targets and diagnosis of related diseases, and enables disease therapy by exploiting the novel properties of aggregates. This review provides an in-depth analysis of the strategies employed to modulate the aggregation of AIEgens, focusing on the importance of molecular modifications to improve hydrophilicity and achieve precise control over the intercellular aggregation of AIEgens. Furthermore, the representative applications of AIEgens in bioimaging, such as enzyme activity monitoring, protein tracking, organelle function monitoring, and in vivo tumor-specific therapeutics, are reviewed. Additionally, we outline the challenges and future opportunities for AIE research, emphasizing the importance of the strategies for realizing the precisely controllable aggregation of AIEgens inside cells and the need for extending AIEgens' absorption and emission wavelengths. This review aims to elucidate the rational development of responsive AIEgens for advanced biomedical applications.
Collapse
Affiliation(s)
- Wen-Jin Wang
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, School of Science and Engineering Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Zhuo-Yang Xin
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, School of Science and Engineering Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Dan Liu
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, School of Science and Engineering Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Yong Liu
- AIE Institute, Guangzhou 510530, China.
| | - Zijie Qiu
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, School of Science and Engineering Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Jianquan Zhang
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, School of Science and Engineering Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Parvej Alam
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, School of Science and Engineering Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Xu-Min Cai
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials College of Chemical Engineering, Nanjing Forestry University, China.
| | - Zheng Zhao
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, School of Science and Engineering Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China.
| | - Ben Zhong Tang
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, School of Science and Engineering Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China; Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Institute of Molecular Functional Materials, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, China.
| |
Collapse
|
47
|
Atta H, Kassem DH, Kamal MM, Hamdy NM. Harnessing the ubiquitin proteasome system as a key player in stem cell biology. Biofactors 2025; 51:e2157. [PMID: 39843166 DOI: 10.1002/biof.2157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025]
Abstract
Intracellular proteins take part in almost every body function; thus, protein homeostasis is of utmost importance. The ubiquitin proteasome system (UPS) has a fundamental role in protein homeostasis. Its main role is to selectively eradicate impaired or misfolded proteins, thus halting any damage that could arise from the accumulation of these malfunctioning proteins. Proteasomes have a critical role in controlling protein homeostasis in all cell types, including stem cells. We will discuss the role of UPS enzymes as well as the 26S proteasome complex in stem cell biology from several angles. First, we shall overview common trends of proteasomal activity and gene expression of different proteasomal subunits and UPS enzymes upon passaging and differentiation of stem cells toward various cell lineages. Second, we shall explore the effect of modulating proteasomal activity in stem cells and navigate through the interrelation between proteasomes' activity and various proteasome-related transcription factors. Third, we will shed light on curated microRNAs and long non-coding RNAs using various bioinformatics tools that might have a possible role in regulating UPS in stem cells and possibly, upon manipulation, can enhance the differentiation process into different lineages and/or delay senescence upon cell passaging. This will help to decipher the role played by individual UPS enzymes and subunits as well as various interrelated molecular mediators in stem cells' maintenance and/or differentiation and open new avenues in stem cell research. This can ultimately provide a leap toward developing novel therapeutic interventions related to proteasome dysregulation.
Collapse
Affiliation(s)
- Hind Atta
- School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, Cairo, Egypt
| | - Dina H Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed M Kamal
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Drug Research and Development Group, Health Research Center of Excellence, The British University in Egypt, Cairo, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
48
|
Lu X, Jin J, Wu Y, Lin J, Zhang X, Lu S, Zhang J, Zhang C, Ren M, Chen H, Zhang W, Luan X. Self-assembled PROTACs enable protein degradation to reprogram the tumor microenvironment for synergistically enhanced colorectal cancer immunotherapy. Bioact Mater 2025; 43:255-272. [PMID: 39386219 PMCID: PMC11461841 DOI: 10.1016/j.bioactmat.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/21/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Both β-catenin and STAT3 drive colorectal cancer (CRC) growth, progression, and immune evasion, and their co-overexpression is strongly associated with a poor prognosis. However, current small molecule inhibitors have limited efficacy due to the reciprocal feedback activation between STAT3 and β-catenin. Inspired by the PROteolysis TArgeting Chimera (PROTAC), a promising pharmacological modality for the selective degradation of proteins, we developed a strategy of nanoengineered peptide PROTACs (NP-PROTACs) to degrade both β-catenin and STAT3 effectively. The NP-PROTACs were engineered by coupling the peptide PROTACs with DSPE-PEG via disulfide bonds and self-assembled into nanoparticles. Notably, the dual degradation of β-catenin and STAT3 mediated by NP-PROTACs led to a synergistic antitumor effect compared to single-target treatment. Moreover, NP-PROTACs treatment enhanced CD103+ dendritic cell infiltration and T-cell cytotoxicity, alleviating the immunosuppressive microenvironment induced by β-catenin/STAT3 in CRC. These results highlight the potential of NP-PROTACs in facilitating the simultaneous degradation of two pathogenic proteins, thereby providing a novel avenue for cancer therapy.
