1
|
Zhang H, Pan Y, Wang M, Wang J, Huang J, Ma R, Yang S, Ma W, Yu S, Cui Y. SETD2 regulates oocytes in vitro maturation through histone methylation and maternal mRNA degradation in yak. Theriogenology 2025; 240:117387. [PMID: 40120144 DOI: 10.1016/j.theriogenology.2025.117387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
In vitro maturation (IVM) of oocytes is a vital aspect of assisted reproductive technology (ART), and its proper application can enhance reproductive efficiency. However, owing to the scarcity of research on the IVM of yak oocytes, its application in yak breeding remains underexplored. Therefore, in this study, we conducted high-throughput mRNA sequencing of immature and mature yak oocytes, which revealed transcriptomic changes during the IVM process in this unique high-altitude domesticated animal. Transcriptomic analysis also identified the histone methyltransferase SET domain-containing 2 (SETD2) as a key factor associated with post-translational modifications during oocyte maturation. To determine the role of SETD2 in oocytes, we employed the SETD2 inhibitor EZM0414 during oocyte maturation. Inhibition of SETD2 resulted in a significant reduction in histone methylation levels, lower oocyte maturation rate in vitro, and suppression of maternal mRNAs degradation suppression (P < 0.05). These findings indicated that SETD2 modulates oocyte maturation by regulating histone methylation and maternal mRNAs degradation. Furthermore, suppression of SETD2 markedly reduced the expression of oocyte secretion-related proteins (TSG6 and GDF9) and cumulus expansion-related protein (PTGS2), demonstrating that oocyte secretion and cumulus expansion were positively correlated with SETD2. Overall, our findings establish SETD2 as an essential regulator of yak oocyte maturation via histone methylation and maternal mRNAs degradation.
Collapse
Affiliation(s)
- Hui Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Provincial Livestock Embryo Engineering Technology Innovation Center, Lanzhou, 730070, China
| | - Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Provincial Livestock Embryo Engineering Technology Innovation Center, Lanzhou, 730070, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Provincial Livestock Embryo Engineering Technology Innovation Center, Lanzhou, 730070, China
| | - Jinglei Wang
- Gansu Provincial Livestock Embryo Engineering Technology Innovation Center, Lanzhou, 730070, China
| | - Jiaxin Huang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Rui Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Provincial Livestock Embryo Engineering Technology Innovation Center, Lanzhou, 730070, China
| | - Shanshan Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Provincial Livestock Embryo Engineering Technology Innovation Center, Lanzhou, 730070, China
| | - Wenbin Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Provincial Livestock Embryo Engineering Technology Innovation Center, Lanzhou, 730070, China
| | - Sijiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Provincial Livestock Embryo Engineering Technology Innovation Center, Lanzhou, 730070, China.
| | - Yan Cui
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Provincial Livestock Embryo Engineering Technology Innovation Center, Lanzhou, 730070, China.
| |
Collapse
|
2
|
Cooke EW, Zeng C, Nur SM, Jia Y, Huang A, Chen J, Gao P, Chen FX, Jin F, Cao K. H3K79 methylation and H3K36 tri-methylation synergistically regulate gene expression in pluripotent stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.08.652740. [PMID: 40463105 PMCID: PMC12132225 DOI: 10.1101/2025.05.08.652740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
In metazoans, nucleosomes harboring H3K79 methylation (H3K79me) deposited by the histone methyltransferase DOT1L decorate actively transcribed genes. Although DOT1L is implicated in transcription regulation and pathogenesis of human diseases such as leukemia and neurological disorders, the role of H3K79me in these biological processes remains elusive. Here, we reveal a novel functional synergism between H3K79me and H3K36 tri-methylation (H3K36me3), another histone modification enriched at active genes, in regulating gene expression and neural cell fate transition. Simultaneous catalytic inactivation of DOT1L and the H3K36 methyltransferase SETD2 via gene editing leads to the global loss of H3K79me and H3K36me3, hyperactive transcription, and failures in neural differentiation. Interestingly, the loss of H3K79me and H3K36me3 causes increased transcription elongation, gained chromatin accessibility at a group of enhancers, and increased binding of TEAD4 transcription factor and its co-activator YAP1 at these enhancers. Furthermore, YAP-TEAD inhibition partially restores the expression levels of hyperactivated genes upon H3K79me/H3K36me3 loss. Taken together, our study demonstrates a synergistic role of H3K79me and H3K36me3 in regulating transcription and cell fate transition, unveils novel mechanisms underlying such synergism, and provides insight into designing therapies that target diseases driven by misregulation or mutations of DOT1L and/or SETD2.
Collapse
Affiliation(s)
| | - Cheng Zeng
- Department of Biochemistry, Case Western Reserve University
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Suza Mohammad Nur
- Department of Biochemistry, Case Western Reserve University
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Yunbo Jia
- Department of Biochemistry, Case Western Reserve University
| | - Aileen Huang
- Department of Biochemistry, Case Western Reserve University
| | - Jiwei Chen
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai, China
| | - Peidong Gao
- Department of Genetics and Genome Sciences, Case Western Reserve University
| | - Fei Xavier Chen
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai, China
| | - Fulai Jin
- Department of Genetics and Genome Sciences, Case Western Reserve University
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Kaixiang Cao
- Department of Biochemistry, Case Western Reserve University
- Department of Genetics and Genome Sciences, Case Western Reserve University
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| |
Collapse
|
3
|
Länger ZM, Israel E, Engelhardt J, Kalita AI, Keller Valsecchi CI, Kurtz J, Prohaska SJ. Multiomics Reveal Associations Between CpG Methylation, Histone Modifications and Transcription in a Species That has Lost DNMT3, the Colorado Potato Beetle. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2025. [PMID: 40351084 DOI: 10.1002/jez.b.23303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/07/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
Insects display exceptional phenotypic plasticity, which can be mediated by epigenetic modifications, including CpG methylation and histone modifications. In vertebrates, both are interlinked and CpG methylation is associated with gene repression. However, little is known about these regulatory systems in invertebrates, where CpG methylation is mainly restricted to gene bodies of transcriptionally active genes. A widely conserved mechanism involves the co-transcriptional deposition of H3K36 trimethylation and the targeted methylation of unmethylated CpGs by the de novo DNA methyltransferase DNMT3. However, DNMT3 has been lost multiple times in invertebrate lineages raising the question of how the links between CpG methylation, histone modifications and gene expression are affected by its loss. Here, we report the epigenetic landscape of Leptinotarsa decemlineata, a beetle species that has lost DNMT3 but retained CpG methylation. We combine RNA-seq, enzymatic methyl-seq and CUT&Tag to study gene expression, CpG methylation and patterns of H3K36me3 and H3K27ac histone modifications on a genome-wide scale. Despite the loss of DNMT3, H3K36me3 mirrors CpG methylation patterns. Together, they give rise to signature profiles for expressed and not expressed genes. H3K27ac patterns show a prominent peak at the transcription start site that is predictive of expressed genes irrespective of their methylation status. Our study provides new insights into the evolutionary flexibility of epigenetic modification systems that urge caution when generalizing across species.
Collapse
Affiliation(s)
- Zoe M Länger
- Institute for Evolution and Biodiversity (IEB), University of Münster, Münster, Germany
| | - Elisa Israel
- Computational EvoDevo Group, Institute of Computer Science, Leipzig University, Leipzig, Germany
| | - Jan Engelhardt
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
| | | | | | - Joachim Kurtz
- Institute for Evolution and Biodiversity (IEB), University of Münster, Münster, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), University of Münster and Bielefeld University, Münster, Germany
| | - Sonja J Prohaska
- Computational EvoDevo Group, Institute of Computer Science, Leipzig University, Leipzig, Germany
| |
Collapse
|
4
|
Costantino S, Mohammed SA, Ambrosini S, Telesca M, Mengozzi A, Walavalkar K, Gorica E, Herwig M, van Heerebeek L, Xia J, Karsai G, Hornemann T, Dzemali O, Santoro R, Lin Q, Ruschitzka F, Hamdani N, Paneni F. Chromatin Rewiring by SETD2 Drives Lipotoxic Injury in Cardiometabolic HFpEF. Circ Res 2025; 136:1079-1095. [PMID: 40211947 DOI: 10.1161/circresaha.124.325310] [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: 07/25/2024] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Cardiometabolic heart failure with preserved ejection fraction (cHFpEF) is a highly prevalent and deadly condition. Histone 3 trimethylation at lysine 36 (H3k36me3)-a chromatin signature induced by the histone methyltransferase SETD2 (SET domain containing 2)-correlates with changes in gene expression in human failing hearts; however, its role remains poorly understood. This study investigates the role of SETD2 in cHFpEF. METHODS Chromatin immunoprecipitation sequencing and RNA sequencing were used to investigate H3k36me3-related transcriptional regulation. Mice with cardiomyocyte-specific deletion of SETD2 (c-SETD2-/-) were generated and subjected to high-fat diet feeding and L-NAME treatment for 15 weeks to induce cHFpEF. Cardiac function and exercise tolerance were assessed by echocardiography and treadmill exhaustion test. A selective pharmacological inhibitor of SETD2, EZM0414, was also tested in cHFpEF mice. Mechanistic experiments were performed in cultured cardiomyocytes exposed to palmitic acid. SETD2 signaling and the effects of EZM0414 were also investigated in cardiomyocytes from patients with cHFpEF and control donors. RESULTS SETD2 was upregulated in cHFpEF mouse hearts, and its chromatin mark H3k36me3 was involved in lipid metabolism and highly enriched on the promoter of the Srebf1 gene, encoding for SREBP1 (sterol regulatory binding protein 1). SETD2 activation in cHFpEF led to SREBP1 upregulation, triglyceride accumulation, and lipotoxic damage. Of note, cardiomyocyte-specific deletion of SETD2 in mice prevented heart failure with preserved ejection fraction-related hypertrophy, diastolic dysfunction, and lung congestion while improving exercise tolerance. SETD2 deletion blunted H3K36me3 enrichment on Srebf1 promoter, thus leading to a marked rewiring of the cardiac lipidome and restoration of autophagic flux. In vivo treatment with the SETD2 inhibitor EZM0414 recapitulated the effects of SETD2 deletion. Silencing of SETD2 in palmitic acid-treated cardiomyocytes prevented SREBP1 upregulation, whereas SETD2 overexpression mirrored lipotoxic damage. Finally, SETD2 was upregulated in left ventricle specimens from patients with cHFpEF while EZM0414 attenuated cardiomyocyte stiffness. CONCLUSIONS Targeting SETD2 might prevent lipotoxic injury in cHFpEF.
