1
|
Yim H, Sun R, Xu Z, Kim HS, Kim M, Cao T, Xie L, Chen X, Kaniskan HÜ, Jin J. Discovery of the first-in-class DOT1L PROTAC degrader. Eur J Med Chem 2025; 291:117595. [PMID: 40186895 PMCID: PMC12045715 DOI: 10.1016/j.ejmech.2025.117595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
DOT1L is the lysine methyltransferase responsible for histone H3 lysine 79 (H3K79) methylation and plays a crucial role in leukemia progression. Furthermore, DOT1L has biological functions that are independent of its methyltransferase activity. Therefore, targeting and degrading DOT1L with PROteolysis TArgeting Chimeras (PROTACs) could represent a promising therapeutic strategy. Here, we report the discovery of the first-in-class DOT1L PROTAC degrader, compound 13 (MS2133), which potently induces DOT1L degradation in a concentration- and time-dependent manner, without affecting DOT1L mRNA expression. The DOT1L degradation induced by 13 requires binding to the E3 ligase von Hippel-Lindau (VHL) and DOT1L and occurs through the ubiquitin-proteasome system. 13 is selective for DOT1L over other methyltransferases and effectively inhibits the growth of mixed lineage leukemia-rearranged (MLL-r) leukemia cells while having no toxicity on normal cells. Overall, 13 is a valuable chemical biology tool for further studying functions of DOT1L and a potential therapeutic for DOT1L-dependent cancers.
Collapse
Affiliation(s)
- Hyerin Yim
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Renhong Sun
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Zhongli Xu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Huen Suk Kim
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Minjeong Kim
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Tao Cao
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - H Ümit Kaniskan
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States.
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Science, Oncological Science and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States.
| |
Collapse
|
2
|
Nozaki T, Onoda M, Habazaki M, Takeuchi Y, Ishida H, Sato Y, Kujirai T, Hanada K, Yamatsugu K, Kurumizaka H, Kimura H, Kono H, Kawashima SA, Kanai M. Designer Catalyst-Enabled Regiodivergent Histone Acetylation. J Am Chem Soc 2025; 147:13732-13743. [PMID: 40223209 DOI: 10.1021/jacs.5c01699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
The "histone code," defined by the combinatorial patterns of post-translational modifications (PTMs) on histones, plays a pivotal role in chromatin structure and gene expression. Tools for the regioselective introduction of histone PTMs in living cells are critical for dissecting the functions of these epigenetic marks. Here, we report the design and development of three regioselective catalysts that acetylate distinct lysine residues (K43, K108, and K120) on histone H2B. Using a combination of molecular dynamics simulations of catalyst-nucleosome complexes and systematic experimental optimization of catalyst structures, we identified key design principles for achieving regioselectivity. Specifically, excluding highly reactive off-target lysine residues from the catalyst effective region (CER) while maintaining proximity to a target lysine residue proved crucial. Biochemical and cellular analyses of the catalytic histone acetylation revealed that each lysine acetylation elicited unique effects on the binding affinity and activity of nucleosome-interacting molecules, as well as on transcriptional programs and cellular phenotypes. These findings establish a framework for designing regioselective histone acetylation catalysts and advance our understanding of the regulatory mechanisms underlying histone PTMs.
Collapse
Affiliation(s)
- Tamiko Nozaki
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Mayu Onoda
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Misuzu Habazaki
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuma Takeuchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hisashi Ishida
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Yuko Sato
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Tomoya Kujirai
- Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Kayo Hanada
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kenzo Yamatsugu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hitoshi Kurumizaka
- Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Hiroshi Kimura
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Hidetoshi Kono
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Center of Quantum Life Science for Structural Therapeutics (cQUEST), Chiba University, Chiba 263-8522, Japan
| | - Shigehiro A Kawashima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
3
|
Wang Y, Yu A, Gao Z, Yuan X, Du X, Shi P, Guan H, Wen S, Wang H, Wang L, Fan B, Liu Z. TET2 gene mutation status associated with poor prognosis of transition zone prostate cancer: a retrospective cohort study based on whole exome sequencing and machine learning models. Front Endocrinol (Lausanne) 2025; 16:1568665. [PMID: 40297177 PMCID: PMC12034557 DOI: 10.3389/fendo.2025.1568665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Prostate cancer (PCa) in the transition zone (TZ) is uncommon and often poses challenges for early diagnosis, but its genomic determinants and therapeutic vulnerabilities remain poorly characterized. Methods Tumor mutational landscape was characterized in nine patients with TZ PCa, identifying somatic variants through whole-exome sequencing (WES). Novel candidate variants relevant to driver gene were selected using rare-variant burden analysis. Kaplan-Meier curves with log-rank testing and Cox regression models were applied to evaluate the prognostic significance of selected mutant driver gene and clinicopathological characteristics in a cohort of 132 patients with TZ PCa. Significant prognostic determinants were integrated into a validated nomogram for individualized prediction of 3-, 4-, and 5-year biochemical recurrence-free survival (BRFS) and overall survival (OS) probabilities. Eight machine learning algorithms were employed to develop BRFS and OS prediction models in a cohort. Results A total of 5,036 somatic single nucleotide variants (SNVs) and 587 somatic insertion and deletion (INDELs) were discovered. Among eight driver gene mutations which were verified through Sanger sequencing, TET2 gene, with high mutation frequency and potential targeted drug relevance (bromodomain inhibitors and DOT1L inhibitors) was selected for further validation. Retrospective cohort study demonstrated that TET2 mutant status was significantly associated with Gleason score (p = 0.004) and distant metastasis (p = 0.002). Furthermore, TET2 mutant status was significantly correlated with inferior BRFS and OS and served as an independent predictor. Comparative evaluation of eight algorithms revealed the GBM model achieved superior discriminative ability for BRFS (AUC for 3-year: 0.752, 4-year: 0.786, 5-year: 0.796). The predictive model based on the GBM machine learning algorithm achieved the best predictive performance for OS (AUC for 3-year: 0.838, 4-year: 0.915, 5-year: 0.868). The constructed predictive nomogram provided evidence that TET2 mutant status integration conferred statistically significant improvements in model accuracy and clinical predictive value. Conclusion Our study elucidated the distinct genetic basis of prostate cancer in the transition zone and identified TET2 mutation as an independent prognostic determinant in TZ PCa. However, the limited sample size of this study necessitates cautious interpretation of these findings, and further validation in larger cohorts is warranted to confirm their generalizability.
Collapse
Affiliation(s)
- Yutong Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Colorectal Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China
| | - Ailing Yu
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- First Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Ziping Gao
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoying Yuan
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaochen Du
- College of Humanities and Social Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Peng Shi
- Second Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Haoyun Guan
- Second Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Shuang Wen
- Department of Pathology, Dalian Friendship Hospital, Dalian, China
| | - Honglong Wang
- Department of Pathology, Dalian Friendship Hospital, Dalian, China
| | - Liang Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bo Fan
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhiyu Liu
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
4
|
Becht DC, Selvam K, Lachance C, Côté V, Li K, Nguyen MC, Pareek A, Shi X, Wen H, Blanco MA, Côté J, Kutateladze TG. A multivalent engagement of ENL with MOZ. Nat Struct Mol Biol 2025; 32:709-718. [PMID: 39794553 DOI: 10.1038/s41594-024-01455-8] [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: 02/26/2024] [Accepted: 11/19/2024] [Indexed: 01/13/2025]
Abstract
The epigenetic cofactor ENL (eleven-nineteen-leukemia) and the acetyltransferase MOZ (monocytic leukemia zinc finger) have vital roles in transcriptional regulation and are implicated in aggressive forms of leukemia. Here, we describe the mechanistic basis for the intertwined association of ENL and MOZ. Genomic analysis shows that ENL and MOZ co-occupy active promoters and that MOZ recruits ENL to its gene targets. Structural studies reveal a multivalent assembly of ENL at the intrinsically disordered region (IDR) of MOZ. While the extraterminal (ET) domain of ENL recognizes the canonical ET-binding motif in IDR, the YEATS domains of ENL and homologous AF9 bind to a set of acetylation sites in the MOZ IDR that are generated by the acetyltransferase CBP (CREB-binding protein). Our findings suggest a multifaceted acetylation-dependent and independent coupling of ENL, MOZ and CBP/p300, which may contribute to leukemogenic activities of the ENL-MOZ assembly and chromosomal translocations of ENL, MOZ and CBP/p300.
Collapse
Affiliation(s)
- Dustin C Becht
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Karthik Selvam
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Catherine Lachance
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Québec City, Québec, Canada
| | - Valérie Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Québec City, Québec, Canada
| | - Kuai Li
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Minh Chau Nguyen
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Akshay Pareek
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Hong Wen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - M Andres Blanco
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA, USA
| | - Jacques Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Québec City, Québec, Canada
| | - Tatiana G Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
5
|
Tang L, Lu M, Du Y, Sun J. Pbx3-mediated suppression of type I interferon response contributes to leukemia progression driven by MLL-AF9. Cancer Gene Ther 2025; 32:475-485. [PMID: 40108441 DOI: 10.1038/s41417-025-00888-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/13/2025] [Accepted: 03/06/2025] [Indexed: 03/22/2025]
Abstract
Cell-intrinsic repression of inflammatory signaling supports the survival of acute myeloid leukemia blasts. However, how the cell-intrinsic inflammation status changes during AML progression remains elusive. Here, we used CRISPR-mediated genome editing to create a murine AML model driven by a chromosomal translocation between the mixed-lineage leukemia (Mll) gene and the Mllt3/Af9 gene. The resulting MLL-AF9 (MA9) fusion protein is sufficient to immortalize hematopoietic stem and progenitor cells (HSPCs) in vitro but insufficient to induce an overt leukemia phenotype in vivo rapidly. Leukemia progression in vivo is associated with a downregulation of type I interferon response genes, and this process depends on the upregulation of MA9 transcriptional target Pbx3 in the progenitor cell compartment. Accordingly, enhancing interferon response by interferon-α (IFNα) administration induces leukemic cell differentiation, and inhibiting MA9 transcriptional activity on top of the enhanced IFN signaling further delays leukemia progression. Our study underscores the importance of Pbx3-mediated suppression of interferon response genes in the progression of MA9-induced AML and highlights the potential application of type I interferon for its treatment.
Collapse
Affiliation(s)
- Li Tang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Meng Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yulong Du
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jianlong Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
6
|
Dong J, Konopleva M. Preclinical targeting of leukemia-initiating cells in the development future biologics for acute myeloid leukemia. Expert Opin Ther Targets 2025; 29:223-237. [PMID: 40304258 DOI: 10.1080/14728222.2025.2500417] [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: 11/16/2024] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Leukemia-initiating cells (LICs) are a critical subset of cells driving acute myeloid leukemia (AML) relapse and resistance to therapy. They possess unique properties, including metabolic, epigenetic, and microenvironmental dependencies, making them promising therapeutic targets. AREAS COVERED This review summarizes preclinical advances in targeting AML LICs, including strategies to exploit metabolic vulnerabilities, such as the reliance on oxidative phosphorylation (OXPHOS), through the use of mitochondrial inhibitors; target epigenetic regulators like DOT1L (Disruptor of Telomeric Silencing 1-like) to disrupt LIC survival mechanisms; develop immunotherapies, including CAR (chimeric antigen receptor) T-cell therapy, and bispecific antibodies; and disrupt LIC interactions with the bone marrow microenvironment by inhibiting supportive niches. EXPERT OPINION LIC-targeted therapies hold significant promise for revolutionizing AML treatment by reducing relapse rates and improving long-term outcomes. However, challenges such as LIC heterogeneity, therapy resistance, and associated toxicity persist. Recent studies have illuminated the distinct biological characteristics of LICs, advancing our understanding of their behavior and vulnerabilities. These insights offer new opportunities to target LICs at earlier disease stages and to explore combination therapies with other targeted treatments, ultimately enhancing therapeutic efficacy and improving patient outcomes.