Collapse
Affiliation(s)
- Xinchen Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jinmei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ye Wu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jiayi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaokun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shengxin Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jiyuan Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Chunling Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Maomao Ren
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weidong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100700, China
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
49
|
Hassan MM, Li YD, Ma MW, Teng M, Byun WS, Puvar K, Lumpkin R, Sandoval B, Rutter JC, Jin CY, Wang MY, Xu S, Schmoker AM, Cheong H, Groendyke BJ, Qi J, Fischer ES, Ebert BL, Gray NS. Exploration of the tunability of BRD4 degradation by DCAF16 trans-labelling covalent glues. Eur J Med Chem 2024; 279:116904. [PMID: 39341093 PMCID: PMC11960843 DOI: 10.1016/j.ejmech.2024.116904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 09/30/2024]
Abstract
Chemically induced proximity modalities such as targeted protein degradation (TPD) hold promise for expanding the number of proteins that can be manipulated pharmacologically. However, current TPD strategies are often limited to proteins with preexisting ligands. Molecular glues (e.g. glutarimide ligands for CUL4CRBN), offer the potential to target undruggable proteins. Yet, their rational design is largely unattainable due to the unpredictability of the 'gain-of-function' nature of the glue interaction upon chemical modification of ligands. We recently reported a covalent trans-labelling glue mechanism which we named 'Template-assisted covalent modification', where an electrophile decorated BRD4 inhibitor was effectively delivered to a cysteine residue on DCAF16 due to an electrophile-induced BRD4-DCAF16 interaction. Herein, we report our efforts to evaluate how various electrophilic modifications to the BRD4 binder, JQ1, affect DCAF16 recruitment and subsequent BRD4 degradation efficiency. We discovered a moderate correlation between the electrophile-induced BRD4-DCAF16 ternary complex formation and BRD4 degradation. Moreover, we show that a more solvent-exposed warhead presentation optimally recruits DCAF16 and promotes BRD4 degradation. The diversity of covalent attachments in this class of BRD4 degraders suggests a high tolerance and tunability for the BRD4-DCAF16 interaction. This offers a new avenue for rational glue design by introducing covalent warheads to known binders.
Collapse
Affiliation(s)
- Muhammad Murtaza Hassan
- Department of Chemical and Systems Biology, ChEM-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA; SPARK Translational Research Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Yen-Der Li
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michelle W Ma
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Mingxing Teng
- Center for Drug Discovery, Department of Pathology & Immunology, and Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Woong Sub Byun
- Department of Chemical and Systems Biology, ChEM-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Kedar Puvar
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Ryan Lumpkin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Brittany Sandoval
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Justine C Rutter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Cyrus Y Jin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Michelle Y Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shawn Xu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anna M Schmoker
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Hakyung Cheong
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Brian J Groendyke
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston, MA, USA.
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, ChEM-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
50
|
Viennet T, Yin M, Jayaraj A, Kim W, Sun ZYJ, Fujiwara Y, Zhang K, Seruggia D, Seo HS, Dhe-Paganon S, Orkin SH, Arthanari H. Structural insights into the DNA-binding mechanism of BCL11A: The integral role of ZnF6. Structure 2024; 32:2276-2286.e4. [PMID: 39423807 PMCID: PMC11625000 DOI: 10.1016/j.str.2024.09.022] [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: 01/17/2024] [Revised: 05/17/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024]
Abstract
The transcription factor BCL11A is a critical regulator of the switch from fetal hemoglobin (HbF: α2γ2) to adult hemoglobin (HbA: α2β2) during development. BCL11A binds at a cognate recognition site (TGACCA) in the γ-globin gene promoter and represses its expression. DNA-binding is mediated by a triple zinc finger domain, designated ZnF456. Here, we report comprehensive investigation of ZnF456, leveraging X-ray crystallography and NMR to determine the structures in both the presence and absence of DNA. We delve into the dynamics and mode of interaction with DNA. Moreover, we discovered that the last zinc finger of BCL11A (ZnF6) plays a different role compared to ZnF4 and 5, providing a positive entropic contribution to DNA binding and γ-globin gene repression. Comprehending the DNA binding mechanism of BCL11A opens avenues for the strategic, structure-based design of novel therapeutics targeting sickle cell disease and β-thalassemia.
Collapse
Affiliation(s)
- Thibault Viennet
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Maolu Yin
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Abhilash Jayaraj
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Woojin Kim
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Zhen-Yu J Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yuko Fujiwara
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Kevin Zhang
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Davide Seruggia
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA; St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Stuart H Orkin
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Haribabu Arthanari
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|