Collapse
Affiliation(s)
- Sarah Costantino
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
| | - Shafeeq A Mohammed
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
| | - Samuele Ambrosini
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
| | - Marialucia Telesca
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.T.)
| | - Alessandro Mengozzi
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (A.M.)
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa (A.M.)
| | - Kaivalya Walavalkar
- Department of Molecular Mechanisms of Disease, DMMD, University of Zurich, Switzerland (K.W., R.S.), University of Zurich, Switzerland
| | - Era Gorica
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
| | - Melissa Herwig
- Institute of Physiology (M.H., N.H.), Ruhr University, Bochum, Germany
- Molecular and Experimental Cardiology (M.H., N.H.), Ruhr University, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital (M.H., N.H.), Ruhr University, Bochum, Germany
| | | | - Junyan Xia
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, China (J.X., Q.L.)
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zurich (G.K., T.H.), University of Zurich, Switzerland
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zurich (G.K., T.H.), University of Zurich, Switzerland
| | - Omer Dzemali
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- Department of Cardiac Surgery (O.D.), University Hospital Zurich, Switzerland
- Departement of Cardiac Surgery, City Hospital Zurich Triemli, Switzerland (O.D.)
| | - Raffaella Santoro
- Department of Molecular Mechanisms of Disease, DMMD, University of Zurich, Switzerland (K.W., R.S.), University of Zurich, Switzerland
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, China (J.X., Q.L.)
| | - Frank Ruschitzka
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- University Heart Center, Cardiology (F.R., F.P.), University Hospital Zurich, Switzerland
| | - Nazha Hamdani
- Institute of Physiology (M.H., N.H.), Ruhr University, Bochum, Germany
- Molecular and Experimental Cardiology (M.H., N.H.), Ruhr University, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital (M.H., N.H.), Ruhr University, Bochum, Germany
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, Hungary (N.H.)
- Department of Physiology, University Maastricht, the Netherlands (N.H.)
| | - Francesco Paneni
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- University Heart Center, Cardiology (F.R., F.P.), University Hospital Zurich, Switzerland
| |
Collapse
|
5
|
Kesler BK, Adams J, Neuert G. Transcriptional stochasticity reveals multiple mechanisms of long non-coding RNA regulation at the Xist-Tsix locus. Nat Commun 2025; 16:4223. [PMID: 40328749 PMCID: PMC12056010 DOI: 10.1038/s41467-025-59496-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Long noncoding RNAs (LncRNAs) are increasingly recognized as being involved in human physiology and diseases, but there is a lack of mechanistic understanding for the majority of lncRNAs. We comparatively test proposed mechanisms of antisense lncRNA regulation at the X-chromosome Inactivation (XCI) locus. We find that due to stochasticity in transcription, different mechanisms based on the act of transcription regulate Xist and Tsix at different levels of nascent transcription. At medium levels, RNA polymerases transcribe Xist and Tsix on each strand at the same transcription site and deposit significant amounts of the histone mark H3K36me3, which inhibits Xist. At high levels of nascent transcription, many RNA polymerases transcribe Xist or Tsix resulting in transcriptional interference. Therefore, lncRNA expression variability is not just a quirk of transcription but an important aspect of regulation that allows multiple mechanisms to be employed by the same gene locus within the same cell population.
Collapse
Affiliation(s)
- Benjamin K Kesler
- Department of Molecular Physiology and Biophysics, Basic Sciences, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - John Adams
- Department of Molecular Physiology and Biophysics, Basic Sciences, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Gregor Neuert
- Department of Molecular Physiology and Biophysics, Basic Sciences, School of Medicine, Vanderbilt University, Nashville, TN, USA.
- Department of Pharmacology, Basic Sciences, School of Medicine, Vanderbilt University, Nashville, TN, USA.
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Center for Computational Systems Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
6
|
Chen B, Zhang C, Rui H, Shen D, Huang Z, Feng W. Histone H3K36 methyltransferases NSD1 and SETD2 are required for brain development. Hum Genet 2025; 144:529-543. [PMID: 40198378 DOI: 10.1007/s00439-025-02740-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Genetic variants in two major histone H3K36 methyltransferases, NSD1 and SETD2, have been identified in patients with neurodevelopmental disorders. We examined the genetic nature of these disease-relevant variants and studied genotype-phenotype correlations using publicly available patient cohorts. To further investigate roles of Nsd1 and Setd2 in brain development, we generated mouse models with conditional knockout of Nsd1 and Setd2 in neuroepithelial cells using the Sox1-cre. Our results showed that conditional Nsd1 knockout mice were viable but exhibited reduced brain size and thinning of neocortex, while Setd2 knockout led to neonatal death with intracerebral hemorrhage and vascular abnormalities. Together, our study demonstrates new roles of Nsd1 and Setd2 in brain development.
Collapse
Affiliation(s)
- Bo Chen
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Chenyang Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Huanwen Rui
- Department of Neurosurgery, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China
| | - Dan Shen
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhuxi Huang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Weijun Feng
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Key Laboratory of Neonatal Diseases, Xiamen Children's Hospital, Children's Hospital of Fudan University at Xiamen, Xiamen, 361006, China.
| |
Collapse
|
7
|
Yang X, Li L, Qu W, Cheng X, Zhang J, Sun Y, Liu S, Peng G, Zheng R, Li X. Zmynd11 is essential for neurogenesis by coordinating H3K36me3 modification of Epha2 and PI3K signaling pathway. Cell Biosci 2025; 15:55. [PMID: 40281637 PMCID: PMC12032794 DOI: 10.1186/s13578-025-01392-z] [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: 01/24/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
10p15.3 deletion syndrome is caused by the deficiency of MYND-type zinc finger domain-containing protein 11 (ZMYND11) and featured by global developmental delay, intellectual disability, behavioral abnormalities, etc. Although the roles of Zmynd11 is intensively studied in cancer, the function and associated mechanisms of Zmynd11 in neurodevelopment remain largely unknown. Here, we show that Zmynd11 displays abundant and dynamic expression pattern during embryonic neurodevelopment. Zmynd11 deficiency impairs embryonic neurogenesis and neurodevelopment in vitro and in vivo, and inhibits morphological maturation of neurons. Mechanistically, Zmynd11 deficiency leads to decreased Epha2 and disrupts PI3K signaling pathway. Under Zmynd11 deficient condition, H3K36me3 modification on Epha2 promoter abnormally increases and the binding of RNA polymerase II decreases. The restoration of PI3K signaling pathway by exogenous Epha2 can rescue aberrant neurogenesis induced by Zmynd11 depletion in vitro and in vivo. Collectively, our study reveals the essential function of Zmynd11 in neurogenesis via coordinating H3K36me3 modification of Epha2 and PI3K signaling pathway.
Collapse
Affiliation(s)
- Xu Yang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- The Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Lan Li
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- The Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenzheng Qu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xuejun Cheng
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jinyu Zhang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Yan Sun
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Suxiao Liu
- Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Guoping Peng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Rui Zheng
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Xuekun Li
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
- The Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Binjiang Institute of Zhejiang University, Hangzhou, China.
| |
Collapse
|
8
|
Wang X, Wang L, Zhou L, Chen L, Shi J, Ge J, Tian S, Yang Z, Zhou Y, Yu Q, Jin J, Ding C, Pan Y, Zou D. NUP62 alleviates senescence and promotes the stemness of human dental pulp stem cells via NSD2-dependent epigenetic reprogramming. Int J Oral Sci 2025; 17:34. [PMID: 40246825 PMCID: PMC12006529 DOI: 10.1038/s41368-025-00362-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: 05/25/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Stem cells play a crucial role in maintaining tissue regenerative capacity and homeostasis. However, mechanisms associated with stem cell senescence require further investigation. In this study, we conducted a proteomic analysis of human dental pulp stem cells (HDPSCs) obtained from individuals of various ages. Our findings showed that the expression of NUP62 was decreased in aged HDPSCs. We discovered that NUP62 alleviated senescence-associated phenotypes and enhanced differentiation potential both in vitro and in vivo. Conversely, the knocking down of NUP62 expression aggravated the senescence-associated phenotypes and impaired the proliferation and migration capacity of HDPSCs. Through RNA-sequence and decoding the epigenomic landscapes remodeled induced by NUP62 overexpression, we found that NUP62 helps alleviate senescence in HDPSCs by enhancing the nuclear transport of the transcription factor E2F1. This, in turn, stimulates the transcription of the epigenetic enzyme NSD2. Finally, the overexpression of NUP62 influences the H3K36me2 and H3K36me3 modifications of anti-aging genes (HMGA1, HMGA2, and SIRT6). Our results demonstrated that NUP62 regulates the fate of HDPSCs via NSD2-dependent epigenetic reprogramming.
Collapse
Affiliation(s)
- Xiping Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Li Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Linxi Zhou
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lu Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiayi Shi
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jing Ge
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Sha Tian
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Zihan Yang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yuqiong Zhou
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Qihao Yu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jiacheng Jin
- Touro College of Dental Medicine, New York Medical College, New York, USA
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yihuai Pan
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
- Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| | - Duohong Zou
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China.
| |
Collapse
|
9
|
Huang Z, Hu L, Liu Z, Wang S. The Functions and Regulatory Mechanisms of Histone Modifications in Skeletal Muscle Development and Disease. Int J Mol Sci 2025; 26:3644. [PMID: 40332229 PMCID: PMC12027200 DOI: 10.3390/ijms26083644] [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: 03/03/2025] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Skeletal muscle development is a complex biological process regulated by many factors, such as transcription factors, signaling pathways, and epigenetic modifications. Histone modifications are important epigenetic regulatory factors involved in various biological processes, including skeletal muscle development, and play a crucial role in the pathogenesis of skeletal muscle diseases. Histone modification regulators affect the expression of many genes involved in skeletal muscle development and disease by adding or removing certain chemical modifications. In this review, we comprehensively summarize the functions and regulatory activities of the histone modification regulators involved in skeletal muscle development, regeneration, and disease.