Collapse
Affiliation(s)
- Jiaxin Dong
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marina Konopleva
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
7
|
Shi X, Li M, Liu Z, Tiessen J, Li Y, Zhou J, Zhu Y, Mahesula S, Ding Q, Tan L, Feng M, Kageyama Y, Hara Y, Tao JJ, Luo X, Patras KA, Lorenzi PL, Huang S, Stevens AM, Takahashi K, Issa GC, Samee MAH, Agathocleous M, Nakada D. Guanine nucleotide biosynthesis blockade impairs MLL complex formation and sensitizes leukemias to menin inhibition. Nat Commun 2025; 16:2641. [PMID: 40102405 PMCID: PMC11920272 DOI: 10.1038/s41467-025-57544-9] [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: 06/13/2024] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
Targeting the dependency of MLL-rearranged (MLLr) leukemias on menin with small molecule inhibitors has opened new therapeutic strategies for these poor-prognosis diseases. However, the rapid development of menin inhibitor resistance calls for combinatory strategies to improve responses and prevent resistance. Here we show that leukemia stem cells (LSCs) of MLLr acute myeloid leukemia (AML) exhibit enhanced guanine nucleotide biosynthesis, the inhibition of which leads to myeloid differentiation and sensitization to menin inhibitors. Mechanistically, targeting inosine monophosphate dehydrogenase 2 (IMPDH2) reduces guanine nucleotides and rRNA transcription, leading to reduced protein expression of LEDGF and menin. Consequently, the formation and chromatin binding of the MLL-fusion complex is impaired, reducing the expression of MLL target genes. Inhibition of guanine nucleotide biosynthesis or rRNA transcription further suppresses MLLr AML when combined with a menin inhibitor. Our findings underscore the requirement of guanine nucleotide biosynthesis in maintaining the function of the LEDGF/menin/MLL-fusion complex and provide a rationale to target guanine nucleotide biosynthesis to sensitize MLLr leukemias to menin inhibitors.
Collapse
MESH Headings
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/genetics
- Humans
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Myeloid-Lymphoid Leukemia Protein/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Histone-Lysine N-Methyltransferase/metabolism
- Histone-Lysine N-Methyltransferase/genetics
- Animals
- Mice
- Cell Line, Tumor
- IMP Dehydrogenase/metabolism
- IMP Dehydrogenase/antagonists & inhibitors
- IMP Dehydrogenase/genetics
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/drug effects
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Transcription Factors/metabolism
- Transcription Factors/genetics
Collapse
Affiliation(s)
- Xiangguo Shi
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Department of Molecular and Precision Medicine, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Minhua Li
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zian Liu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jonathan Tiessen
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuan Li
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jing Zhou
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yudan Zhu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Swetha Mahesula
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Qing Ding
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mengdie Feng
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuki Kageyama
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yusuke Hara
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jacob J Tao
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xuan Luo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Kathryn A Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Suming Huang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Alexandra M Stevens
- Section of Hematology/Oncology, Department of Pediatrics, Texas Children's Cancer and Hematology Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Michalis Agathocleous
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Daisuke Nakada
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Xue Z, Xuan H, Lau K, Su Y, Wegener M, Li K, Turner L, Adams M, Shi X, Wen H. Expression of ENL YEATS domain tumor mutations in nephrogenic or stromal lineage impairs kidney development. Nat Commun 2025; 16:2531. [PMID: 40087269 PMCID: PMC11909213 DOI: 10.1038/s41467-025-57926-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 03/05/2025] [Indexed: 03/17/2025] Open
Abstract
Recurrent gain-of-function mutations in the histone reader protein ENL have been identified in Wilms tumor, the most prevalent pediatric kidney cancer. However, their pathological significance in kidney development and tumorigenesis in vivo remains elusive. Here, we generate mouse models mimicking ENL tumor (ENLT) mutations and show that heterozygous mutant expression in Six2+ nephrogenic or Foxd1+ stromal lineages leads to severe, lineage-specific kidney defects, both resulting in neonatal lethality. Six2-ENLT mutant kidneys display compromised cap mesenchyme, scant nephron tubules, and cystic glomeruli, indicative of premature progenitor commitment and blocked differentiation. Bulk and spatial transcriptomic analyses reveal aberrant activation of Hox and Wnt signaling genes in mutant nephrogenic cells. In contrast, Foxd1-ENLT mutant kidneys exhibit expansion in renal capsule and cap mesenchyme, with dysregulated stromal gene expression affecting stroma-epithelium crosstalk. Our findings uncover distinct pathways through which ENL mutations disrupt nephrogenesis, providing a foundation for further investigations into their role in tumorigenesis.
Collapse
Affiliation(s)
- Zhaoyu Xue
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Hongwen Xuan
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Kin Lau
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Yangzhou Su
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Marc Wegener
- Genomics Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Kuai Li
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Lisa Turner
- Pathology Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Marie Adams
- Genomics Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Hong Wen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
9
|
Mendoza-Castrejon J, Yang W, Denby E, Wang H, Casey EB, Muthukumar R, Patel RM, Yoon J, Li Y, White JM, Chen R, Batista LFZ, Magee JA. Fetal context conveys heritable protection against MLL-rearranged leukemia that depends on MLL3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642680. [PMID: 40161712 PMCID: PMC11952435 DOI: 10.1101/2025.03.11.642680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
MLL rearrangements ( MLL r) are the most common cause of congenital and infant leukemias. MLL r arise prior to birth and require few cooperating mutations for transformation, yet congenital leukemias are 10-fold less common than infant leukemias and >100-fold less common than childhood leukemias overall. This raises the question of whether mechanisms exist to suppress leukemic transformation during fetal life, thereby protecting the developing fetus from malignancy during a period of rapid hematopoietic progenitor expansion. Here, we use mouse models to show that fetal MLL::ENL exposure creates a heritable, leukemia-resistant state. MLL::ENL imposes a negative selective pressure on fetal hematopoietic progenitors. It leads to postnatal loss of self-renewal gene expression and enhanced myeloid differentiation that precludes transformation. These changes do not occur when MLL::ENL is induced shortly after birth, and transformation proceeds efficiently in this context. The fetal barrier to transformation is enforced by the histone methyltransferase MLL3. It can be overcome by cooperating mutations, such as Nras G12D , or through somatic or germline inactivation of MLL3. Heritable fetal protection against leukemic transformation may explain the low incidence of congenital leukemias in humans despite prenatal MLL rearrangement.
Collapse
|
10
|
Yang Y, Cathelin S, Liu ACH, Subedi A, Maher A, Hosseini M, Manikoth Ayyathan D, Vanner R, Chan SM. TET2 deficiency increases the competitive advantage of hematopoietic stem and progenitor cells through upregulation of thrombopoietin receptor signaling. Nat Commun 2025; 16:2384. [PMID: 40064887 PMCID: PMC11894142 DOI: 10.1038/s41467-025-57614-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Ten-Eleven Translocation-2 (TET2) mutations drive the expansion of mutant hematopoietic stem cells (HSCs) in clonal hematopoiesis (CH). However, the precise mechanisms by which TET2 mutations confer a competitive advantage to HSCs remain unclear. Here, through an epigenetic drug screen, we discover that inhibition of disruptor of telomeric silencing 1-like (DOT1L), a H3K79 methyltransferase, selectively reduces the fitness of Tet2 knockout (Tet2KO) hematopoietic stem and progenitor cells (HSPCs). Mechanistically, we find that TET2 deficiency increases H3K79 dimethylation and expression of Mpl, which encodes the thrombopoietin receptor (TPO-R). Correspondingly, TET2 deficiency is associated with a higher proportion of primitive Mpl-expressing (Mpl+) cells in the HSC compartment. Importantly, inhibition of Mpl expression or the signaling downstream of TPO-R is sufficient to reduce the competitive advantage of murine and human TET2-deficient HSPCs. Our findings demonstrate a critical role for aberrant TPO-R signaling in TET2 mutation-driven CH and uncover potential therapeutic strategies against this condition.
Collapse
Affiliation(s)
- Yitong Yang
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Severine Cathelin
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Alex C H Liu
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Amit Subedi
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Abdula Maher
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, M5S 1A8, Canada
| | - Mohsen Hosseini
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | | | - Robert Vanner
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Steven M Chan
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada.
| |
Collapse
|
11
|
Sturgeon CM, Wagenblast E, Izzo F, Papapetrou EP. The Crossroads of Clonal Evolution, Differentiation Hierarchy, and Ontogeny in Leukemia Development. Blood Cancer Discov 2025; 6:94-109. [PMID: 39652739 PMCID: PMC11876951 DOI: 10.1158/2643-3230.bcd-24-0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/19/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
SIGNIFICANCE In recent years, remarkable technological advances have illuminated aspects of the pathogenesis of myeloid malignancies-yet outcomes for patients with these devastating diseases have not significantly improved. We posit that a synthesized view of the three dimensions through which hematopoietic cells transit during their healthy and diseased life-clonal evolution, stem cell hierarchy, and ontogeny-promises high yields in new insights into disease pathogenesis and new therapeutic avenues.
Collapse
Affiliation(s)
- Christopher M. Sturgeon
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elvin Wagenblast
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pediatrics, Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Franco Izzo
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eirini P. Papapetrou
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
12
|
Schurer A, Glushakow-Smith SG, Gritsman K. Targeting chromatin modifying complexes in acute myeloid leukemia. Stem Cells Transl Med 2025; 14:szae089. [PMID: 39607901 PMCID: PMC11878770 DOI: 10.1093/stcltm/szae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/22/2024] [Indexed: 11/30/2024] Open
Abstract
Acute myeloid leukemia (AML) is a devastating hematologic malignancy with high rates of relapse, which can, in part, be attributed to the dysregulation of chromatin modifications. These epigenetic modifications can affect the capacity of hematopoietic cells to self-renew or differentiate, which can lead to transformation. Aberrant histone modifications contribute to the derepression of self-renewal genes such as HOXA/B and MEIS1 in committed hematopoietic progenitors, which is considered a key mechanism of leukemogenesis in MLL-rearranged (MLL-r) and NPM1-mutated AML. As regulators of some of the key histone modifications in this disease, the menin-KMT2A and polycomb repressive (PRC1/2) complexes have been identified as promising targets for the treatment of AML. This review explores recent discoveries of how leukemic cells hijack these complexes and their interactions with other chromatin regulators to promote disease progression. We also discuss inhibitors targeting these complexes that have demonstrated therapeutic efficacy in preclinical and clinical studies and propose novel therapeutic combinations targeting the KMT2A and PRC1/2 broader interacting networks to overcome issues of resistance to existing monotherapies.
Collapse
Affiliation(s)
- Alexandra Schurer
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
- The Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Shira G Glushakow-Smith
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
- The Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Kira Gritsman
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
- The Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, United States
- Department of Medical Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, United States
- Center for Tumor Dormancy, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461,United States
- Marilyn and Stanley M. Katz Institute for Immunotherapy for Cancer and Inflammatory Disorders, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| |
Collapse
|
13
|
Bret C, Desmots-Loyer F, Moreaux J, Fest T. BHLHE41, a transcriptional repressor involved in physiological processes and tumor development. Cell Oncol (Dordr) 2025; 48:43-66. [PMID: 39254779 PMCID: PMC11850569 DOI: 10.1007/s13402-024-00973-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2024] [Indexed: 09/11/2024] Open
Abstract
BHLHE41 is a nuclear transcriptional repressor that belongs to the basic helix-loop-helix protein superfamily. BHLHE41 expression tends to be restricted to specific tissues and is regulated by environmental cues and biological events. BHLHE41 homodimerizes or heterodimerizes with various partners, influencing its transcription factor function. BHLHE41 is involved in the regulation of many physiological processes implicated in tissue/organ homeostasis, such as myogenesis, adipogenesis, circadian rhythms and DNA repair. At cellular level, BHLHE41 is involved in the regulation of mesenchymal stem cell properties, tissue-specific macrophage functions and lymphoid lineage physiology. In several cancer types, BHLHE41 modulates the expression of different transcriptional programs influencing cell cycle control, apoptosis, invasiveness, epithelial to mesenchymal transition and hypoxia response in the tumor environment. Depending on the cancer cell type, BHLHE41 can act as a tumor suppressor or an oncogene, and could be a target for innovative therapies. This review summarizes the available knowledge on BHLHE41 structure, biological functions, regulation and potential partners, as well as its role in physiological processes, and its implication in major cancer steps.
Collapse
Affiliation(s)
- Caroline Bret
- Department of Biological Hematology, CHU Montpellier, Montpellier, 34295, France.
- Faculty of Medicine of Montpellier and Nîmes, University of Montpellier, Montpellier, 34090, France.
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, 34396, France.
| | - Fabienne Desmots-Loyer
- UMR 1236, University of Rennes 1, INSERM, Établissement Français du Sang Bretagne, Rennes, France.
- Pôle de Biologie, Rennes University Medical Center, Rennes, France.
| | - Jérôme Moreaux
- Department of Biological Hematology, CHU Montpellier, Montpellier, 34295, France.
- Faculty of Medicine of Montpellier and Nîmes, University of Montpellier, Montpellier, 34090, France.
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, 34396, France.
- Institut Universitaire de France, Paris, France.
| | - Thierry Fest
- UMR 1236, University of Rennes 1, INSERM, Établissement Français du Sang Bretagne, Rennes, France.
- Pôle de Biologie, Rennes University Medical Center, Rennes, France.
| |
Collapse
|
14
|
Perner F, Gadrey JY, Armstrong SA, Kühn MWM. Targeting the Menin-KMT2A interaction in leukemia: Lessons learned and future directions. Int J Cancer 2025. [PMID: 39887730 DOI: 10.1002/ijc.35332] [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: 11/25/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 02/01/2025]
Abstract
Chromosomal rearrangements involving the Mixed Lineage Leukemia gene (MLL1, KMT2A) are defining a genetically distinct subset in about 10% of human acute leukemias. Translocations involving the KMT2A-locus at chromosome 11q23 are resulting in the formation of a chimeric oncogene, where the N-terminal part of KMT2A is fused to a variety of translocation partners. The most frequently found fusion partners of KMT2A in acute leukemia are the C-terminal parts of AFF1, MLLT3, MLLT1 and MLLT10. Unfortunately, the presence of an KMT2A-rearrangements is associated with adverse outcomes in leukemia patients. Moreover, non-rearranged KMT2A-complexes have been demonstrated to be crucial for disease development and maintenance in NPM1-mutated and NUP98-rearranged leukemia, expanding the spectrum of genetic disease subtypes that are dependent on KMT2A. Recent advances in the development of targeted therapy strategies to disrupt the function of KMT2A-complexes in leukemia have led to the establishment of Menin-KMT2A interaction inhibitors that effectively eradicate leukemia in preclinical model systems and show favorable tolerability and significant efficacy in early-phase clinical trials. Indeed, one Menin inhibitor, Revumenib, was recently approved for the treatment of patients with relapsed or refractory KMT2A-rearranged acute leukemia. However, single agent therapy can lead to resistance. In this Review article we summarize our current understanding about the biology of pathogenic KMT2A-complex function in cancer, specifically leukemia, and give a systematic overview of lessons learned from recent clinical and preclinical studies using Menin inhibitors.
Collapse
Affiliation(s)
- Florian Perner
- Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany
- DGHO, Deutsche Gesellschaft für Hämatologie und Medizinische Onkologie e.V. working group, Clinical and Translational Epigenetics, Berlin, Germany
| | - Jayant Y Gadrey
- Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Scott A Armstrong
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael W M Kühn
- DGHO, Deutsche Gesellschaft für Hämatologie und Medizinische Onkologie e.V. working group, Clinical and Translational Epigenetics, Berlin, Germany
- Department of Hematology and Medical Oncology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
15
|
Coulée M, de la Iglesia A, Blanco M, Gobé C, Lapoujade C, Ialy-Radio C, Alvarez-Gonzalez L, Meurice G, Ruiz-Herrera A, Fouchet P, Cocquet J, El Khattabi L. Chromatin environment-dependent effects of DOT1L on gene expression in male germ cells. Commun Biol 2025; 8:138. [PMID: 39875559 PMCID: PMC11775102 DOI: 10.1038/s42003-024-07393-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 12/09/2024] [Indexed: 01/30/2025] Open
Abstract
The H3K79 methyltransferase DOT1L is essential for multiple aspects of mammalian development where it has been shown to regulate gene expression. Here, by producing and integrating epigenomic and spike-in RNA-seq data, we decipher the molecular role of DOT1L during mouse spermatogenesis and show that it has opposite effects on gene expression depending on chromatin environment. On one hand, DOT1L represses autosomal genes that are devoid of H3K79me2 at their bodies and located in H3K27me3-rich/H3K27ac-poor environments. On the other hand, it activates the expression of genes enriched in H3K79me2 and located in H3K27me3-poor/H3K27ac-rich environments, predominantly X chromosome-linked genes, after meiosis I. This coincides with a significant increase in DOT1L expression at this stage and a genome-wide acquisition of H3K79me2, particularly on the sex chromosomes. Taken together, our results show that H3K79me2 positively correlates with male germ cell genetic program throughout spermatogenesis, with DOT1L predominantly inhibiting rather than activating gene expression. Interestingly, while DOT1L appears to directly regulate the (re)activation of X genes following meiotic sex chromosome inactivation, it also controls the timely expression of (autosomal) differentiation genes during spermatogenesis.
Collapse
Affiliation(s)
- Manon Coulée
- Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014, Paris, France
| | | | - Mélina Blanco
- Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014, Paris, France
- Université Paris Cité, CEA, Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, Paris, France
- Laboratoire des Cellules Souches Germinales, Université Paris-Saclay, CEA, Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, Paris, France
| | - Clara Gobé
- Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014, Paris, France
| | - Clémentine Lapoujade
- Université Paris Cité, CEA, Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, Paris, France
- Laboratoire des Cellules Souches Germinales, Université Paris-Saclay, CEA, Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, Paris, France
| | - Côme Ialy-Radio
- Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014, Paris, France
| | - Lucia Alvarez-Gonzalez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | | | - Aurora Ruiz-Herrera
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Pierre Fouchet
- Université Paris Cité, CEA, Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, Paris, France
- Laboratoire des Cellules Souches Germinales, Université Paris-Saclay, CEA, Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, Paris, France
| | - Julie Cocquet
- Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014, Paris, France.
| | - Laïla El Khattabi
- Université Paris Cité, CNRS, Inserm, Institut Cochin, F-75014, Paris, France.
- Sorbonne Université, APHP Hôpital Pitié-Salpêtrière, Paris Brain Institute-ICM, INSERM U1127, CNRS, UMR 7225, Paris, France.
| |
Collapse
|
16
|
Yusoff NA, Abd Hamid Z, Taib IS, Abdul Razak SR, Budin SB. Exploring Epigenetic Complexity in Regulation of Hematopoietic Stem Cells Niche: A Mechanistic Journey from Normal to Malignant Hematopoiesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 39841383 DOI: 10.1007/5584_2024_846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Epigenetic regulation in hematopoietic stem cells (HSCs) research has emerged as a transformative molecular approach that enhances understanding of hematopoiesis and hematological disorders. This chapter investigates the intricate epigenetic mechanisms that control HSCs function, including deoxyribonucleic acid (DNA) methylation, histone modifications, and chromatin remodeling. It also explores the role of non-coding ribonucleic acid (RNAs) as epigenetic regulators, highlighting how changes in gene expression can occur without alterations to the DNA sequence. Epigenetic mechanisms play a pivotal in regulating HSC self-renewal and differentiation, processes essential for maintaining a balanced hematopoietic system in which lineage-specific hematopoietic stem and progenitor cells (HSPCs) pool is sustained. Recent advancements in epigenetic mapping and sequencing technologies have illuminated the dynamic epigenetic landscapes that characterize HSCs and their progeny. Numerous studies have revealed that dysregulation of epigenetic pathways is a hallmark of various hematological malignancies, including leukemias, lymphomas, and myelodysplastic syndromes. This review highlights key findings that demonstrate the impact of epigenetic abnormalities on the disruption of HSPC niches and the progression of oncogenesis in hematological malignancies. Furthermore, this chapter explores the therapeutic potential of targeting epigenetic modifications that are critical in formation and progression of hematologic malignancies. It also discusses the latest developments in epigenetic therapies, including the use of DNA methyltransferase inhibitors, histone deacetylase inhibitors, and emerging drugs targeting other epigenetic regulators. These therapies represent a promising strategy for resetting aberrant epigenetic states, potentially restoring normal hematopoiesis. Conclusively, this chapter offers a thorough overview of the current landscape and future directions of epigenetic research related to the maintenance of the HSPC niches. The insights presented here aim to contribute significantly to the field, offering a reference point for molecular approaches that enhance our understanding of hematopoiesis and its associated hematological malignancies.
Collapse
Affiliation(s)
- Nur Afizah Yusoff
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Zariyantey Abd Hamid
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Izatus Shima Taib
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Siti Razila Abdul Razak
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Pulau Pinang, Malaysia
| | - Siti Balkis Budin
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
17
|
Heikamp EB, Armstrong SA. Revumenib Revises the Treatment Landscape for KMT2A-r Leukemia. J Clin Oncol 2025; 43:85-88. [PMID: 39509656 PMCID: PMC11771282 DOI: 10.1200/jco-24-01265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/02/2024] [Indexed: 11/15/2024] Open
Affiliation(s)
- Emily B. Heikamp
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA
| | - Scott A. Armstrong
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA
| |
Collapse
|
18
|
Gordon SJV, Perner F, MacPherson L, Wenge DV, Bourgeois W, Fennell K, Klaus T, Petrovic J, Horvath J, Cao J, Lapek J, Uryu S, White J, Lam EYN, Mu XJ, Chan YC, Gillespie A, Blyth B, Camerino MA, Bozikis YE, Holze H, Knezevic K, Balic J, Stupple PA, Street IP, Monahan BJ, Sharma S, Wainwright EN, Vassiliadis D, Paul TA, Armstrong SA, Dawson MA. Catalytic inhibition of KAT6/KAT7 enhances the efficacy and overcomes primary and acquired resistance to Menin inhibitors in MLL leukaemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627663. [PMID: 39713447 PMCID: PMC11661155 DOI: 10.1101/2024.12.11.627663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Understanding the molecular pathogenesis of MLL fusion oncoprotein (MLL-FP) leukaemia has spawned epigenetic therapies that have improved clinical outcomes in this often-incurable disease. Using genetic and pharmacological approaches, we define the individual and combined contribution of KAT6A, KAT6B and KAT7, in MLL-FP leukaemia. Whilst inhibition of KAT6A/B is efficacious in some pre-clinical models, simultaneous targeting of KAT7, with the novel inhibitor PF-9363, increases the therapeutic efficacy. KAT7 interacts with Menin and the MLL complex and is co-localised at chromatin to co-regulate the MLL-FP transcriptional program. Inhibition of KAT6/KAT7 provides an orthogonal route to targeting Menin to disable the transcriptional activity of MLL-FP. Consequently, combined inhibition rapidly evicts the MLL-FP from chromatin, potently represses oncogenic transcription and overcomes primary resistance to Menin inhibitors. Moreover, PF-9363 or genetic depletion of KAT7 can also overcome acquired genetic/non-genetic resistance to Menin inhibition. These data provide the molecular rationale for rapid clinical translation of combination therapy in MLL-FP leukaemia.