Collapse
Affiliation(s)
- Zining Huang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan 430062, China; (Z.H.); (L.H.)
| | - Linqing Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan 430062, China; (Z.H.); (L.H.)
| | - Zhiwei Liu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Shanshan Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan 430062, China; (Z.H.); (L.H.)
| |
Collapse
|
10
|
Mancini M, De Santis S, Monaldi C, Castagnetti F, Iezza M, Iurlo A, Cattaneo D, Galimberti S, Cerrano M, Capodanno I, Bonifacio M, Rossi M, Agostinelli C, Meggendorfer M, Haferlach T, Cavo M, Gugliotta G, Soverini S. SETD2 loss of function is a recurrent event in advanced-phase chronic myeloid leukemia and contributes to genomic instability: SETD2 loss in Chronic Myeloid Leukemia. Clin Transl Med 2025; 15:e70163. [PMID: 40275711 PMCID: PMC12022228 DOI: 10.1002/ctm2.70163] [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: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 04/26/2025] Open
Abstract
The SETD2 tumour suppressor encodes a histone methyltransferase that specifically trimethylates histone H3 on lysine 36 (H3K36me3), a key histone mark implicated in the maintenance of genomic integrity among other functions. We found that SETD2 protein deficiency, mirrored by H3K36me3 deficiency, is a nearly universal event in advanced-phase chronic myeloid leukemia (CML) patients. Similarly, K562 and KCL22 cell lines exhibited markedly reduced or undetectable SETD2/H3K36me3 levels, respectively. This resulted from altered SETD2 protein turnover rather than mutations or transcriptional downregulation, and proteasome inhibition led to the accumulation of hyper-ubiquitinated SETD2 and to H3K36me3 rescue suggesting that a functional SETD2 protein is produced but abnormally degraded. We demonstrated that phosphorylation by Aurora-A kinase and ubiquitination by MDM2 plays a key role in the proteasome-mediated degradation of SETD2. Moreover, we found that SETD2 and H3K36me3 loss impinges on the activation and proficiency of homologous recombination and mismatch repair. Finally, we showed that proteasome and Aurora-A kinase inhibitors, acting via SETD2/H3K36me3 rescue, are effective in inducing apoptosis and reducing clonogenic growth in cell lines and primary cells from advanced-phase patients. Taken together, our results point to SETD2/H3K36me3 deficiency as a mechanism, already identified by our group in systemic mastocytosis, that is reversible, druggable, and BCR::ABL1-independent, able to cooperate with BCR::ABL1 in driving genetic instability in CML. KEY POINTS: Virtually all CML patients in blast crisis display SETD2 loss of function. SETD2 loss seems to be accomplished at the posttranslational level rather than being the result of genetic/genomic hits or transcriptional repression. Phosphorylation by Aurora kinase A and ubiquitination by MDM2 contribute to SETD2 proteasome-mediated degradation in blast crisis CML patients. Loss of SETD2 results in increased DNA damage.
Collapse
Affiliation(s)
- Manuela Mancini
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaIstituto di Ematologia “Seràgnoli”BolognaItaly
| | - Sara De Santis
- Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Cecilia Monaldi
- Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Fausto Castagnetti
- Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Miriam Iezza
- Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Alessandra Iurlo
- Hematology DivisionFoundation IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanoItaly
| | - Daniele Cattaneo
- Hematology DivisionFoundation IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanoItaly
- Department of Oncology and Hemato‐OncologyUniversity of MilanMilanoItaly
| | - Sara Galimberti
- Clinical and Experimental MedicineHematologyUniversity of PisaPisaItaly
| | - Marco Cerrano
- Azienda Ospedaliera Citta' Della Salute E Della Scienza Di TorinoTorinoItaly
| | | | - Massimiliano Bonifacio
- Section of HematologyDepartment of MedicineAzienda Ospedaliera Universitaria Integrata di VeronaVeronaItaly
| | - Maura Rossi
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaIstituto di Ematologia “Seràgnoli”BolognaItaly
| | - Claudio Agostinelli
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaIstituto di Ematologia “Seràgnoli”BolognaItaly
- Haematopathology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | | | | | - Michele Cavo
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaIstituto di Ematologia “Seràgnoli”BolognaItaly
| | - Gabriele Gugliotta
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaIstituto di Ematologia “Seràgnoli”BolognaItaly
| | - Simona Soverini
- Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| |
Collapse
|
11
|
Lucain M, Vitobello A, Sadikovic B, Albuisson J, Gaudillat L, Chevarin M, Maraval J, Thauvin-Robinet C, Kerkhof J, Philippe C, Nambot S, Faivre L. Abnormal DNA Methylation Profile Suggests the Extension of the Clinical Spectrum of the SETD2-Related Disorders to a Syndromic Multiple Tumor Phenotype. Am J Med Genet A 2025:e64043. [PMID: 40104911 DOI: 10.1002/ajmg.a.64043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 01/27/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
SETD2 has an essential role in epigenetic regulation. SETD2 pathogenic variants cause neurodevelopmental disorders (SETD2-NDDs) that most commonly include various degrees of intellectual disability and behavioral disorders, macrocephaly, brain malformations, and generalized overgrowth. A distinctive DNA methylation episignature has been identified for Luscan-Lumish syndrome. A less common phenotype, denoted SETD2-NDD with multiple congenital anomalies, failure to thrive, and profound intellectual disability, has been reported in association with a particular pathogenic variant (p.Arg1740Trp). To date, about 50 patients have been described in the literature with SETD2 causative variants. We report here an individual with a phenotype distinct from SETD2-NDDs, including normal cognition, distinctive facial features, and multiple tumor histories, including a sacral osteoblastoma at age 7, a benign femoral bone tumor at age 17, a peritoneal pseudomyxoma at age 27, and a hypophyseal macroadenoma and a low-grade optochiasmatic glioma at age 37 years. Trio exome sequencing identified a de novo heterozygous missense variant of unknown significance (p.Ser1658Leu) in the SETD2 gene. DNA methylation study by EpiSign assay confirmed the presence of an episignature profile compatible with SETD2-related disorders. Given the implication of somatic SETD2 variants in benign and malignant tumors, the implication of these SETD2 constitutional variants in tumorigenesis is discussed.
Collapse
Affiliation(s)
- Marie Lucain
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
| | - Antonio Vitobello
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
| | - Bekim Sadikovic
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, Canada, Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | | | - Léa Gaudillat
- Centre de Génétique, Centre de Référence Maladies Rares "Anomalies du Développement et Syndromes Malformatifs", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Martin Chevarin
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
| | - Julien Maraval
- Centre de Génétique, Centre de Référence Maladies Rares "Anomalies du Développement et Syndromes Malformatifs", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Christel Thauvin-Robinet
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
- Centre de Référence Maladies Rares "Déficiences Intellectuelles de Causes Rares", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Jennifer Kerkhof
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, Canada, Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Christophe Philippe
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
| | - Sophie Nambot
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
- Centre de Référence Maladies Rares "Déficiences Intellectuelles de Causes Rares", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Laurence Faivre
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
- Centre de Génétique, Centre de Référence Maladies Rares "Anomalies du Développement et Syndromes Malformatifs", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| |
Collapse
|
12
|
Bahia RK, Lopez C, Nardocci G, Davie JR. Differential H3K4me3 Domains in Normal and Colorectal Cancer Cells Reveal Novel Epigenetic Targets. Int J Mol Sci 2025; 26:2546. [PMID: 40141189 PMCID: PMC11942224 DOI: 10.3390/ijms26062546] [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: 02/11/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Histone H3 trimethylated at lysine 4 (H3K4me3) is an histone mark associated with transcriptionally active genes. H3K4me3 has two types of distribution: a sharp distribution of approximately 500 bp and a broad H3K4me3 domain that may extend 4 kb and longer through the gene body. Most transcribed genes have a narrow H3K4me3 configuration, whereas genes involved in cell identity and tumor suppression have a broad arrangement in normal cells. In cancer cells, genes that promote cancer possess a broad H3K4me3 domain. In this study, we performed H3K4me3 chromatin immunoprecipitation sequencing to determine the genes with narrow and broad H3K4me3 configurations in normal colon epithelial cells and three colon cancer cell lines. The analysis revealed that genes involved in cell adhesion and nervous system development had an H3K4me3 peak next to their transcription start site in normal cells but not in colon cancer cells. Genes coding for long non-coding RNA (lncRNA) were differentially marked with a broad H3K4me3 domain in normal colon versus colon cancer cells (FENDRR in normal colon; ELFN1-AS1 in colon cancer). Identifying the genes that are silenced or activated, particularly in colon cancer, provides a list of actionable targets for designing effective treatments for this prevalent human disease.
Collapse
Affiliation(s)
- Ravinder Kaur Bahia
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Camila Lopez
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Gino Nardocci
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile
- Molecular Biology and Bioinformatics Lab, Program in Molecular Biology and Bioinformatics, Center for Biomedical Research and Innovation (CIIB), Universidad de los Andes, Santiago 7620001, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7620001, Chile
| | - James R. Davie
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
13
|
Wagner RT, Hlady RA, Pan X, Wang L, Kim S, Zhao X, El Khoury LY, Shaikh S, Zhong J, Lee JH, Grembecka J, Cierpicki T, Ho TH, Robertson KD. SETD2 loss-of-function uniquely sensitizes cells to epigenetic targeting of NSD1-directed H3K36 methylation. Genome Biol 2025; 26:22. [PMID: 39910618 PMCID: PMC11800516 DOI: 10.1186/s13059-025-03483-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND SETD2 is the sole epigenetic factor responsible for catalyzing histone 3, lysine 36, tri-methylation (H3K36me3) in mammals. Its role in regulating cellular processes such as RNA splicing, DNA repair, and spurious transcription initiation underlies its broader tumor suppressor function. SETD2 mutation promotes the epithelial-mesenchymal transition and is clinically associated with adverse outcomes highlighting a therapeutic need to develop targeted therapies against this dangerous mutation. RESULTS We employ an unbiased genome-wide synthetic lethal screen, which identifies another H3K36me writer, NSD1, as a synthetic lethal modifier in SETD2-mutant cells. Confirmation of this synthetic lethal interaction is performed in isogenic clear cell renal cell carcinoma and immortalized renal epithelial cell lines, in mouse and human backgrounds. Depletion of NSD1 using a CRISPRi targeting approach promotes the loss of SETD2-mutant cells coincident with elevated levels of DNA damage and apoptosis. Surprisingly, only suppression of NSD1, but not related H3K36-methyltransferases, promotes synthetic lethality in these models. Mapping of genomic H3K36me2 targeting by NSD1 and NSD2 individually highlights the independent functions of these epigenetic writers. Furthermore, as a proof-of-principle, we demonstrate the therapeutic feasibility of targeting this synthetic lethal interaction by recapitulating the phenotype using BT5, a first-in-class pharmacologic inhibitor against NSD1. CONCLUSIONS These findings unify genome-wide screening approaches with the latest genetic and pharmacologic modeling methodologies to reveal an entirely novel epigenetic approach to individualize therapies against a challenging loss-of-function SETD2 mutation in cancer.
Collapse
Affiliation(s)
- Ryan T Wagner
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Ryan A Hlady
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Xiaoyu Pan
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Sungho Kim
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Xia Zhao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Louis Y El Khoury
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Shafiq Shaikh
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jian Zhong
- Epigenomics Development Laboratory, Mayo Clinic, CIM Epigenomics Program, Rochester, MN, USA
| | - Jeong-Heon Lee
- Epigenomics Development Laboratory, Mayo Clinic, CIM Epigenomics Program, Rochester, MN, USA
| | | | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Thai H Ho
- Division of Hematology and Oncology, Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA.