Collapse
|
19
|
Hojjatipour T, Ajeli M, Maali A, Azad M. Epigenetic-modifying agents: The potential game changers in the treatment of hematologic malignancies. Crit Rev Oncol Hematol 2024; 204:104498. [PMID: 39244179 DOI: 10.1016/j.critrevonc.2024.104498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024] Open
Abstract
Hematologic malignancies are lethal diseases arising from accumulated leukemic cells with substantial genetic or epigenetic defects in their natural development. Epigenetic modifications, including DNA methylation and histone modifications, are critical in hematologic malignancy formation, propagation, and treatment response. Both mutations and aberrant recruitment of epigenetic modifiers are reported in different hematologic malignancies, which regarding the reversible nature of epigenetic regulations, make them a potential target for cancer treatment. Here, we have first outlined a comprehensive overview of current knowledge related to epigenetic regulation's impact on the development and prognosis of hematologic malignancies. Furthermore, we have presented an updated overview regarding the current status of epigenetic-based drugs in hematologic malignancies treatment. And finally, discuss current challenges and ongoing clinical trials based on the manipulation of epigenetic modifies in hematologic malignancies.
Collapse
Affiliation(s)
- Tahereh Hojjatipour
- Cancer Immunology Group, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Mina Ajeli
- Department of Medical Laboratory Sciences, Guilan University of Medical Sciences, Guilan, Iran
| | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran; Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, School of Paramedicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
20
|
Ma F, Ren M, Li Z, Tang Y, Sun X, Wang Y, Cao N, Zhu X, Xu Y, Wang R, Shen Y, Zhao R, Li Z, Ashrafizadeh M, Sethi G, Wang F, Zhao A. ARID1A is a coactivator of STAT5 that contributes to CD8 + T cell dysfunction and anti-PD-1 resistance in gastric cancer. Pharmacol Res 2024; 210:107499. [PMID: 39549895 DOI: 10.1016/j.phrs.2024.107499] [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: 07/24/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
ARID1A deletion mutation contributes to improved treatment of several malignancies with immune checkpoint inhibitors (ICIs). However, its role in modulating of tumor immune microenvironment (TIME) of gastric cancer (GC) remains unclear. Here, we report an increase of CD8+ T cells infiltration in GC patients with ARID1A-mutation (MUT), which enhances sensitivity to ICIs. Kaplan-Meier survival analysis showed that ARID1A-mutation patients with gastrointestinal malignancies benefit from immunotherapy. Transcriptome analysis implicated that ARID1A regulates STAT5 downstream targets to inhibit T-cell mediated toxicity. Integrated dual luciferase assay and ChIP-qPCR analyses indicated that ARID1A coordinated with STAT5 to facilitate the transcription of the immunosuppressive factors TGF-β1 and NOX4. ARID1A recruited canonical BAF complex (cBAF) subunits, including SMARCB1 and SMARCD1, to sustain DNA accessibility. Downregulation of ARID1A reduced chromatin remodeling into configurations which make GC more sensitive to ICIs. In addition, targeting STAT5 effectively improved anti-PD-1 efficiency in ARID1A-wild type (WT) GC patients. Taken together, ARID1A is a coactivator of STAT5, function as a chromatin organizer in GC ICIs resistance, and targeting STAT5 is an effective strategy to improve the efficiency of ICIs in GC.
Collapse
Affiliation(s)
- Fangqi Ma
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Department of Traditional Chinese Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Mingming Ren
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zhongqiu Li
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan 450004, China
| | - Yujing Tang
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 611756, China
| | - Xiaoyu Sun
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yi Wang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Nida Cao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xiaohong Zhu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan Xu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Rui Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Yumiao Shen
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ruohan Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zhaoyan Li
- Department of Traditional Chinese Medicine, School of Medicine Affiliated Ruijin Hospital, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR) Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Furong Wang
- Department of Pathology, the Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, Guangdong 516002, China.
| | - Aiguang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
21
|
Sethy B, Yu ZY, Narwanti I, Upadhyay R, Lai MJ, Lee SB, Liou JP. Design, synthesis, and biological evaluation of adenosine derivatives targeting DOT1L and HAT as anti-leukemia agents. Bioorg Chem 2024; 153:107771. [PMID: 39299178 DOI: 10.1016/j.bioorg.2024.107771] [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: 02/24/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024]
Abstract
Disruptor of telomeric silencing 1-like (DOT1L) is a key hub in histone lysine methyltransferase and an attractive therapeutic target for treating hematological malignancies including acute myeloid leukemia (AML). In this study, we report the design and synthesis of a new series of adenosine derivatives as DOT1L inhibitors by accommodating a basic linker piperidine-4-ylmethyl motif to respective aryl-urea/benzimidazole scaffolds. The anti-DOT1L enzyme activity analysis demonstrated that compounds 8, 12, and 13 strongly suppressed DOT1L activity with IC50 values ranging from 0.125 to 0.408 µM among all the synthetics, and the structure-activity relationships were summarized. Moreover, compound 12 possessed relatively potent DOT1L inhibitory activity by significantly reduced histone H3 di-methylation at lysine 79 (H3K79me2) level in cells. Subsequently, all the synthetics were screened against various leukemia cell lines, indicating the DOT1L active adenosine derivatives exhibited low to moderate while compound 15 showed strong cellular inhibition despite its unsuccessful DOT1L inhibition. Therefore, acknowledging the distinctive potency of compound 15 against five different leukemia cell lines, including MLL-r (MV4-11) and non-MLL-r cell lines (HL-60, HH, K562, and KG-1), with IC50 values in the 0.45 ∼ 1.66 μM range and its mode of action was explored. Furthermore, compound 15 hindered histone acetylation, induced remarkable DNA damage, and triggered apoptosis. Importantly, normal T lymphocytes only showed moderate response to compound 15. These findings provide a basis for future studies on its potential application against AML.
Collapse
Affiliation(s)
- Bidyadhar Sethy
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Zih-Yao Yu
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Iin Narwanti
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Faculty of Pharmacy, Universitas Ahmad Dahlan, Yogyakarta, Indonesia
| | - Richa Upadhyay
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Mei-Jung Lai
- TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Sung-Bau Lee
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
22
|
Bonilla G, Morris A, Kundu S, DuCasse A, Kirkpatrick G, Jeffries NE, Chetal K, Yvanovich EE, Milosevic J, Zhao T, Xia J, Barghout R, Scadden D, Mansour MK, Kingston RE, Sykes DB, Mercier FE, Sadreyev RI. Leukemia aggressiveness is driven by chromatin remodeling and expression changes of core regulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582846. [PMID: 38496490 PMCID: PMC10942317 DOI: 10.1101/2024.02.29.582846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Molecular mechanisms driving clonal aggressiveness in leukemia are not fully understood. We tracked and analyzed MLL-rearranged leukemic clones independently evolving towards higher aggressiveness. More aggressive subclones lost their growth differential ex vivo but restored it upon secondary transplantation, suggesting molecular memory of aggressiveness. Development of aggressiveness was associated with clone-specific gradual modulation of chromatin states and expression levels across the genome, with a surprising preferential trend of reversing the earlier changes between normal and leukemic progenitors. To focus on the core aggressiveness program, we identified genes with consistent changes of expression and chromatin marks that were maintained in vivo and ex vivo in both clones. Overexpressing selected core genes (Smad1 as aggressiveness driver, Irx5 and Plag1 as suppressors) affected leukemic progenitor growth in the predicted way and had convergent downstream effects on central transcription factors and repressive epigenetic modifiers, suggesting a broader regulatory network of leukemic aggressiveness.
Collapse
|
23
|
Nadiminti KVG, Sahasrabudhe KD, Liu H. Menin inhibitors for the treatment of acute myeloid leukemia: challenges and opportunities ahead. J Hematol Oncol 2024; 17:113. [PMID: 39558390 PMCID: PMC11575055 DOI: 10.1186/s13045-024-01632-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024] Open
Abstract
The AML treatment landscape has significantly changed in recent years with the approval of targeted therapies in the front-line and relapsed/refractory settings, including inhibitors of FLT3 and IDH1/2 mutations. More importantly, approval of the combination of the BCl-2 inhibitor, venetoclax, and hypomethylating agents or low dose cytarabine provided unprecedented breakthrough for the frontline treatment of older, unfit AML patients. Even with all this exciting progress, more targeted therapies for AML treatment are needed. Recent development of menin inhibitors targeting AML with KMT2A rearrangements or NPM1 mutations could represent a promising new horizon of treatment for patients within these subsets of AML. Our current review will focus on a summary and updates of recent developments of menin inhibitors in the treatment of AML, on the challenges ahead arising from drug resistance, as well as on the opportunities of novel combinations with menin inhibitors.
Collapse
Affiliation(s)
- Kalyan V G Nadiminti
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI, 53705-2281, USA.
| | - Kieran D Sahasrabudhe
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI, 53705-2281, USA
| | - Hongtao Liu
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI, 53705-2281, USA
| |
Collapse
|
24
|
Janssens DH, Duran M, Otto DJ, Wu W, Xu Y, Kirkey D, Mullighan CG, Yi JS, Meshinchi S, Sarthy JF, Ahmad K, Henikoff S. MLL oncoprotein levels influence leukemia lineage identities. Nat Commun 2024; 15:9341. [PMID: 39472576 PMCID: PMC11522475 DOI: 10.1038/s41467-024-53399-8] [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: 02/16/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024] Open
Abstract
Chromosomal translocations involving the mixed-lineage leukemia (MLL) locus generate potent oncogenic fusion proteins (oncoproteins) that disrupt regulation of developmental gene expression. By profiling the oncoprotein-target sites of 36 broadly representative MLL-rearranged leukemia samples, including three samples that underwent a lymphoid-to-myeloid lineage-switching event in response to therapy, we find the genomic enrichment of the oncoprotein is highly variable between samples and subject to dynamic regulation. At high levels of expression, the oncoproteins preferentially activate either an acute lymphoblastic leukemia (ALL) program, enriched for pro-B-cell genes, or an acute myeloid leukemia (AML) program, enriched for hematopoietic-stem-cell genes. The fusion-partner-specific-binding patterns over these gene sets are highly correlated with the prevalence of each mutation in ALL versus AML. In lineage-switching samples the oncoprotein levels are reduced and the oncoproteins preferentially activate granulocyte-monocyte progenitor (GMP) genes. In a sample that lineage switched during treatment with the menin inhibitor revumenib, the oncoprotein and menin are reduced to undetectable levels, but ENL, a transcriptional cofactor of the oncoprotein, persists on numerous oncoprotein-target loci, including genes in the GMP-like lineage-switching program. We propose MLL oncoproteins promote lineage-switching events through dynamic chromatin binding at lineage-specific target genes, and may support resistance to menin inhibitors through similar changes in chromatin occupancy.