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA.
| |
Collapse
|
14
|
Jin YJ, Liang G, Li R, Wang S, Alnouri MW, Bentsen M, Kuenne C, Günther S, Yan Y, Li Y, Wettschureck N, Offermanns S. Phosphorylation of endothelial histone H3.3 serine 31 by PKN1 links flow-induced signaling to proatherogenic gene expression. NATURE CARDIOVASCULAR RESEARCH 2025; 4:180-196. [PMID: 39779823 PMCID: PMC11825370 DOI: 10.1038/s44161-024-00593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025]
Abstract
Atherosclerotic lesions develop preferentially in arterial regions exposed to disturbed blood flow, where endothelial cells acquire an inflammatory phenotype. How disturbed flow induces endothelial cell inflammation is incompletely understood. Here we show that histone H3.3 phosphorylation at serine 31 (H3.3S31) regulates disturbed-flow-induced endothelial inflammation by allowing rapid induction of FOS and FOSB, required for inflammatory gene expression. We identified protein kinase N1 (PKN1) as the kinase responsible for disturbed-flow-induced H3.3S31 phosphorylation. Disturbed flow activates PKN1 in an integrin α5β1-dependent manner and induces its translocation into the nucleus, and PKN1 is also involved in the phosphorylation of the AP-1 transcription factor JUN. Mice with endothelium-specific PKN1 loss or endothelial expression of S31 phosphorylation-deficient H.3.3 mutants show reduced endothelial inflammation and disturbed-flow-induced vascular remodeling in vitro and in vivo. Together, we identified a pathway whereby disturbed flow through PKN1-mediated histone phosphorylation and FOS/FOSB induction promotes inflammatory gene expression and vascular inflammation.
Collapse
Affiliation(s)
- Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Guozheng Liang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rui Li
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mohamad Wessam Alnouri
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mette Bentsen
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Carsten Kuenne
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Yang Yan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yongxin Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany
- Cardiopulmonary Institute (CPI), Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany.
- Cardiopulmonary Institute (CPI), Frankfurt, Germany.
- German Center for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.
| |
Collapse
|
15
|
Lee MK, Park NH, Lee SY, Kim T. Context-Dependent and Locus-Specific Role of H3K36 Methylation in Transcriptional Regulation. J Mol Biol 2025; 437:168796. [PMID: 39299382 DOI: 10.1016/j.jmb.2024.168796] [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: 06/17/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
H3K36 methylation is a critical histone modification involved in transcription regulation. It involves the mono (H3K36me1), di (H3K36me2), and/or tri-methylation (H3K36me3) of lysine 36 on histone H3 by methyltransferases. In yeast, Set2 catalyzes all three methylation states. By contrast, in higher eukaryotes, at least eight methyltransferases catalyze different methylation states, including SETD2 for H3K36me3 and the NSD family for H3K36me2 in vivo. Both Set2 and SETD2 interact with the phosphorylated CTD of RNA Pol II, which links H3K36 methylation to transcription. In yeast, H3K36me3 and H3K36me2 peak at the 3' ends of genes. In higher eukaryotes, this is also true for H3K36me3 but not for H3K36me2, which is enriched at the 5' ends of genes and intergenic regions, suggesting that H3K36me2 and H3K36me3 may play different regulatory roles. Whether H3K36me1 demonstrates preferential distribution remains unclear. H3K36me3 is essential for inhibiting transcription elongation. It also suppresses cryptic transcription by promoting histone deacetylation by the histone deacetylases Rpd3S (yeast) and variant NuRD (higher eukaryotes). H3K36me3 also facilitates DNA methylation by DNMT3B, thereby preventing spurious transcription initiation. H3K36me3 not only represses transcription since it promotes the activation of mRNA and cryptic promoters in response to environmental changes by targeting the histone acetyltransferase NuA3 in yeast. Further research is needed to elucidate the methylation state- and locus-specific functions of H3K36me1 and the mechanisms that regulate it.
Collapse
Affiliation(s)
- Min Kyung Lee
- Department of Life Sciences and Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Na Hyun Park
- Department of Life Sciences and Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Soo Young Lee
- Department of Life Sciences and Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Republic of Korea
| | - TaeSoo Kim
- Department of Life Sciences and Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
16
|
Shipman GA, Padilla R, Horth C, Hu B, Bareke E, Vitorino FN, Gongora JM, Garcia BA, Lu C, Majewski J. Systematic perturbations of SETD2, NSD1, NSD2, NSD3, and ASH1L reveal their distinct contributions to H3K36 methylation. Genome Biol 2024; 25:263. [PMID: 39390582 PMCID: PMC11465688 DOI: 10.1186/s13059-024-03415-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Methylation of histone 3 lysine 36 (H3K36me) has emerged as an essential epigenetic component for the faithful regulation of gene expression. Despite its importance in development and disease, how the molecular agents collectively shape the H3K36me landscape is unclear. RESULTS We use mouse mesenchymal stem cells to perturb the H3K36me methyltransferases (K36MTs) and infer the activities of the five most prominent enzymes: SETD2, NSD1, NSD2, NSD3, and ASH1L. We find that H3K36me2 is the most abundant of the three methylation states and is predominantly deposited at intergenic regions by NSD1, and partly by NSD2. In contrast, H3K36me1/3 are most abundant within exons and are positively correlated with gene expression. We demonstrate that while SETD2 deposits most H3K36me3, it may also deposit H3K36me2 within transcribed genes. Additionally, loss of SETD2 results in an increase of exonic H3K36me1, suggesting other (K36MTs) prime gene bodies with lower methylation states ahead of transcription. While NSD1/2 establish broad intergenic H3K36me2 domains, NSD3 deposits H3K36me2 peaks on active promoters and enhancers. Meanwhile, the activity of ASH1L is restricted to the regulatory elements of developmentally relevant genes, and our analyses implicate PBX2 as a potential recruitment factor. CONCLUSIONS Within genes, SETD2 primarily deposits H3K36me3, while the other K36MTs deposit H3K36me1/2 independently of SETD2 activity. For the deposition of H3K36me1/2, we find a hierarchy of K36MT activities where NSD1 > NSD2 > NSD3 > ASH1L. While NSD1 and NSD2 are responsible for most genome-wide propagation of H3K36me2, the activities of NSD3 and ASH1L are confined to active regulatory elements.
Collapse
Affiliation(s)
- Gerry A Shipman
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1B1, Canada
- McGill University Genome Centre, Montreal, QC, H3A 0G1, Canada
| | - Reinnier Padilla
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1B1, Canada
- McGill University Genome Centre, Montreal, QC, H3A 0G1, Canada
| | - Cynthia Horth
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1B1, Canada
- McGill University Genome Centre, Montreal, QC, H3A 0G1, Canada
| | - Bo Hu
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1B1, Canada
- McGill University Genome Centre, Montreal, QC, H3A 0G1, Canada
| | - Eric Bareke
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1B1, Canada
- McGill University Genome Centre, Montreal, QC, H3A 0G1, Canada
| | - Francisca N Vitorino
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joanna M Gongora
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Benjamin A Garcia
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chao Lu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jacek Majewski
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1B1, Canada.
- McGill University Genome Centre, Montreal, QC, H3A 0G1, Canada.
| |
Collapse
|
17
|
Chen S, Liu D, Chen B, Li Z, Chang B, Xu C, Li N, Feng C, Hu X, Wang W, Zhang Y, Xie Y, Huang Q, Wang Y, Nimer SD, Chen S, Chen Z, Wang L, Sun X. Catalytic activity of Setd2 is essential for embryonic development in mice: establishment of a mouse model harboring patient-derived Setd2 mutation. Front Med 2024; 18:831-849. [PMID: 39115793 DOI: 10.1007/s11684-024-1095-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/08/2024] [Indexed: 11/01/2024]
Abstract
SETD2 is the only enzyme responsible for transcription-coupled histone H3 lysine 36 trimethylation (H3K36me3). Mutations in SETD2 cause human diseases including cancer and developmental defects. In mice, Setd2 is essential for embryonic vascular remodeling. Given that many epigenetic modifiers have recently been found to possess noncatalytic functions, it is unknown whether the major function(s) of Setd2 is dependent on its catalytic activity or not. Here, we established a site-specific knockin mouse model harboring a cancer patient-derived catalytically dead Setd2 (Setd2-CD). We found that the essentiality of Setd2 in mouse development is dependent on its methyltransferase activity, as the Setd2CD/CD and Setd2-/- mice showed similar embryonic lethal phenotypes and largely comparable gene expression patterns. However, compared with Setd2-/-, the Setd2CD/CD mice showed less severe defects in allantois development, and single-cell RNA-seq analysis revealed differentially regulated allantois-specific 5' Hoxa cluster genes in these two models. Collectively, this study clarifies the importance of Setd2 catalytic activity in mouse development and provides a new model for comparative study of previously unrecognized Setd2 functions.
Collapse
Affiliation(s)
- Shubei Chen
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Dianjia Liu
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Bingyi Chen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zijuan Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Binhe Chang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chunhui Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ningzhe Li
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Changzhou Feng
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
- Department of Clinical Laboratory, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222000, China
| | - Xibo Hu
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Weiying Wang
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yuanliang Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yinyin Xie
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Qiuhua Huang
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yingcai Wang
- Sylvester Comprehensive Cancer Center and Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Stephen D Nimer
- Sylvester Comprehensive Cancer Center and Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Saijuan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Zhu Chen
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Lan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiaojian Sun
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
18
|
Weng Y, Xue J, Niu N. SETD2 in cancer: functions, molecular mechanisms, and therapeutic regimens. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0201. [PMID: 39302028 PMCID: PMC11414219 DOI: 10.20892/j.issn.2095-3941.2024.0201] [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: 06/06/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024] Open
Affiliation(s)
- Yawen Weng
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jing Xue
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ningning Niu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| |
Collapse
|
19
|
Ma S, Long G, Jiang Z, Zhang Y, Sun L, Pan Y, You Q, Guo X. Recent advances in targeting histone H3 lysine 36 methyltransferases for cancer therapy. Eur J Med Chem 2024; 274:116532. [PMID: 38805937 DOI: 10.1016/j.ejmech.2024.116532] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
Histone H3 lysine 36 (H3K36) methylation is a typical epigenetic histone modification that is involved in various biological processes such as DNA transcription, repair and recombination in vivo. Mutations, translocations, and aberrant gene expression associated with H3K36 methyltransferases have been implicated in different malignancies such as acute myeloid leukemia, lung cancer, multiple myeloma, and others. Herein, we provided a comprehensive overview of the latest advances in small molecule inhibitors targeting H3K36 methyltransferases. We analyzed the structures and biological functions of the H3K36 methyltransferases family members. Additionally, we discussed the potential directions for future development of inhibitors targeting H3K36 methyltransferases.
Collapse
Affiliation(s)
- Sai Ma
- Jiangsu Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Guanlu Long
- Jiangsu Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Zheng Jiang
- Jiangsu Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Yan Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Liangkui Sun
- Jiangsu Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Yun Pan
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- Jiangsu Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiaoke Guo
- Jiangsu Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
20
|
Park J, Wu J, Szkop KJ, Jeong J, Jovanovic P, Husmann D, Flores NM, Francis JW, Chen YJC, Benitez AM, Zahn E, Song S, Ajani JA, Wang L, Singh K, Larsson O, Garcia BA, Topisirovic I, Gozani O, Mazur PK. SMYD5 methylation of rpL40 links ribosomal output to gastric cancer. Nature 2024; 632:656-663. [PMID: 39048817 PMCID: PMC11625416 DOI: 10.1038/s41586-024-07718-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
Dysregulated transcription due to disruption in histone lysine methylation dynamics is an established contributor to tumorigenesis1,2. However, whether analogous pathologic epigenetic mechanisms act directly on the ribosome to advance oncogenesis is unclear. Here we find that trimethylation of the core ribosomal protein L40 (rpL40) at lysine 22 (rpL40K22me3) by the lysine methyltransferase SMYD5 regulates mRNA translation output to promote malignant progression of gastric adenocarcinoma (GAC) with lethal peritoneal ascites. A biochemical-proteomics strategy identifies the monoubiquitin fusion protein partner rpL40 (ref. 3) as the principal physiological substrate of SMYD5 across diverse samples. Inhibiting the SMYD5-rpL40K22me3 axis in GAC cell lines reprogrammes protein synthesis to attenuate oncogenic gene expression signatures. SMYD5 and rpL40K22me3 are upregulated in samples from patients with GAC and negatively correlate with clinical outcomes. SMYD5 ablation in vivo in familial and sporadic mouse models of malignant GAC blocks metastatic disease, including peritoneal carcinomatosis. Suppressing SMYD5 methylation of rpL40 inhibits human cancer cell and patient-derived GAC xenograft growth and renders them hypersensitive to inhibitors of PI3K and mTOR. Finally, combining SMYD5 depletion with PI3K-mTOR inhibition and chimeric antigen receptor T cell administration cures an otherwise lethal in vivo mouse model of aggressive GAC-derived peritoneal carcinomatosis. Together, our work uncovers a ribosome-based epigenetic mechanism that facilitates the evolution of malignant GAC and proposes SMYD5 targeting as part of a potential combination therapy to treat this cancer.
Collapse
Affiliation(s)
- Juhyung Park
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jibo Wu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Krzysztof J Szkop
- Department of Oncology-Pathology, Science for Life Laboratories, Karolinska Institute, Stockholm, Sweden
| | - Jinho Jeong
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Predrag Jovanovic
- Lady Davis Institute and Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Dylan Husmann
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Natasha M Flores
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joel W Francis
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Ying-Jiun C Chen
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Morales Benitez
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emily Zahn
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kamini Singh
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Montefiore Einstein Cancer Center, Bronx, NY, USA
| | - Ola Larsson
- Department of Oncology-Pathology, Science for Life Laboratories, Karolinska Institute, Stockholm, Sweden
| | - Benjamin A Garcia
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, USA
| | - Ivan Topisirovic
- Lady Davis Institute and Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Or Gozani
- Department of Biology, Stanford University, Stanford, CA, USA.
| | - Pawel K Mazur
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
21
|
Boulanger C, Haidara N, Yague-Sanz C, Larochelle M, Jacques PÉ, Hermand D, Bachand F. Repression of pervasive antisense transcription is the primary role of fission yeast RNA polymerase II CTD serine 2 phosphorylation. Nucleic Acids Res 2024; 52:7572-7589. [PMID: 38801067 PMCID: PMC11260464 DOI: 10.1093/nar/gkae436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
The RNA polymerase II carboxy-terminal domain (CTD) consists of conserved heptapeptide repeats that can be phosphorylated to influence distinct stages of the transcription cycle, including RNA processing. Although CTD-associated proteins have been identified, phospho-dependent CTD interactions have remained elusive. Proximity-dependent biotinylation (PDB) has recently emerged as an alternative approach to identify protein-protein associations in the native cellular environment. In this study, we present a PDB-based map of the fission yeast RNAPII CTD interactome in living cells and identify phospho-dependent CTD interactions by using a mutant in which Ser2 was replaced by alanine in every repeat of the fission yeast CTD. This approach revealed that CTD Ser2 phosphorylation is critical for the association between RNAPII and the histone methyltransferase Set2 during transcription elongation, but is not required for 3' end processing and transcription termination. Accordingly, loss of CTD Ser2 phosphorylation causes a global increase in antisense transcription, correlating with elevated histone acetylation in gene bodies. Our findings reveal that the fundamental role of CTD Ser2 phosphorylation is to establish a chromatin-based repressive state that prevents cryptic intragenic transcription initiation.
Collapse
Affiliation(s)
- Cédric Boulanger
- RNA Group, Dept of Biochemistry & Functional Genomics, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Nouhou Haidara
- RNA Group, Dept of Biochemistry & Functional Genomics, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Carlo Yague-Sanz
- URPHYM-GEMO, The University of Namur, rue de Bruxelles, 61, Namur 5000, Belgium
| | - Marc Larochelle
- RNA Group, Dept of Biochemistry & Functional Genomics, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | | | - Damien Hermand
- URPHYM-GEMO, The University of Namur, rue de Bruxelles, 61, Namur 5000, Belgium
- The Francis Crick Institute, 1 Midland Road London NW1 1AT, UK
| | - Francois Bachand
- RNA Group, Dept of Biochemistry & Functional Genomics, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| |
Collapse
|
22
|
Chen L, Zhang M, Yang X, Wang Y, Huang T, Li X, Ban Y, Li Q, Yang Q, Zhang Y, Zheng Y, Wang D, Wang X, Shi X, Zhang M, Sun Y, Wu J. Methyl-CpG-binding 2 K271 lactylation-mediated M2 macrophage polarization inhibits atherosclerosis. Theranostics 2024; 14:4256-4277. [PMID: 39113793 PMCID: PMC11303070 DOI: 10.7150/thno.94738] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/18/2024] [Indexed: 08/10/2024] Open
Abstract
Rationale: Posttranslational modifications of proteins have not been addressed in studies aimed at elucidating the cardioprotective effect of exercise in atherosclerotic cardiovascular disease (ASCVD). In this study, we reveal a novel mechanism by which exercise ameliorates atherosclerosis via lactylation. Methods: Using ApoE-/- mice in an exercise model, proteomics analysis was used to identify exercise-induced specific lactylation of MeCP2 at lysine 271 (K271). Mutation of the MeCP2 K271 lactylation site in aortic plaque macrophages was achieved by recombinant adenoviral transfection. Explore the molecular mechanisms by which motility drives MeCP2 K271 lactylation to improve plaque stability using ATAC-Seq, CUT &Tag and molecular biology. Validation of the potential target RUNX1 for exercise therapy using Ro5-3335 pharmacological inhibition. Results: we showed that in ApoE-/- mice, methyl-CpG-binding protein 2 (MeCP2) K271 lactylation was observed in aortic root plaque macrophages, promoting pro-repair M2 macrophage polarization, reducing the plaque area, shrinking necrotic cores, reducing plaque lipid deposition, and increasing collagen content. Adenoviral transfection, by introducing a mutant at lysine 271, overexpressed MeCP2 K271 lactylation, which enhanced exercise-induced M2 macrophage polarization and increased plaque stability. Mechanistically, the exercise-induced atheroprotective effect requires an interaction between MeCP2 K271 lactylation and H3K36me3, leading to increased chromatin accessibility and transcriptional repression of RUNX1. In addition, the pharmacological inhibition of the transcription factor RUNX1 exerts atheroprotective effects by promoting the polarization of plaque macrophages towards the pro-repair M2 phenotype. Conclusions: These findings reveal a novel mechanism by which exercise ameliorates atherosclerosis via MeCP2 K271 lactylation-H3K36me3/RUNX1. Interventions that enhance MeCP2 K271 lactylation have been shown to increase pro-repair M2 macrophage infiltration, thereby promoting plaque stabilization and reducing the risk of atherosclerotic cardiovascular disease. We also established RUNX1 as a potential drug target for exercise therapy, thereby providing guidance for the discovery of new targets.
Collapse
Affiliation(s)
- Liangqi Chen
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Meiju Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Xueyan Yang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yanan Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Tuo Huang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Xin Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yunting Ban
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Qifeng Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Qingyuan Yang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yongxiang Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yang Zheng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Di Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Xiaoqi Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Xiujie Shi
- The Clinical Skills Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Maomao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yong Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Jian Wu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
- Cardiac Rehabilitation Center, Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
23
|
Pathak R, Esnault C, Radhakrishnan R, Singh PK, Zhang H, Dale R, Anand A, Bedwell GJ, Engelman AN, Rabi A, Hormoz S, Singh P, Levin HL. The role of LEDGF in transcription is exploited by HIV-1 to position integration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.29.601340. [PMID: 39005447 PMCID: PMC11244883 DOI: 10.1101/2024.06.29.601340] [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
HIV-1 integration occurs across actively transcribed genes due to the interaction of integrase with host chromatin factor LEDGF. Although LEDGF was originally isolated as a co-activator that stimulates promoter activity in purified systems, this role is inconsistent with LEDGF-mediated integration across gene bodies and with data indicating LEDGF is a histone chaperone that promotes transcriptional elongation. We found LEDGF is enriched in pronounced peaks that match the enrichments of H3K4me3 and RNA Pol II at transcription start sites (TSSs) of active promoters. Our genome-wide chromatin mapping revealed that MLL1 had a dominant role in recruiting LEDGF to promoters and the presence of LEDGF recruits RNA Pol II. Enrichment of LEDGF at TSSs correlates strongly with levels of integration across the transcribed sequences, indicating that LEDGF at TSSs contributed to integration across gene bodies. Although the N-terminal Pro-Trp-Trp-Pro (PWWP) domain of LEDGF interacts with nucleosomes containing H3K36me3, a modification thought to recruit LEDGF to chromatin, we found H3K36me3 does not contribute to gene specificity of integration. These data support a dual role model of LEDGF where it is tethered to promoters by MLL1 and recruits RNA Pol II. Subsequently, LEDGF travels across genes to effect HIV-1 integration. Our data also provides a mechanistic context for the contribution made by LEDGF to MLL1-based infant acute leukemia and acute myeloid leukemia in adults.