Collapse
Affiliation(s)
- Derek H Janssens
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Melodie Duran
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dominik J Otto
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Data Science IRC, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Weifang Wu
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yiling Xu
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Danielle Kirkey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Seattle Children's Hospital, Seattle, WA, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center of Excellence for Leukemia Studies, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Joanna S Yi
- Pediatric Hematology and Oncology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Soheil Meshinchi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Seattle Children's Hospital, Seattle, WA, USA
| | - Jay F Sarthy
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Kami Ahmad
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Steven Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
25
|
Chen Q, Wang S, Zhang J, Xie M, Lu B, He J, Zhen Z, Li J, Zhu J, Li R, Li P, Wang H, Vakoc C, Roeder RG, Chen M. JMJD1C forms condensates to facilitate a RUNX1-dependent gene expression program shared by multiple types of AML cells. Protein Cell 2024:pwae059. [PMID: 39450904 DOI: 10.1093/procel/pwae059] [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: 04/25/2024] [Indexed: 10/26/2024] Open
Abstract
JMJD1C, a member of the lysine demethylase 3 (KDM3) family, is universally required for the survival of several types of acute myeloid leukemia (AML) cells with different genetic mutations, representing a therapeutic opportunity with broad application. Yet how JMJD1C regulates the leukemic programs of various AML cells is largely unexplored. Here we show that JMJD1C interacts with the master hematopoietic transcription factor RUNX1, which thereby recruits JMJD1C to the genome to facilitate a RUNX1-driven transcriptional program that supports leukemic cell survival. The underlying mechanism hinges on the long N-terminal disordered region of JMJD1C, which harbors two inseparable abilities: condensate formation and direct interaction with RUNX1. This dual capability of JMJD1C may influence enhancer-promoter contacts crucial for the expression of key leukemic genes regulated by RUNX1. Our findings demonstrate a previously unappreciated role for the non-catalytic function of JMJD1C in transcriptional regulation, underlying a mechanism shared by different types of leukemias.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Saisai Wang
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Juqing Zhang
- School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China
| | - Min Xie
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Bin Lu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Jie He
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of People Liberation Army (PLA), Second Military Medical University (Naval Medical University), Shanghai 200052, China
| | - Zhuoran Zhen
- State Key Laboratory of Molecular Oncology, Tsinghua-Peking Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Jing Li
- Department of Precision Medicine, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Jiajun Zhu
- State Key Laboratory of Molecular Oncology, Tsinghua-Peking Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Rong Li
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of People Liberation Army (PLA), Second Military Medical University (Naval Medical University), Shanghai 200052, China
| | - Pilong Li
- State Key Laboratory of Membrane Biology, Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Haifeng Wang
- School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China
| | | | - Robert G Roeder
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| | - Mo Chen
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan 030607, China
| |
Collapse
|
26
|
Hu Y, Ye S, Kong J, Zhou Q, Wang Z, Zhang Y, Yan H, Wang Y, Li T, Xie Y, Chen B, Zhao Y, Zhang T, Zheng X, Niu J, Hu B, Wang S, Chen Z, Zheng C. DOT1L protects against podocyte injury in diabetic kidney disease through phospholipase C-like 1. Cell Commun Signal 2024; 22:519. [PMID: 39456056 PMCID: PMC11515305 DOI: 10.1186/s12964-024-01895-1] [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: 08/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Podocyte injury causes proteinuria and accelerates glomerular sclerosis during diabetic kidney disease (DKD). Disruptor of telomeric silencing 1-like (DOT1L), an evolutionarily conserved histone methyltransferase, has been reported in preventing kidney fibrosis in chronic kidney disease models. However, whether DOT1L exerts beneficial effects in diabetes induced podocyte injury and the underlying molecular mechanisms need further exploration. METHODS The expression of DOT1L was confirmed by Western blotting in MPC-5 cells and cortex of kidney from db/db mice, as well as immunofluorescence staining in human renal biopsy samples. The effect of DOT1L on podocyte injury was obtained using MPC-5 cells and db/db mice. The potential target genes regulated by DOT1L was measured by RNA-sequencing. Then, a series of molecular biological experiments was performed to investigate the regulation of PLCL1 by DOT1L in MCP-5 cells and db/db mice. Lipid accumulation was assessed by UPLC-MS/MS analysis and Oil Red O staining. RESULTS DOT1L expression was significantly declined in high glucose (HG)-treated MPC-5 cells, podocyte regions of kidney tissues from db/db mice and human renal biopsy samples. Subsequent investigations revealed that upregulation of DOT1L ameliorated HG-induced cell apoptosis in MPC-5 cells as well as primary podocytes. Furthermore, podocyte-specific DOT1L overexpression inhibited diabetic podocyte injury in db/db mice. Mechanistically, we revealed that DOT1L upregulated phospholipase C-like 1 (PLCL1) expression by mediating H3K79me2 at its promoter and PLCL1 silencing suppressed the protective role of DOT1L on podocyte injury. Moreover, DOT1L improved diabetes induced abnormal fatty acid metabolism in podocytes and PLCL1 knockdown reversed its protective effects. CONCLUSIONS Taken together, our results indicate that DOT1L protects podocyte injury via PLCL1-mediated fatty acid metabolism and provides new insights into the therapeutic target of DKD.
Collapse
Affiliation(s)
- Yepeng Hu
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Shu Ye
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Jing Kong
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Qiao Zhou
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Zhe Wang
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Yikai Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Han Yan
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Yaqiong Wang
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Tiekun Li
- Nanjing Kingmed Center for Clinical Laboratory Co., Ltd., 11 Yaogu Avenue, Nanjing, Jiangsu, China
| | - Yi Xie
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Bingbing Chen
- Nursing Department, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Yiming Zhao
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Tianyue Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Xianan Zheng
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Junjia Niu
- Department of Nephrology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Bibi Hu
- Nursing Department, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Shengyao Wang
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Zhida Chen
- Department of Nephrology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China.
| | - Chao Zheng
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China.
| |
Collapse
|
27
|
Tan Z, Guo N, Liu S, Li J, Chen Y, Cui J, Lei H, Jiang N, Wang L, Zhai X. Discovery of Novel Non-nucleoside DOT1L R231Q Inhibitors with Improved Pharmacokinetic Properties and Anti-lung Cancer Efficacy. J Med Chem 2024; 67:16248-16269. [PMID: 39255403 DOI: 10.1021/acs.jmedchem.4c01096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Given the considerable potential of DOT1LR231Q inhibitors in lung cancer therapy and the problematic pharmacokinetics of nucleoside inhibitors, our group launched a development program of non-nucleoside DOT1LR231Q inhibitors to improve the pharmacokinetic properties. Herein, two series of non-nucleoside compounds bearing piperidine or 3-(aminomethyl)pyrrolidin-3-ol as "ribose mimics" were designed and evaluated through antiproliferation assay and western blot analysis. The optimal TB22 inhibited the proliferation of H460R231Q cells with an IC50 value of 2.85 μM, about 13-fold more potent than SGC0946. Notably, TB22 demonstrated significant in vivo efficacy (TGI = 60.57%) in H460R231Q cell-derived xenograft models and improved pharmacokinetic properties (t1/2 = 6.06 ± 2.94 h and CL = 55.18 ± 8.56 mL/kg/min). Moreover, a mechanism study validated that TB22 suppressed malignant phenotypes of lung cancer cells harboring R231Q mutation via the MAPK/ERK signaling pathway. This work provides a promising molecule for lung cancer therapy in favor of clinical patients.
Collapse
Affiliation(s)
- Zehui Tan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ning Guo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuyu Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiandong Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Chen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jianming Cui
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hongrui Lei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Nan Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
28
|
Perlman BS, Burget N, Zhou Y, Schwartz GW, Petrovic J, Modrusan Z, Faryabi RB. Enhancer-promoter hubs organize transcriptional networks promoting oncogenesis and drug resistance. Nat Commun 2024; 15:8070. [PMID: 39277592 PMCID: PMC11401928 DOI: 10.1038/s41467-024-52375-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 09/04/2024] [Indexed: 09/17/2024] Open
Abstract
Recent advances in high-resolution mapping of spatial interactions among regulatory elements support the existence of complex topological assemblies of enhancers and promoters known as enhancer-promoter hubs or cliques. Yet, organization principles of these multi-interacting enhancer-promoter hubs and their potential role in regulating gene expression in cancer remain unclear. Here, we systematically identify enhancer-promoter hubs in breast cancer, lymphoma, and leukemia. We find that highly interacting enhancer-promoter hubs form at key oncogenes and lineage-associated transcription factors potentially promoting oncogenesis of these diverse cancer types. Genomic and optical mapping of interactions among enhancer and promoter elements further show that topological alterations in hubs coincide with transcriptional changes underlying acquired resistance to targeted therapy in T cell leukemia and B cell lymphoma. Together, our findings suggest that enhancer-promoter hubs are dynamic and heterogeneous topological assemblies with the potential to control gene expression circuits promoting oncogenesis and drug resistance.
Collapse
Affiliation(s)
- Brent S Perlman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, USA
| | - Noah Burget
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, USA
| | - Yeqiao Zhou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, USA
| | - Gregory W Schwartz
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Jelena Petrovic
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, USA
| | - Zora Modrusan
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, USA
| | - Robert B Faryabi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA.
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, USA.
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
29
|
Wang Y, Zhang N, Shang W, Peng H, Hu Z, Yang Y, Tan L, Zhang L, He F, Rao X. Dexamethasone Inhibits the Growth of B-Lymphoma Cells by Downregulating DOT1L. Cancer Rep (Hoboken) 2024; 7:e2150. [PMID: 39307938 PMCID: PMC11417011 DOI: 10.1002/cnr2.2150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 07/18/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Dexamethasone (Dex), a synthetic glucocorticoid that acts by binding to the glucocorticoid receptor (GR), has been widely applied to treat leukemia and lymphoma; however, the precise mechanism underlying Dex action is still not well elucidated. DOT1L, a histone H3-lysine79 (H3K79) methyltransferase, has been linked to multiple cancer types, particularly mixed lineage leukemia (MLL) gene rearranged leukemia, but its contribution to lymphoma is yet to be delineated. Analysis from the TCGA database displayed that DOT1L was highly expressed in lymphoma and leukemia. RESULTS We initially demonstrated that DOT1L served as a new target gene controlled by GR, and the downregulation of DOT1L was critical for the killing of B-lymphoma cells by Dex. Further study revealed that Dex had no impact on the transcriptional activity of the DOT1L promoter, rather it reduced the mRNA level of DOT1L at the posttranscriptional level. In addition, knockdown of DOT1L remarkably inhibited the B-lymphoma cell growth. CONCLUSIONS Overall, our findings indicated that DOT1L may serve as a potential drug target and a promising biomarker of Dex sensitivity when it comes to treating B lymphoma.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Nan Zhang
- Department of HematologyPeople's Liberation Army the General Hospital of Western Theater CommandChengduChina
| | - Weilong Shang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Huagang Peng
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Zhen Hu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Yi Yang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Li Tan
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| | - Li Zhang
- Department of OncologyXiangya Hospital, Central South UniversityChangshaChina
| | - Fengtian He
- Department of Biochemistry and Molecular BiologyCollege of Basic Medical Sciences, Army Medical UniversityChongqingChina
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in ChongqingArmy Medical UniversityChongqingChina
| |
Collapse
|
30
|
Xue Z, Qin L, Xuan H, Luo K, Huang M, Xie L, Su Y, Xu L, Harsh J, Dale B, Shi X, Chen X, Kaniskan HÜ, Jin J, Wen H. A potent and selective ENL degrader suppresses oncogenic gene expression and leukemia progression. SCIENCE ADVANCES 2024; 10:eado1432. [PMID: 39196923 PMCID: PMC11352836 DOI: 10.1126/sciadv.ado1432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/24/2024] [Indexed: 08/30/2024]
Abstract
The histone acylation reader eleven-nineteen leukemia (ENL) plays a pivotal role in sustaining oncogenesis in acute leukemias, particularly in mixed-lineage leukemia-rearranged (MLL-r) leukemia. ENL relies on its reader domain to recognize histone lysine acylation promoting oncogenic gene expression and leukemia progression. Here, we report the development of MS41, a highly potent and selective von Hippel-Lindau-recruiting ENL degrader that effectively inhibits the growth of ENL-dependent leukemia cells. MS41-induced ENL degradation reduces the chromatin occupancy of ENL-associated transcription elongation machinery, resulting in the suppression of key oncogenic gene expression programs and the activation of differentiation genes. MS41 is well-tolerated in vivo and substantially suppresses leukemia progression in a xenograft mouse model of MLL-r leukemia. Notably, MS41 also induces the degradation of mutant ENL proteins identified in Wilms' tumors. Our findings emphasize the therapeutic potential of pharmacological ENL degradation for treating ENL-dependent cancers, making MS41 not only a valuable chemical probe but also potential anticancer therapeutic for further development.