Collapse
|
24
|
Larue AEM, Atlasi Y. The epigenetic landscape in intestinal stem cells and its deregulation in colorectal cancer. Stem Cells 2024; 42:509-525. [PMID: 38597726 PMCID: PMC11177158 DOI: 10.1093/stmcls/sxae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Epigenetic mechanisms play a pivotal role in controlling gene expression and cellular plasticity in both normal physiology and pathophysiological conditions. These mechanisms are particularly important in the regulation of stem cell self-renewal and differentiation, both in embryonic development and within adult tissues. A prime example of this finely tuned epigenetic control is observed in the gastrointestinal lining, where the small intestine undergoes renewal approximately every 3-5 days. How various epigenetic mechanisms modulate chromatin functions in intestinal stem cells (ISCs) is currently an active area of research. In this review, we discuss the main epigenetic mechanisms that control ISC differentiation under normal homeostasis. Furthermore, we explore the dysregulation of these mechanisms in the context of colorectal cancer (CRC) development. By outlining the main epigenetic mechanisms contributing to CRC, we highlight the recent therapeutics development and future directions for colorectal cancer research.
Collapse
Affiliation(s)
- Axelle E M Larue
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, United Kingdom
| | - Yaser Atlasi
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, United Kingdom
| |
Collapse
|
25
|
Bussey-Sutton CR, Ward A, Fox JA, Turner AMW, Peterson JJ, Emery A, Longoria AR, Gomez-Martinez I, Jones C, Hepperla A, Margolis DM, Strahl BD, Browne EP. The histone methyltransferase SETD2 regulates HIV expression and latency. PLoS Pathog 2024; 20:e1012281. [PMID: 38848441 PMCID: PMC11189200 DOI: 10.1371/journal.ppat.1012281] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/20/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Understanding the mechanisms that drive HIV expression and latency is a key goal for achieving an HIV cure. Here we investigate the role of the SETD2 histone methyltransferase, which deposits H3K36 trimethylation (H3K36me3), in HIV infection. We show that prevention of H3K36me3 by a potent and selective inhibitor of SETD2 (EPZ-719) leads to reduced post-integration viral gene expression and accelerated emergence of latently infected cells. CRISPR/Cas9-mediated knockout of SETD2 in primary CD4 T cells confirmed the role of SETD2 in HIV expression. Transcriptomic profiling of EPZ-719-exposed HIV-infected cells identified numerous pathways impacted by EPZ-719. Notably, depletion of H3K36me3 prior to infection did not prevent HIV integration but resulted in a shift of integration sites from highly transcribed genes to quiescent chromatin regions and to polycomb repressed regions. We also observed that SETD2 inhibition did not apparently affect HIV RNA levels, indicating a post-transcriptional mechanism affecting HIV expression. Viral RNA splicing was modestly reduced in the presence of EPZ-719. Intriguingly, EPZ-719 exposure enhanced responsiveness of latent HIV to the HDAC inhibitor vorinostat, suggesting that H3K36me3 can contribute to a repressive chromatin state at the HIV locus. These results identify SETD2 and H3K36me3 as novel regulators of HIV integration, expression and latency.
Collapse
Affiliation(s)
- Cameron R. Bussey-Sutton
- Department of Biochemistry, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Airlie Ward
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Joshua A. Fox
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Anne-Marie W. Turner
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jackson J. Peterson
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ann Emery
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Arturo R. Longoria
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ismael Gomez-Martinez
- Department of Genetics, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Corbin Jones
- Department of Genetics, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Austin Hepperla
- Department of Genetics, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David M. Margolis
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Brian D. Strahl
- Department of Biochemistry, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Edward P. Browne
- Department of Medicine, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC HIV Cure Center, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, UNC Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
26
|
Chen L, Liu J, Chen K, Su Y, Chen Y, Lei Y, Si J, Zhang J, Zhang Z, Zou W, Zhang X, Rondina MT, Wang QF, Li Y. SET domain containing 2 promotes megakaryocyte polyploidization and platelet generation through methylation of α-tubulin. J Thromb Haemost 2024; 22:1727-1741. [PMID: 38537781 DOI: 10.1016/j.jtha.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/23/2024] [Accepted: 03/12/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Megakaryocytes (MKs) are polyploid cells responsible for producing ∼1011 platelets daily in humans. Unraveling the mechanisms regulating megakaryopoiesis holds the promise for the production of clinical-grade platelets from stem cells, overcoming significant current limitations in platelet transfusion medicine. Previous work identified that loss of the epigenetic regulator SET domain containing 2 (SETD2) was associated with an increased platelet count in mice. However, the role of SETD2 in megakaryopoiesis remains unknown. OBJECTIVES Here, we examined how SETD2 regulated MK development and platelet production using complementary murine and human systems. METHODS We manipulated the expression of SETD2 in multiple in vitro and ex vivo models to assess the ploidy of MKs and the function of platelets. RESULTS The genetic ablation of Setd2 increased the number of high-ploidy bone marrow MKs. Peripheral platelet counts in Setd2 knockout mice were significantly increased ∼2-fold, and platelets exhibited normal size, morphology, and function. By knocking down and overexpressing SETD2 in ex vivo human cell systems, we demonstrated that SETD2 negatively regulated MK polyploidization by controlling methylation of α-tubulin, microtubule polymerization, and MK nuclear division. Small-molecule inactivation of SETD2 significantly increased the production of high-ploidy MKs and platelets from human-induced pluripotent stem cells and cord blood CD34+ cells. CONCLUSION These findings identify a previously unrecognized role for SETD2 in regulating megakaryopoiesis and highlight the potential of targeting SETD2 to increase platelet production from human cells for transfusion practices.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jingkun Liu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Kunying Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yanxun Su
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yihe Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ying Lei
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jia Si
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jie Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhaojun Zhang
- University of Chinese Academy of Sciences, Beijing, China; Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center of Bioinformation, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Weiguo Zou
- Shanghai Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Matthew T Rondina
- Departments of Internal Medicine and Pathology, Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA; Department of Internal Medicine and the Geriatric Research, Education, and Clinical Center, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah, USA.
| | - Qian-Fei Wang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Yueying Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
27
|
Hong Y, Bie L, Zhang T, Yan X, Jin G, Chen Z, Wang Y, Li X, Pei G, Zhang Y, Hong Y, Gong L, Li P, Xie W, Zhu Y, Shen X, Liu N. SAFB restricts contact domain boundaries associated with L1 chimeric transcription. Mol Cell 2024; 84:1637-1650.e10. [PMID: 38604171 DOI: 10.1016/j.molcel.2024.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/05/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Long interspersed element-1 (LINE-1 or L1) comprises 17% of the human genome, continuously generates genetic variations, and causes disease in certain cases. However, the regulation and function of L1 remain poorly understood. Here, we uncover that L1 can enrich RNA polymerase IIs (RNA Pol IIs), express L1 chimeric transcripts, and create contact domain boundaries in human cells. This impact of L1 is restricted by a nuclear matrix protein scaffold attachment factor B (SAFB) that recognizes transcriptionally active L1s by binding L1 transcripts to inhibit RNA Pol II enrichment. Acute inhibition of RNA Pol II transcription abolishes the domain boundaries associated with L1 chimeric transcripts, indicating a transcription-dependent mechanism. Deleting L1 impairs domain boundary formation, and L1 insertions during evolution have introduced species-specific domain boundaries. Our data show that L1 can create RNA Pol II-enriched regions that alter genome organization and that SAFB regulates L1 and RNA Pol II activity to preserve gene regulation.
Collapse
Affiliation(s)
- Yaqiang Hong
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Luyao Bie
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tao Zhang
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaohan Yan
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Guangpu Jin
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhuo Chen
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yang Wang
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiufeng Li
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Gaofeng Pei
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yongyan Zhang
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yantao Hong
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Liang Gong
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
| | - Pilong Li
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wei Xie
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanfen Zhu
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xiaohua Shen
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Nian Liu
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
28
|
Brockett JS, Manalo T, Zein-Sabatto H, Lee J, Fang J, Chu P, Feng H, Patil D, Davidson P, Ogan K, Master VA, Pattaras JG, Roberts DL, Bergquist SH, Reyna MA, Petros JA, Lerit DA, Arnold RS. A missense SNP in the tumor suppressor SETD2 reduces H3K36me3 and mitotic spindle integrity in Drosophila. Genetics 2024; 226:iyae015. [PMID: 38290049 PMCID: PMC10990431 DOI: 10.1093/genetics/iyae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
Mutations in SETD2 are among the most prevalent drivers of renal cell carcinoma (RCC). We identified a novel single nucleotide polymorphism (SNP) in SETD2, E902Q, within a subset of RCC patients, which manifests as both an inherited or tumor-associated somatic mutation. To determine if the SNP is biologically functional, we used CRISPR-based genome editing to generate the orthologous mutation within the Drosophila melanogaster Set2 gene. In Drosophila, the homologous amino acid substitution, E741Q, reduces H3K36me3 levels comparable to Set2 knockdown, and this loss is rescued by reintroduction of a wild-type Set2 transgene. We similarly uncovered significant defects in spindle morphogenesis, consistent with the established role of SETD2 in methylating α-Tubulin during mitosis to regulate microtubule dynamics and maintain genome stability. These data indicate the Set2 E741Q SNP affects both histone methylation and spindle integrity. Moreover, this work further suggests the SETD2 E902Q SNP may hold clinical relevance.
Collapse
Affiliation(s)
- Jovan S Brockett
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tad Manalo
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hala Zein-Sabatto
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jina Lee
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Junnan Fang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Philip Chu
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Harry Feng
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dattatraya Patil
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Priscilla Davidson
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kenneth Ogan
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Viraj A Master
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John G Pattaras
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - David L Roberts
- Emory University Department of Medicine, Division of General Internal Medicine, Atlanta, GA 30322, USA
| | - Sharon H Bergquist
- Emory University Department of Medicine, Division of General Internal Medicine, Atlanta, GA 30322, USA
| | - Matthew A Reyna
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John A Petros
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Dorothy A Lerit
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Rebecca S Arnold
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
29
|
Jia L, Cowell LG, Kapur P. Understanding Factors that Influence Prognosis and Response to Therapy in Clear Cell Renal Cell Carcinoma. Adv Anat Pathol 2024; 31:96-104. [PMID: 38179997 DOI: 10.1097/pap.0000000000000428] [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: 01/06/2024]
Abstract
In this review, we highlight and contextualize emerging morphologic prognostic and predictive factors in renal cell carcinoma. We focus on clear cell renal cell carcinoma (ccRCC), the most common histologic subtype. Our understanding of the molecular characterization of ccRCC has dramatically improved in the last decade. Herein, we highlight how these discoveries have laid the foundation for new approaches to prognosis and therapeutic decision-making for patients with ccRCC. We explore the clinical relevance of common mutations, established gene expression signatures, intratumoral heterogeneity, sarcomatoid/rhabdoid morphology and PD-L1 expression, and discuss their impact on predicting response to therapy.
Collapse
Affiliation(s)
| | - Lindsay G Cowell
- Peter O'Donnell School of Public Health
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, Dallas, TX
| | - Payal Kapur
- Department of Pathology
- Department of Urology, University of Texas Southwestern Medical Center
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, Dallas, TX
| |
Collapse
|
30
|
Febres-Aldana CA, Fanaroff R, Offin M, Zauderer MG, Sauter JL, Yang SR, Ladanyi M. Diffuse Pleural Mesothelioma: Advances in Molecular Pathogenesis, Diagnosis, and Treatment. ANNUAL REVIEW OF PATHOLOGY 2024; 19:11-42. [PMID: 37722697 DOI: 10.1146/annurev-pathol-042420-092719] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
Diffuse pleural mesothelioma (DPM) is a highly aggressive malignant neoplasm arising from the mesothelial cells lining the pleural surfaces. While DPM is a well-recognized disease linked to asbestos exposure, recent advances have expanded our understanding of molecular pathogenesis and transformed our clinical practice. This comprehensive review explores the current concepts and emerging trends in DPM, including risk factors, pathobiology, histologic subtyping, and therapeutic management, with an emphasis on a multidisciplinary approach to this complex disease.
Collapse
Affiliation(s)
- Christopher A Febres-Aldana
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| | - Rachel Fanaroff
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| | - Michael Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Marjorie G Zauderer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jennifer L Sauter
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| | - Soo-Ryum Yang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| |
Collapse
|
31
|
Shao R, Suo J, Zhang Z, Kong M, Ma Y, Wen Y, Liu M, Zhuang L, Ge K, Bi Q, Zhang C, Zou W. H3K36 methyltransferase NSD1 protects against osteoarthritis through regulating chondrocyte differentiation and cartilage homeostasis. Cell Death Differ 2024; 31:106-118. [PMID: 38012390 PMCID: PMC10781997 DOI: 10.1038/s41418-023-01244-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common joint diseases, there are no effective disease-modifying drugs, and the pathological mechanisms of OA need further investigation. Here, we show that H3K36 methylations were decreased in senescent chondrocytes and age-related osteoarthritic cartilage. Prrx1-Cre inducible H3.3K36M transgenic mice showed articular cartilage destruction and osteophyte formation. Conditional knockout Nsd1Prrx1-Cre mice, but not Nsd2Prrx1-Cre or Setd2Prrx1-Cre mice, replicated the phenotype of K36M/+; Prrx1-Cre mice. Immunostaining results showed decreased anabolic and increased catabolic activities in Nsd1Prrx1-Cre mice, along with decreased chondrogenic differentiation. Transcriptome and ChIP-seq data revealed that Osr2 was a key factor affected by Nsd1. Intra-articular delivery of Osr2 adenovirus effectively improved the homeostasis of articular cartilage in Nsd1Prrx1-Cre mice. In human osteoarthritic cartilages, both mRNA and protein levels of NSD1 and OSR2 were decreased. Our results indicate that NSD1-induced H3K36 methylations and OSR2 expression play important roles in articular cartilage homeostasis and OA. Targeting H3K36 methylation and OSR2 would be a novel strategy for OA treatment.
Collapse
Affiliation(s)
- Rui Shao
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhong Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mingxiang Kong
- Department of Orthopedics, Rehabilitation center, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China
| | - Yiyang Ma
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yang Wen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengxue Liu
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Lenan Zhuang
- Adipocyte Biology and Gene Regulation Section, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, 20892, USA
| | - Kai Ge
- Adipocyte Biology and Gene Regulation Section, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, 20892, USA
| | - Qing Bi
- Department of Orthopedics, Rehabilitation center, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China
| | - Changqing Zhang
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Weiguo Zou
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
32
|
Lewerissa EI, Nadif Kasri N, Linda K. Epigenetic regulation of autophagy-related genes: Implications for neurodevelopmental disorders. Autophagy 2024; 20:15-28. [PMID: 37674294 PMCID: PMC10761153 DOI: 10.1080/15548627.2023.2250217] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionarily highly conserved catabolic process that is important for the clearance of cytosolic contents to maintain cellular homeostasis and survival. Recent findings point toward a critical role for autophagy in brain function, not only by preserving neuronal health, but especially by controlling different aspects of neuronal development and functioning. In line with this, mutations in autophagy-related genes are linked to various key characteristics and symptoms of neurodevelopmental disorders (NDDs), including autism, micro-/macrocephaly, and epilepsy. However, the group of NDDs caused by mutations in autophagy-related genes is relatively small. A significant proportion of NDDs are associated with mutations in genes encoding epigenetic regulatory proteins that modulate gene expression, so-called chromatinopathies. Intriguingly, several of the NDD-linked chromatinopathy genes have been shown to regulate autophagy-related genes, albeit in non-neuronal contexts. From these studies it becomes evident that tight transcriptional regulation of autophagy-related genes is crucial to control autophagic activity. This opens the exciting possibility that aberrant autophagic regulation might underly nervous system impairments in NDDs with disturbed epigenetic regulation. We here summarize NDD-related chromatinopathy genes that are known to regulate transcriptional regulation of autophagy-related genes. Thereby, we want to highlight autophagy as a candidate key hub mechanism in NDD-related chromatinopathies.Abbreviations: ADNP: activity dependent neuroprotector homeobox; ASD: autism spectrum disorder; ATG: AutTophaGy related; CpG: cytosine-guanine dinucleotide; DNMT: DNA methyltransferase; EHMT: euchromatic histone lysine methyltransferase; EP300: E1A binding protein p300; EZH2: enhancer of zeste 2 polycomb repressive complex 2 subunit; H3K4me3: histone 3 lysine 4 trimethylation; H3K9me1/2/3: histone 3 lysine 9 mono-, di-, or trimethylation; H3K27me2/3: histone 3 lysine 27 di-, or trimethylation; hiPSCs: human induced pluripotent stem cells; HSP: hereditary spastic paraplegia; ID: intellectual disability; KANSL1: KAT8 regulatory NSL complex subunit 1; KAT8: lysine acetyltransferase 8; KDM1A/LSD1: lysine demethylase 1A; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin complex 1; NDD: neurodevelopmental disorder; PHF8: PHD finger protein 8; PHF8-XLID: PHF8-X linked intellectual disability syndrome; PTM: post-translational modification; SESN2: sestrin 2; YY1: YY1 transcription factor; YY1AP1: YY1 associated protein 1.
Collapse
Affiliation(s)
- Elly I. Lewerissa
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Gelderland, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Gelderland, The Netherlands
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behavior, Nijmegen, Gelderland, The Netherlands
| | - Katrin Linda
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Gelderland, The Netherlands
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Flemish Brabant, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Flemish Brabant, Belgium
| |
Collapse
|
33
|
Contreras Yametti GP, Robbins G, Chowdhury A, Narang S, Ostrow TH, Kilberg H, Greenberg J, Kramer L, Raetz E, Tsirigos A, Evensen NA, Carroll WL. SETD2 mutations do not contribute to clonal fitness in response to chemotherapy in childhood B cell acute lymphoblastic leukemia. Leuk Lymphoma 2024; 65:78-90. [PMID: 37874744 PMCID: PMC11874253 DOI: 10.1080/10428194.2023.2273752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 10/26/2023]
Abstract
Mutations in genes encoding epigenetic regulators are commonly observed at relapse in B cell acute lymphoblastic leukemia (B-ALL). Loss-of-function mutations in SETD2, an H3K36 methyltransferase, have been observed in B-ALL and other cancers. Previous studies on mutated SETD2 in solid tumors and acute myelogenous leukemia support a role in promoting resistance to DNA damaging agents. We did not observe chemoresistance, an impaired DNA damage response, nor increased mutation frequency in response to thiopurines using CRISPR-mediated knockout in wild-type B-ALL cell lines. Likewise, restoration of SETD2 in cell lines with hemizygous mutations did not increase sensitivity. SETD2 mutations affected the chromatin landscape and transcriptional output that was unique to each cell line. Collectively our data does not support a role for SETD2 mutations in driving clonal evolution and relapse in B-ALL, which is consistent with the lack of enrichment of SETD2 mutations at relapse in most studies.
Collapse
Affiliation(s)
- Gloria P. Contreras Yametti
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Gabriel Robbins
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Ashfiyah Chowdhury
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Sonali Narang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Talia H. Ostrow
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Harrison Kilberg
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Joshua Greenberg
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Lindsay Kramer
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Elizabeth Raetz
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Aristotelis Tsirigos
- Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Nikki A. Evensen
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - William L. Carroll
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Perlmutter Cancer Center, NYU Langone Health, New York, NY
- Department of Pathology, NYU Langone Health, New York, NY
| |
Collapse
|
34
|
Zheng X, Luo Y, Xiong Y, Liu X, Zeng C, Lu X, Wang X, Cheng Y, Wang S, Lan H, Wang K, Weng Z, Bi W, Gan X, Jia X, Wang L, Wang Y. Tumor cell-intrinsic SETD2 inactivation sensitizes cancer cells to immune checkpoint blockade through the NR2F1-STAT1 pathway. J Immunother Cancer 2023; 11:e007678. [PMID: 38056895 PMCID: PMC10711831 DOI: 10.1136/jitc-2023-007678] [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: 11/16/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Cancer immunotherapies can induce durable tumor regression, but most patients do not respond. SETD2 mutation has been linked to the efficacy of immune checkpoint inhibitors (ICIs) immunotherapy. The functional importance of the SETD2 inactivation and how to modulate immunotherapy response remains unclear. METHODS To explore the function of SETD2 in immunotherapy, knockout and subsequent functional experiments were conducted. Bulk RNA-seq, ATAC-seq, Chip-seq and single-cell RNA-seq were performed to dissect the mechanism and explore the immune microenvironment of mouse tumor. Flow cytometry was used to assess cell surface antigen and intratumoral T cell levels. RESULTS We comprehensively determine the effect of SETD2 inactivation in ICIs therapy and elucidate the mechanistic impact on tumor immunity. Murine syngeneic tumors harboring Setd2 inactivation are sensitive to ICIs. By bulk and single-cell RNA-seq, we further reveal that SETD2 inactivation reprograms intratumoral immune cells and inflames the tumor microenvironment, which is characterized by high infiltration of T cells and enhanced antigen presentation to activate CD8+ T cell-mediated killing. Mechanistically, via an integrated multiomics analysis using ATAC-seq, ChIP-seq and RNA-seq, we demonstrate that SETD2 inactivation reduces NR2F1 transcription by impairing H3K36me3 deposition and chromatin accessibility, which activates the STAT1 signaling pathway to promote chemokines and programmed cell death protein-1 (PD-1) expression and enhance antigen presentation. All these regulatory mechanisms synergistically promote the effects of anti-programmed cell death ligand 1 immunotherapy in Setd2-knockout syngeneic mouse models. The SETD2-NR2F1-STAT1 regulatory axis is conserved in human and murine cancers. Finally, cancer patients harboring SETD2 mutations who received ICIs show increased durable clinical benefits and survival. CONCLUSIONS These findings provide novel insights into the biology of SETD2 inactivation regulation and reveal a new potential therapeutic biomarker for ICIs immunotherapy in various refractory cancers.