Collapse
Affiliation(s)
- Zhaoyu Xue
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Lihuai Qin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences, and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, New York, USA
| | - Hongwen Xuan
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Kaixiu Luo
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences, and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, New York, USA
| | - Mengying Huang
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yangzhou Su
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Longxia Xu
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Josiah Harsh
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Brandon Dale
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences, and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, New York, USA
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - H Ümit Kaniskan
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences, and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, New York, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences, and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, New York, USA
| | - Hong Wen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
31
|
Yang Z, Zhang G, Zhao R, Tian T, Zhi J, Wei G, Roeder RG, Jing L, Yu M. MLL-AF9 regulates transcriptional initiation in mixed lineage leukemic cells. J Biol Chem 2024; 300:107566. [PMID: 39002676 PMCID: PMC11345648 DOI: 10.1016/j.jbc.2024.107566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/15/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024] Open
Abstract
Mixed lineage leukemia-fusion proteins (MLL-FPs) are believed to maintain gene activation and induce MLL through aberrantly stimulating transcriptional elongation, but the underlying mechanisms are incompletely understood. Here, we show that both MLL1 and AF9, one of the major fusion partners of MLL1, mainly occupy promoters and distal intergenic regions, exhibiting chromatin occupancy patterns resembling that of RNA polymerase II in HEL, a human erythroleukemia cell line without MLL1 rearrangement. MLL1 and AF9 only coregulate over a dozen genes despite of their co-occupancy on thousands of genes. They do not interact with each other, and their chromatin occupancy is also independent of each other. Moreover, AF9 deficiency in HEL cells decreases global TBP occupancy while decreases CDK9 occupancy on a small number of genes, suggesting an accessory role of AF9 in CDK9 recruitment and a possible major role in transcriptional initiation via initiation factor recruitment. Importantly, MLL1 and MLL-AF9 occupy promoters and distal intergenic regions, exhibiting identical chromatin occupancy patterns in MLL cells, and MLL-AF9 deficiency decreased occupancy of TBP and TFIIE on major target genes of MLL-AF9 in iMA9, a murine acute myeloid leukemia cell line inducibly expressing MLL-AF9, suggesting that it can also regulate initiation. These results suggest that there is no difference between MLL1 and MLL-AF9 with respect to location and size of occupancy sites, contrary to what people have believed, and that MLL-AF9 may also regulate transcriptional initiation in addition to widely believed elongation.
Collapse
Affiliation(s)
- Zimei Yang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ge Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ruoyu Zhao
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Tian Tian
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Junhong Zhi
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Gang Wei
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Robert G Roeder
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, New York, USA
| | - Lili Jing
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Yu
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
32
|
Tan Z, Guo N, Cao Z, Liu S, Zhang J, Ma D, Zhang J, Lv W, Jiang N, Zang L, Wang L, Zhai X. Discovery of first-in-class DOT1L inhibitors against the R231Q gain-of-function mutation in the catalytic domain with therapeutic potential of lung cancer. Acta Pharm Sin B 2024; 14:3605-3623. [PMID: 39220866 PMCID: PMC11365375 DOI: 10.1016/j.apsb.2024.03.018] [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: 01/19/2024] [Revised: 02/18/2024] [Accepted: 03/01/2024] [Indexed: 09/04/2024] Open
Abstract
Recent research certified that DOT1L and its mutations represented by R231Q were potential targets for the treatment of lung cancer. Herein, a series of adenosine-containing derivatives were identified with DOT1LR231Q inhibition through antiproliferation assay and Western blot analysis in the H460R231Q cell. The most promising compound 37 significantly reduced DOT1LR231Q mediated H3K79 methylation and effectively inhibited the proliferation, self-renewal, migration, and invasion of lung cancer cell lines at low micromolar concentrations. The cell permeability and cellular target engagement of 37 were verified by both CETSA and DARTS assays. In the H460R231Q OE cell-derived xenograft (CDX) model, 37 displayed pronounced tumor growth inhibition after intraperitoneal administration at 20 mg/kg dose for 3 weeks (TGI = 54.38%), without obvious toxicities. A pharmacokinetic study revealed that 37 possessed tolerable properties (t 1/2 = 1.93 ± 0.91 h, F = 97.2%) after intraperitoneal administration in rats. Mechanism study confirmed that 37 suppressed malignant phenotypes of lung cancer carrying R231Q gain-of-function mutation via the MAPK/ERK signaling pathway. Moreover, analysis of the binding modes between molecules and DOT1LWT/R231Q proteins put forward the "Induced-fit" allosteric model in favor to the discovery of potent DOT1L candidates.
Collapse
Affiliation(s)
- Zehui Tan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ning Guo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhi Cao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuyu Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiayu Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Deyi Ma
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiahao Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wencai Lv
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Nan Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linghe Zang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
33
|
Perlman BS, Burget N, Zhou Y, Schwartz GW, Petrovic J, Modrusan Z, Faryabi RB. Enhancer-promoter hubs organize transcriptional networks promoting oncogenesis and drug resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601745. [PMID: 39005446 PMCID: PMC11244972 DOI: 10.1101/2024.07.02.601745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Recent advances in high-resolution mapping of spatial interactions among regulatory elements support the existence of complex topological assemblies of enhancers and promoters known as enhancer-promoter hubs or cliques. Yet, organization principles of these multi-interacting enhancer-promoter hubs and their potential role in regulating gene expression in cancer remains unclear. Here, we systematically identified enhancer-promoter hubs in breast cancer, lymphoma, and leukemia. We found that highly interacting enhancer-promoter hubs form at key oncogenes and lineage-associated transcription factors potentially promoting oncogenesis of these diverse cancer types. Genomic and optical mapping of interactions among enhancer and promoter elements further showed that topological alterations in hubs coincide with transcriptional changes underlying acquired resistance to targeted therapy in T cell leukemia and B cell lymphoma. Together, our findings suggest that enhancer-promoter hubs are dynamic and heterogeneous topological assemblies with the potential to control gene expression circuits promoting oncogenesis and drug resistance.
Collapse
|
34
|
Sudholz H, Schuster IS, Foroutan M, Sng X, Andoniou CE, Doan A, Camilleri T, Shen Z, Zaph C, Degli-Esposti MA, Huntington ND, Scheer S. DOT1L maintains NK cell phenotype and function for optimal tumor control. Cell Rep 2024; 43:114333. [PMID: 38865244 DOI: 10.1016/j.celrep.2024.114333] [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: 11/06/2023] [Revised: 03/06/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
Histone methyltransferases (HMTs) are crucial in gene regulation and function, yet their role in natural killer (NK) cell biology within the tumor microenvironment (TME) remains largely unknown. We demonstrate that the HMT DOT1L limits NK cell conversion to CD49a+ CD49b+ intILC1, a subset that can be observed in the TME in response to stimulation with transforming growth factor (TGF)-β and is correlated with impaired tumor control. Deleting Dot1l in NKp46-expressing cells reveals its pivotal role in maintaining NK cell phenotype and function. Loss of DOT1L skews NK cells toward intILC1s even in the absence of TGF-β. Transcriptionally, DOT1L-null NK cells closely resemble intILC1s and ILC1s, correlating with altered NK cell responses and impaired solid tumor control. These findings deepen our understanding of NK cell biology and could inform approaches to prevent NK cell conversion to intILC1s in adoptive NK cell therapies for cancer.
Collapse
Affiliation(s)
- Harrison Sudholz
- Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry, Monash University, Clayton, VIC 3800, Australia
| | - Iona S Schuster
- Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA 6009, Australia
| | - Momeneh Foroutan
- Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry, Monash University, Clayton, VIC 3800, Australia; oNKo-Innate Pty Ltd, Moonee Ponds, VIC 3039, Australia
| | - Xavier Sng
- Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Christopher E Andoniou
- Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA 6009, Australia
| | - Anh Doan
- Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry, Monash University, Clayton, VIC 3800, Australia
| | - Tania Camilleri
- Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry, Monash University, Clayton, VIC 3800, Australia
| | - Zihan Shen
- Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry, Monash University, Clayton, VIC 3800, Australia
| | - Colby Zaph
- Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry, Monash University, Clayton, VIC 3800, Australia
| | - Mariapia A Degli-Esposti
- Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA 6009, Australia
| | - Nicholas D Huntington
- Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry, Monash University, Clayton, VIC 3800, Australia; oNKo-Innate Pty Ltd, Moonee Ponds, VIC 3039, Australia.
| | - Sebastian Scheer
- Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
35
|
Kealy L, Runting J, Thiele D, Scheer S. An emerging maestro of immune regulation: how DOT1L orchestrates the harmonies of the immune system. Front Immunol 2024; 15:1385319. [PMID: 38962004 PMCID: PMC11219580 DOI: 10.3389/fimmu.2024.1385319] [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: 02/12/2024] [Accepted: 04/04/2024] [Indexed: 07/05/2024] Open
Abstract
The immune system comprises a complex yet tightly regulated network of cells and molecules that play a critical role in protecting the body from infection and disease. The activity and development of each immune cell is regulated in a myriad of ways including through the cytokine milieu, the availability of key receptors, via tailored intracellular signalling cascades, dedicated transcription factors and even by directly modulating gene accessibility and expression; the latter is more commonly known as epigenetic regulation. In recent years, epigenetic regulators have begun to emerge as key players involved in modulating the immune system. Among these, the lysine methyltransferase DOT1L has gained significant attention for its involvement in orchestrating immune cell formation and function. In this review we provide an overview of the role of DOT1L across the immune system and the implications of this role on health and disease. We begin by elucidating the general mechanisms of DOT1L-mediated histone methylation and its impact on gene expression within immune cells. Subsequently, we provide a detailed and comprehensive overview of recent studies that identify DOT1L as a crucial regulator of immune cell development, differentiation, and activation. Next, we discuss the potential mechanisms of DOT1L-mediated regulation of immune cell function and shed light on how DOT1L might be contributing to immune cell homeostasis and dysfunction. We then provide food for thought by highlighting some of the current obstacles and technical limitations precluding a more in-depth elucidation of DOT1L's role. Finally, we explore the potential therapeutic implications of targeting DOT1L in the context of immune-related diseases and discuss ongoing research efforts to this end. Overall, this review consolidates the current paradigm regarding DOT1L's role across the immune network and emphasises its critical role in governing the healthy immune system and its potential as a novel therapeutic target for immune-related diseases. A deeper understanding of DOT1L's immunomodulatory functions could pave the way for innovative therapeutic approaches which fine-tune the immune response to enhance or restore human health.
Collapse
Affiliation(s)
- Liam Kealy
- Immunity Program, The Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jessica Runting
- Immunity Program, The Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Daniel Thiele
- Immunity Program, The Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sebastian Scheer
- Immunity Program, The Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
36
|
Gao X, Zuo X, Min T, Wan Y, He Y, Jiang B. Traditional Chinese medicine for acute myelocytic leukemia therapy: exploiting epigenetic targets. Front Pharmacol 2024; 15:1388903. [PMID: 38895633 PMCID: PMC11183326 DOI: 10.3389/fphar.2024.1388903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy with historically high mortality rates. The treatment strategies for AML is still internationally based on anthracyclines and cytarabine, which remained unchanged for decades. With the rapid advance on sequencing technology, molecular targets of leukemogenesis and disease progression related to epigenetics are constantly being discovered, which are important for the prognosis and treatment of AML. Traditional Chinese medicine (TCM) is characterized by novel pharmacological mechanisms, low toxicity and limited side effects. Several biologically active ingredients of TCM are effective against AML. This review focuses on bioactive compounds in TCM targeting epigenetic mechanisms to address the complexities and heterogeneity of AML.
Collapse
Affiliation(s)
- Xinlong Gao
- Naval Medical Center of PLA, Shanghai, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Xu Zuo
- Naval Medical Center of PLA, Shanghai, China
| | | | - Yu Wan
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Ying He
- Naval Medical Center of PLA, Shanghai, China
| | - Beier Jiang
- Naval Medical Center of PLA, Shanghai, China
| |
Collapse
|
37
|
Wang G, Zhang W, Ren J, Zeng Y, Dang X, Tian X, Yu W, Li Z, Ma Y, Yang P, Lu J, Zheng J, Lu B, Xu J, Liang A. The DNA damage-independent ATM signalling maintains CBP/DOT1L axis in MLL rearranged acute myeloid leukaemia. Oncogene 2024; 43:1900-1916. [PMID: 38671157 PMCID: PMC11178498 DOI: 10.1038/s41388-024-02998-2] [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: 11/21/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 04/28/2024]
Abstract
The long-term maintenance of leukaemia stem cells (LSCs) is responsible for the high degree of malignancy in MLL (mixed-lineage leukaemia) rearranged acute myeloid leukaemia (AML). The DNA damage response (DDR) and DOT1L/H3K79me pathways are required to maintain LSCs in MLLr-AML, but little is known about their interplay. This study revealed that the DDR enzyme ATM regulates the maintenance of LSCs in MLLr-AML with a sequential protein-posttranslational-modification manner via CBP-DOT1L. We identified the phosphorylation of CBP by ATM, which confers the stability of CBP by preventing its proteasomal degradation, and characterised the acetylation of DOT1L by CBP, which mediates the high level of H3K79me2 for the expression of leukaemia genes in MLLr-AML. In addition, we revealed that the regulation of CBP-DOT1L axis in MLLr-AML by ATM was independent of DNA damage activation. Our findings provide insight into the signalling pathways involoved in MLLr-AML and broaden the understanding of the role of DDR enzymes beyond processing DNA damage, as well as identigying them as potent cancer targets.
Collapse
Affiliation(s)
- Guangming Wang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Postdoctoral Station of Clinical Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Wenjun Zhang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Jie Ren
- Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yu Zeng
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiuyong Dang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiaoxue Tian
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wenlei Yu
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Zheng Li
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yuting Ma
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Pingping Yang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Jinyuan Lu
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Bing Lu
- East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Jun Xu
- East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Aibin Liang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
38
|
Separovich RJ, Karakatsanis NM, Gao K, Fuh D, Hamey JJ, Wilkins MR. Proline-directed yeast and human MAP kinases phosphorylate the Dot1p/DOT1L histone H3K79 methyltransferase. FEBS J 2024; 291:2590-2614. [PMID: 38270553 DOI: 10.1111/febs.17062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 10/24/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Disruptor of telomeric silencing 1 (Dot1p) is an exquisitely conserved histone methyltransferase and is the sole enzyme responsible for H3K79 methylation in the budding yeast Saccharomyces cerevisiae. It has been shown to be highly phosphorylated in vivo; however, the upstream kinases that act on Dot1p are almost entirely unknown in yeast and all other eukaryotes. Here, we used in vitro and in vivo kinase discovery approaches to show that mitogen-activated protein kinase HOG1 (Hog1p) is a bona fide kinase of the Dot1p methyltransferase. In vitro kinase assays showed that Hog1p phosphorylates Dot1p at multiple sites, including at several proline-adjacent sites that are consistent with known Hog1p substrate preferences. The activity of Hog1p was specifically enhanced at these proline-adjacent sites on Dot1p upon Hog1p activation by the osmostress-responsive MAP kinase kinase PBS2 (Pbs2p). Genomic deletion of HOG1 reduced phosphorylation at specific sites on Dot1p in vivo, providing further evidence for Hog1p kinase activity on Dot1p in budding yeast cells. Phenotypic analysis of knockout and phosphosite mutant yeast strains revealed the importance of Hog1p-catalysed phosphorylation of Dot1p for cellular responses to ultraviolet-induced DNA damage. In mammalian systems, this kinase-substrate relationship was found to be conserved: human DOT1L (the ortholog of yeast Dot1p) can be phosphorylated by the proline-directed kinase p38β (also known as MAPK11; the ortholog of yeast Hog1p) at multiple sites in vitro. Taken together, our findings establish Hog1p and p38β as newly identified upstream kinases of the Dot1p/DOT1L H3K79 methyltransferase enzymes in eukaryotes.
Collapse
Affiliation(s)
- Ryan J Separovich
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Nicola M Karakatsanis
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Kelley Gao
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - David Fuh
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Joshua J Hamey
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Marc R Wilkins
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
39
|
Wang L, Chen Y, Wang Q, Xiang M, Zeng Z, Zhang Z, Zhang F, Chen S, Xue M. Identification and clinical implications of recurrent PAX5::MLLT3 fusion in lymphoid blastic phase chronic myeloid leukemia. Int J Lab Hematol 2024; 46:571-574. [PMID: 38389449 DOI: 10.1111/ijlh.14245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024]
Affiliation(s)
- Lijun Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Chen
- Department of Hematology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Qingyuan Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Xiang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhao Zeng
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhibo Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fenghong Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Mengxing Xue
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
40
|
Adriaanse FRS, Schneider P, Arentsen-Peters STCJM, da Fonseca AMN, Stutterheim J, Pieters R, Zwaan CM, Stam RW. Distinct Responses to Menin Inhibition and Synergy with DOT1L Inhibition in KMT2A-Rearranged Acute Lymphoblastic and Myeloid Leukemia. Int J Mol Sci 2024; 25:6020. [PMID: 38892207 PMCID: PMC11173273 DOI: 10.3390/ijms25116020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Pediatric acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) exhibit favorable survival rates. However, for AML and ALL patients carrying KMT2A gene translocations clinical outcome remains unsatisfactory. Key players in KMT2A-fusion-driven leukemogenesis include menin and DOT1L. Recently, menin inhibitors like revumenib have garnered attention for their potential therapeutic efficacy in treating KMT2A-rearranged acute leukemias. However, resistance to menin inhibition poses challenges, and identifying which patients would benefit from revumenib treatment is crucial. Here, we investigated the in vitro response to revumenib in KMT2A-rearranged ALL and AML. While ALL samples show rapid, dose-dependent induction of leukemic cell death, AML responses are much slower and promote myeloid differentiation. Furthermore, we reveal that acquired resistance to revumenib in KMT2A-rearranged ALL cells can occur either through the acquisition of MEN1 mutations or independently of mutations in MEN1. Finally, we demonstrate significant synergy between revumenib and the DOT1L inhibitor pinometostat in KMT2A-rearranged ALL, suggesting that such drug combinations represent a potent therapeutic strategy for these patients. Collectively, our findings underscore the complexity of resistance mechanisms and advocate for precise patient stratification to optimize the use of menin inhibitors in KMT2A-rearranged acute leukemia.
Collapse
Affiliation(s)
- Fabienne R. S. Adriaanse
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC-Sophia’s Children’s Hospital, 3015 CN Rotterdam, The Netherlands
| | - Pauline Schneider
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | | | | | - Janine Stutterheim
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Rob Pieters
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - C. Michel Zwaan
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC-Sophia’s Children’s Hospital, 3015 CN Rotterdam, The Netherlands
| | - Ronald W. Stam
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
41
|
Nakamura A, Masuya M, Shinmei M, Tawara I, Nosaka T, Ono R. Bahcc1 is critical for the aberrant epigenetic program in a mouse model of MLL-ENL-mediated leukemia. Blood Adv 2024; 8:2193-2206. [PMID: 38452334 PMCID: PMC11061229 DOI: 10.1182/bloodadvances.2023011320] [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: 07/27/2023] [Revised: 02/16/2024] [Accepted: 03/06/2024] [Indexed: 03/09/2024] Open
Abstract
ABSTRACT In leukemogenesis, genotoxic stress in hematopoietic stem and progenitor cells (HSPCs) drives individual context-dependent programs of malignant transformation. In light of the various differentiation stages of HSPCs based on a recently revised definition using CD150/CD48, our analyses showed that a subpopulation of long-term repopulating HSCs was most susceptible to MLL-ENL-mediated transformation. An analysis of the molecular mechanism identified Bromo-adjacent homology domain and coiled-coil containing 1 (Bahcc1), which encodes a reader molecule of trimethylated histone H3 lysine 27 (H3K27me3), as a candidate gene involved in distinct susceptibility to leukemic transformation. Interestingly, Bahcc1 was previously reported to be highly expressed in acute myeloid leukemia (AML) with an unfavorable prognosis, including some cases of MLL-rearranged AML. We found that MLL-ENL upregulated Bahcc1 through binding to its promoter, and that Bahcc1 was involved in MLL-ENL-mediated immortalization at least partly through repression of H3K27me3-marked Cdkn1c. Analyses using bone marrow transplantation in mice showed that depletion of Bahcc1 suppressed the leukemogenic activity of MLL-ENL. In a public database, high BAHCC1 expression was found to be associated with a poor prognosis in pediatric AML, in which BAHCC1 expression was significantly lower in MLL-AF9-AML than in other MLL-fusion-AML. These findings shed light on the distinct immortalization potential of HSPCs and suggest a novel MLL-fusion-Bahcc1 axis, which may lead to development of molecular targeted therapy against MLL-fusion-mediated leukemia.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Disease Models, Animal
- Epigenesis, Genetic
- Gene Expression Regulation, Leukemic
- Hematopoietic Stem Cells/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
Collapse
Affiliation(s)
- Akihide Nakamura
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu, Japan
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masahiro Masuya
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Makoto Shinmei
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Tetsuya Nosaka
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Ryoichi Ono
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
42
|
Liu J, Huang XJ. [Progress of allogeneic hematopoietic stem cell transplantation in KMT2A-rearranged acute leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:514-520. [PMID: 38964931 PMCID: PMC11270489 DOI: 10.3760/cma.j.cn121090-20231026-00230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Indexed: 07/06/2024]
Abstract
KMT2A (lysine methyltransferase 2A) -rearranged acute leukemia is a class of leukemia with unique biological characteristics with moderate or poor prognosis. In recent years, allogeneic hematopoietic stem cell transplantation (allo-HSCT) has been increasingly indicated for patients with KMT2A-rearranged acute leukemia. By reviewing the clinical studies of allo-HSCT in KMT2A-rearranged acute leukemia, the efficacy of allo-HSCT in children and adults with KMT2A-rearranged acute myeloid leukemia and acute lymphoblastic leukemia was assessed, the factors affecting the prognosis of allo-HSCT were summarized, and the methods that may improve the outcomes of allo-HSCT were explored.
Collapse
Affiliation(s)
- J Liu
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - X J Huang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| |
Collapse
|
43
|
Mas-Ponte D, Supek F. Mutation rate heterogeneity at the sub-gene scale due to local DNA hypomethylation. Nucleic Acids Res 2024; 52:4393-4408. [PMID: 38587182 PMCID: PMC11077091 DOI: 10.1093/nar/gkae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024] Open
Abstract
Local mutation rates in human are highly heterogeneous, with known variability at the scale of megabase-sized chromosomal domains, and, on the other extreme, at the scale of oligonucleotides. The intermediate, kilobase-scale heterogeneity in mutation risk is less well characterized. Here, by analyzing thousands of somatic genomes, we studied mutation risk gradients along gene bodies, representing a genomic scale spanning roughly 1-10 kb, hypothesizing that different mutational mechanisms are differently distributed across gene segments. The main heterogeneity concerns several kilobases at the transcription start site and further downstream into 5' ends of gene bodies; these are commonly hypomutated with several mutational signatures, most prominently the ubiquitous C > T changes at CpG dinucleotides. The width and shape of this mutational coldspot at 5' gene ends is variable across genes, and corresponds to variable interval of lowered DNA methylation depending on gene activity level and regulation. Such hypomutated loci, at 5' gene ends or elsewhere, correspond to DNA hypomethylation that can associate with various landmarks, including intragenic enhancers, Polycomb-marked regions, or chromatin loop anchor points. Tissue-specific DNA hypomethylation begets tissue-specific local hypomutation. Of note, direction of mutation risk is inverted for AID/APOBEC3 cytosine deaminase activity, whose signatures are enriched in hypomethylated regions.
Collapse
Affiliation(s)
- David Mas-Ponte
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Fran Supek
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
44
|
Yang X, Liu J, Liu W, Wu H, Wei Y, Guo X, Jia H, Can C, Wang D, Hu X, Ma D. circFAM193B interaction with PRMT6 regulates AML leukemia stem cells chemoresistance through altering the oxidative metabolism and lipid peroxidation. Leukemia 2024; 38:1057-1071. [PMID: 38424136 DOI: 10.1038/s41375-024-02189-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
Most forms of chemotherapy for acute myeloid leukemia (AML) are often ineffective in eliminating leukemic stem cells (LSCs), as their underlying mechanisms remain unclear. Here, we have identified circFAM193B, which regulates the redox biology of LSCs and is associated with unfavorable outcomes in AML patients. In vitro and in vivo assays suggested that circFAM193B significantly inhibits LSCs chemotherapy resistance and AML progression. Knockdown circFAM193B enhances mitochondrial OXPHOS function and inhibits the accumulation of reactive oxygen species and lipid peroxidation mediated by chemotherapy, which protects AML cells from oxidative stress-induced cell death. Mechanistically, circFAM193B physically interacts with arginine methyltransferase PRMT6 catalytic domain and enhances the transcription efficiency of key lipid peroxidation factor ALOX15 by decreasing H3R2me2a modification. In summary, we have identified circFAM193B was downregulated in LSCs to promote the survival of LSC by modulating energy metabolism and the redox balance in the postchemotherapy persistence of LSC. Our studies provide a conceptual advance and biological insights regarding the drug resistance of LSCs via circRNA mediated PRMT6-deposited methylarginine signaling.
Collapse
MESH Headings
- Humans
- Protein-Arginine N-Methyltransferases/metabolism
- Protein-Arginine N-Methyltransferases/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Drug Resistance, Neoplasm
- Mice
- Animals
- Lipid Peroxidation
- Oxidative Stress
- Cell Line, Tumor
- Reactive Oxygen Species/metabolism
- Nuclear Proteins
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Jinting Liu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Wancheng Liu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Hanyang Wu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Yihong Wei
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Xiaodong Guo
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Hexiao Jia
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Can Can
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Dongmei Wang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Xiang Hu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
- Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
45
|
Gao Y, Vakoc CR. Therapeutic index of targeting select chromatin complexes in human cancer patients. Curr Opin Genet Dev 2024; 85:102162. [PMID: 38401489 PMCID: PMC11072572 DOI: 10.1016/j.gde.2024.102162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/21/2024] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
Aberrant chromatin regulation can promote the initiation and progression of human cancer. An improved understanding of such mechanisms has resulted in the identification of cancers with an enhanced dependency on specific chromatin regulatory proteins relative to nonmalignant cell types. Hence, targeting of such complexes with small molecules has significant therapeutic potential in oncology. In recent years, several drugs have been developed and evaluated in human cancer patients, which can influence tumor biology by reprogramming of chromatin structure. In this review, we summarize several of the known mechanisms that endow cancer cells with a powerful dependency on chromatin regulation that exceeds the requirements for normal tissue homeostasis. We also summarize the remarkable small-molecule inhibitors that exploit chromatin regulator dependencies with a clear therapeutic benefit in human cancer patients.
Collapse
Affiliation(s)
- Yuan Gao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA. https://twitter.com/@yuangao_yg
| | | |
Collapse
|
46
|
Chen DW, Fan JM, Schrey JM, Mitchell DV, Jung SK, Hurwitz SN, Perez EB, Muraro MJ, Carroll M, Taylor DM, Kurre P. Inflammatory recruitment of healthy hematopoietic stem and progenitor cells in the acute myeloid leukemia niche. Leukemia 2024; 38:741-750. [PMID: 38228679 PMCID: PMC10997516 DOI: 10.1038/s41375-024-02136-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
Inflammation in the bone marrow (BM) microenvironment is a constitutive component of leukemogenesis in acute myeloid leukemia (AML). Current evidence suggests that both leukemic blasts and stroma secrete proinflammatory factors that actively suppress the function of healthy hematopoietic stem and progenitor cells (HSPCs). HSPCs are also cellular components of the innate immune system, and we reasoned that they may actively propagate the inflammation in the leukemic niche. In two separate congenic models of AML we confirm by evaluation of the BM plasma secretome and HSPC-selective single-cell RNA sequencing (scRNA-Seq) that multipotent progenitors and long-lived stem cells adopt inflammatory gene expression programs, even at low leukemic infiltration of the BM. In particular, we observe interferon gamma (IFN-γ) pathway activation, along with secretion of its chemokine target, CXCL10. We show that AML-derived nanometer-sized extracellular vesicles (EVAML) are sufficient to trigger this inflammatory HSPC response, both in vitro and in vivo. Altogether, our studies indicate that HSPCs are an unrecognized component of the inflammatory adaptation of the BM by leukemic cells. The pro-inflammatory conversion and long-lived presence of HSPCs in the BM along with their regenerative re-expansion during remission may impact clonal selection and disease evolution.
Collapse
Affiliation(s)
- Ding-Wen Chen
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jian-Meng Fan
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Julie M Schrey
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dana V Mitchell
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Seul K Jung
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephanie N Hurwitz
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | - Martin Carroll
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Deanne M Taylor
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Peter Kurre
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
47
|
Ling RE, Cross JW, Roy A. Aberrant stem cell and developmental programs in pediatric leukemia. Front Cell Dev Biol 2024; 12:1372899. [PMID: 38601080 PMCID: PMC11004259 DOI: 10.3389/fcell.2024.1372899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/11/2024] [Indexed: 04/12/2024] Open
Abstract
Hematopoiesis is a finely orchestrated process, whereby hematopoietic stem cells give rise to all mature blood cells. Crucially, they maintain the ability to self-renew and/or differentiate to replenish downstream progeny. This process starts at an embryonic stage and continues throughout the human lifespan. Blood cancers such as leukemia occur when normal hematopoiesis is disrupted, leading to uncontrolled proliferation and a block in differentiation of progenitors of a particular lineage (myeloid or lymphoid). Although normal stem cell programs are crucial for tissue homeostasis, these can be co-opted in many cancers, including leukemia. Myeloid or lymphoid leukemias often display stem cell-like properties that not only allow proliferation and survival of leukemic blasts but also enable them to escape treatments currently employed to treat patients. In addition, some leukemias, especially in children, have a fetal stem cell profile, which may reflect the developmental origins of the disease. Aberrant fetal stem cell programs necessary for leukemia maintenance are particularly attractive therapeutic targets. Understanding how hijacked stem cell programs lead to aberrant gene expression in place and time, and drive the biology of leukemia, will help us develop the best treatment strategies for patients.
Collapse
Affiliation(s)
- Rebecca E. Ling
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Joe W. Cross
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Anindita Roy
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Haematology, Great Ormond Street Hospital for Children, London, United Kingdom
| |
Collapse
|
48
|
Chan AK, Han L, Delaney CD, Wang X, Mukhaleva E, Li M, Yang L, Pokharel SP, Mattson N, Garcia M, Wang B, Xu X, Zhang L, Singh P, Elsayed Z, Chen R, Kuang B, Wang J, Yuan YC, Chen B, Chan LN, Rosen ST, Horne D, Müschen M, Chen J, Vaidehi N, Armstrong SA, Su R, Chen CW. Therapeutic targeting Tudor domains in leukemia via CRISPR-Scan Assisted Drug Discovery. SCIENCE ADVANCES 2024; 10:eadk3127. [PMID: 38394203 PMCID: PMC10889360 DOI: 10.1126/sciadv.adk3127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024]
Abstract
Epigenetic dysregulation has been reported in multiple cancers including leukemias. Nonetheless, the roles of the epigenetic reader Tudor domains in leukemia progression and therapy remain unexplored. Here, we conducted a Tudor domain-focused CRISPR screen and identified SGF29, a component of SAGA/ATAC acetyltransferase complexes, as a crucial factor for H3K9 acetylation, ribosomal gene expression, and leukemogenesis. To facilitate drug development, we integrated the CRISPR tiling scan with compound docking and molecular dynamics simulation, presenting a generally applicable strategy called CRISPR-Scan Assisted Drug Discovery (CRISPR-SADD). Using this approach, we identified a lead inhibitor that selectively targets SGF29's Tudor domain and demonstrates efficacy against leukemia. Furthermore, we propose that the structural genetics approach used in our study can be widely applied to diverse fields for de novo drug discovery.
Collapse
Affiliation(s)
- Anthony K.N. Chan
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Division of Epigenetic and Transcriptional Engineering, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Li Han
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Christopher D. Delaney
- Duke University School of Medicine, Durham, NC, USA
- Department of Pediatrics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Xueer Wang
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Elizaveta Mukhaleva
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Mingli Li
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Division of Epigenetic and Transcriptional Engineering, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Sheela Pangeni Pokharel
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Division of Epigenetic and Transcriptional Engineering, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Nicole Mattson
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Michelle Garcia
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Department of Chemistry, Dartmouth College, Hanover, NH, USA
| | - Bintao Wang
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Xiaobao Xu
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Leisi Zhang
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Priyanka Singh
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Zeinab Elsayed
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Renee Chen
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Benjamin Kuang
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Jinhui Wang
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Yate-Ching Yuan
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Bryan Chen
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Lai N. Chan
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - David Horne
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Markus Müschen
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Nagarajan Vaidehi
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, CA, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Scott A. Armstrong
- Department of Pediatrics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Chun-Wei Chen
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Division of Epigenetic and Transcriptional Engineering, Beckman Research Institute, City of Hope, Duarte, CA, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
49
|
Barbosa K, Deshpande A, Perales M, Xiang P, Murad R, Pramod AB, Minkina A, Robertson N, Schischlik F, Lei X, Sun Y, Brown A, Amend D, Jeremias I, Doench JG, Humphries RK, Ruppin E, Shendure J, Mali P, Adams PD, Deshpande AJ. Transcriptional control of leukemogenesis by the chromatin reader SGF29. Blood 2024; 143:697-712. [PMID: 38048593 PMCID: PMC10900139 DOI: 10.1182/blood.2023021234] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/06/2023] Open
Abstract
ABSTRACT Aberrant expression of stem cell-associated genes is a common feature in acute myeloid leukemia (AML) and is linked to leukemic self-renewal and therapy resistance. Using AF10-rearranged leukemia as a prototypical example of the recurrently activated "stemness" network in AML, we screened for chromatin regulators that sustain its expression. We deployed a CRISPR-Cas9 screen with a bespoke domain-focused library and identified several novel chromatin-modifying complexes as regulators of the TALE domain transcription factor MEIS1, a key leukemia stem cell (LSC)-associated gene. CRISPR droplet sequencing revealed that many of these MEIS1 regulators coordinately controlled the transcription of several AML oncogenes. In particular, we identified a novel role for the Tudor-domain-containing chromatin reader protein SGF29 in the transcription of AML oncogenes. Furthermore, SGF29 deletion impaired leukemogenesis in models representative of multiple AML subtypes in multiple AML subtype models. Our studies reveal a novel role for SGF29 as a nononcogenic dependency in AML and identify the SGF29 Tudor domain as an attractive target for drug discovery.
Collapse
Affiliation(s)
- Karina Barbosa
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Anagha Deshpande
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Marlenne Perales
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Ping Xiang
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Rabi Murad
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Akula Bala Pramod
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Anna Minkina
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Neil Robertson
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Fiorella Schischlik
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Xue Lei
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Younguk Sun
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Adam Brown
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Munich, Germany
| | - Diana Amend
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Munich, Germany
| | - Irmela Jeremias
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Munich, Germany
| | | | | | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, San Diego, CA
| | - Peter D. Adams
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Aniruddha J. Deshpande
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| |
Collapse
|
50
|
Weinzapfel EN, Fedder-Semmes KN, Sun ZW, Keogh MC. Beyond the tail: the consequence of context in histone post-translational modification and chromatin research. Biochem J 2024; 481:219-244. [PMID: 38353483 PMCID: PMC10903488 DOI: 10.1042/bcj20230342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
The role of histone post-translational modifications (PTMs) in chromatin structure and genome function has been the subject of intense debate for more than 60 years. Though complex, the discourse can be summarized in two distinct - and deceptively simple - questions: What is the function of histone PTMs? And how should they be studied? Decades of research show these queries are intricately linked and far from straightforward. Here we provide a historical perspective, highlighting how the arrival of new technologies shaped discovery and insight. Despite their limitations, the tools available at each period had a profound impact on chromatin research, and provided essential clues that advanced our understanding of histone PTM function. Finally, we discuss recent advances in the application of defined nucleosome substrates, the study of multivalent chromatin interactions, and new technologies driving the next era of histone PTM research.
Collapse
|