Collapse
Affiliation(s)
- Xufen Zheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuxiang Luo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yangjie Xiong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoxiao Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chunling Zeng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaojing Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaofang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yumei Cheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Simin Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haoqi Lan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kai Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhonghui Weng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenbo Bi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xinxin Gan
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaona Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yuexiang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
35
|
Su Z, Liu Z, Lei W, Xia K, Xiao A, Hu Z, Zhou M, Zhu F, Tian J, Yang M, Wang D, Xiang AP, Nie J. Hyperhomocysteinemia lowers serum testosterone concentration via impairing testosterone production in Leydig cells. Cell Biol Toxicol 2023; 39:3077-3100. [PMID: 37495868 DOI: 10.1007/s10565-023-09819-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Hyperhomocysteinemia (HHcy) plays a salient role in male infertility. However, whether HHcy interferes with testosterone production remains inconclusive. Here, we reported a lower serum testosterone level in HHcy mice. Single-cell RNA sequencing revealed that genes related to testosterone biosynthesis, together with nuclear receptor subfamily 5 group A member 1 (Nr5a1), a key transcription factor for steroidogenic genes, were downregulated in the Leydig cells (LCs) of HHcy mice. Mechanistically, Hcy lowered trimethylation of histone H3 on lysine 4 (H3K4me3), which was bound on the promoter region of Nr5a1, resulting in downregulation of Nr5a1. Intriguingly, we identified an unknown cell cluster annotated as Macrophage-like Leydig cells (McLCs), expressing both LCs and macrophages markers. In HHcy mice, McLCs were shifted toward pro-inflammatory phenotype and thus promoted inflammatory response in LC. Betaine supplementation rescued the downregulation of NR5A1 and restored the serum testosterone level in HHcy mice. Overall, our study highlights an etiological role of HHcy in LCs dysfunction.
Collapse
Affiliation(s)
- Zhiyuan Su
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhuoliang Liu
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Wenjing Lei
- Department of Nephrology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Kai Xia
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - An Xiao
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zheng Hu
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Miaomiao Zhou
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Fengxin Zhu
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jianwei Tian
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Manqiu Yang
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Dong Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Jing Nie
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
36
|
Wang C, Wang T, Li KJ, Hu LH, Li Y, Yu YZ, Xie T, Zhu S, Fu DJ, Wang Y, Zeng XZ, Liu FP, Chen H, Chen ZS, Feng NH, Liu J, Jiang Y, Zhao SC. SETD4 inhibits prostate cancer development by promoting H3K27me3-mediated NUPR1 transcriptional repression and cell cycle arrest. Cancer Lett 2023; 579:216464. [PMID: 37879429 DOI: 10.1016/j.canlet.2023.216464] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/05/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023]
Abstract
The suppressor of variegation enhancer of zeste-trithorax (SET) domain methyltransferases have been reported to function as key regulators in multiple tumor types by catalyzing histone lysine methylation. Nevertheless, our understanding on the role of these lysine methyltransferases, including SETD4, in prostate cancer (PCa) remains limited. Hence, the specific role of SETD4 in PCa was investigated in this study. The expression of SETD4 in PCa cells and tissue samples was downregulated in PCa cells and tissue specimens, and decreased SETD4 expression led to inferior clinicopathological characteristics in patients with PCa. knockdown of SETD4 facilitated the proliferation of PCa cells and accelerated cell cycle progression. Mechanistically, SETD4 repressed NUPR1 transcription by methylating H3K27 to generate H3K27me3, subsequently inactivated Akt pathway and impeded the tumorigenesis of PCa. Our results highlight that SETD4 prevents the development of PCa by catalyzing the methylation of H3K27 and suppressing NUPR1 transcription, subsequently inactivating the Akt signaling pathway. The findings suggest the potential application of SETD4 in PCa prognosis and therapeutics.
Collapse
Affiliation(s)
- Chong Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Tao Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510900, China
| | - Kang-Jing Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ling-Hong Hu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yue Li
- Laboratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yu-Zhong Yu
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510500, China
| | - Tao Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Sha Zhu
- Department of Urology, Jiangnan University Medical Center, Wuxi, 214002, China; Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, China
| | - Du-Jiang Fu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yang Wang
- Department of Urology, Jiangnan University Medical Center, Wuxi, 214002, China; Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, China
| | - Xian-Zi Zeng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Feng-Ping Liu
- Department of Urology, Jiangnan University Medical Center, Wuxi, 214002, China; Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, China
| | - Hong Chen
- Luoyang Key Laboratory of Organic Functional Molecules, College of Food and Drug, Luoyang Normal University, Luoyang, Henan, 471934, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Ning-Han Feng
- Department of Urology, Jiangnan University Medical Center, Wuxi, 214002, China; Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, China.
| | - Jinghua Liu
- Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Shan-Chao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510500, China.
| |
Collapse
|
37
|
Sun Y, Ramesh V, Wei F, Locasale JW. Methionine availability influences essential H3K36me3 dynamics during cell differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568331. [PMID: 38045360 PMCID: PMC10690240 DOI: 10.1101/2023.11.22.568331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Histone modifications are integral to epigenetics through their influence on gene expression and cellular status. While it's established that metabolism, including methionine metabolism, can impact histone methylation, the direct influence of methionine availability on crucial histone marks that determine the epigenomic process remains poorly understood. In this study, we demonstrate that methionine, through its metabolic product, S-adenosylmethionine (SAM), dynamically regulates H3K36me3, a cancer-associated histone modification known to influence cellular status, and myogenic differentiation of mouse myoblast cells. We further demonstrate that the methionine-dependent effects on differentiation are mediated in part through the histone methyltransferase SETD2. Methionine restriction leads to preferential decreases in H3K36me3 abundance and genome accessibility of genes involved in myogenic differentiation. Importantly, the effects of methionine restriction on differentiation and chromatin accessibility can be phenocopied by the deletion of Setd2. Collectively, this study demonstrates that methionine metabolism through its ability to be sensed by chromatin modifying enzymes can have a direct role in influencing cell fate determination.
Collapse
|
38
|
Depierre D, Perrois C, Schickele N, Lhoumaud P, Abdi-Galab M, Fosseprez O, Heurteau A, Margueron R, Cuvier O. Chromatin in 3D distinguishes dMes-4/NSD and Hypb/dSet2 in protecting genes from H3K27me3 silencing. Life Sci Alliance 2023; 6:e202302038. [PMID: 37684044 PMCID: PMC10491495 DOI: 10.26508/lsa.202302038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Cell type-specific barcoding of genomes requires the establishment of hundreds of heterochromatin domains where heterochromatin-associated repressive complexes hinder chromatin accessibility thereby silencing genes. At heterochromatin-euchromatin borders, regulation of accessibility not only depends on the delimitation of heterochromatin but may also involve interplays with nearby genes and their transcriptional activity, or alternatively on histone modifiers, chromatin barrier insulators, and more global demarcation of chromosomes into 3D compartmentalized domains and topological-associating domain (TADs). Here, we show that depletion of H3K36 di- or tri-methyl histone methyltransferases dMes-4/NSD or Hypb/dSet2 induces reproducible increasing levels of H3K27me3 at heterochromatin borders including in nearby promoters, thereby repressing hundreds of genes. Furthermore, dMes-4/NSD influences genes demarcated by insulators and TAD borders, within chromatin hubs, unlike transcription-coupled action of Hypb/dSet2 that protects genes independently of TADs. Insulator mutants recapitulate the increase of H3K27me3 upon dMes-4/NSD depletion unlike Hypb/dSet2. Hi-C data demonstrate how dMes-4/NSD blocks propagation of long-range interactions onto active regions. Our data highlight distinct mechanisms protecting genes from H3K27me3 silencing, highlighting a direct influence of H3K36me on repressive TADs.
Collapse
Affiliation(s)
- David Depierre
- Chromatin Dynamics and Cell Proliferation, Center of Integrative Biology, Molecular, Cellular and Developmental Biology (MCD/UMR5087), CNRS, Université Paul Sabatier de Toulouse, Toulouse, France
| | - Charlène Perrois
- Chromatin Dynamics and Cell Proliferation, Center of Integrative Biology, Molecular, Cellular and Developmental Biology (MCD/UMR5087), CNRS, Université Paul Sabatier de Toulouse, Toulouse, France
| | - Naomi Schickele
- Chromatin Dynamics and Cell Proliferation, Center of Integrative Biology, Molecular, Cellular and Developmental Biology (MCD/UMR5087), CNRS, Université Paul Sabatier de Toulouse, Toulouse, France
| | - Priscillia Lhoumaud
- Chromatin Dynamics and Cell Proliferation, Center of Integrative Biology, Molecular, Cellular and Developmental Biology (MCD/UMR5087), CNRS, Université Paul Sabatier de Toulouse, Toulouse, France
| | - Mahdia Abdi-Galab
- Chromatin Dynamics and Cell Proliferation, Center of Integrative Biology, Molecular, Cellular and Developmental Biology (MCD/UMR5087), CNRS, Université Paul Sabatier de Toulouse, Toulouse, France
| | - Olivier Fosseprez
- Chromatin Dynamics and Cell Proliferation, Center of Integrative Biology, Molecular, Cellular and Developmental Biology (MCD/UMR5087), CNRS, Université Paul Sabatier de Toulouse, Toulouse, France
| | - Alexandre Heurteau
- Chromatin Dynamics and Cell Proliferation, Center of Integrative Biology, Molecular, Cellular and Developmental Biology (MCD/UMR5087), CNRS, Université Paul Sabatier de Toulouse, Toulouse, France
| | - Raphaël Margueron
- Institut Curie, Paris Sciences et Lettres Research University; INSERM U934/ CNRS UMR3215, Paris, France
| | - Olivier Cuvier
- Chromatin Dynamics and Cell Proliferation, Center of Integrative Biology, Molecular, Cellular and Developmental Biology (MCD/UMR5087), CNRS, Université Paul Sabatier de Toulouse, Toulouse, France
| |
Collapse
|
39
|
Mitchell B, Thor S, Piper M. Cellular and molecular functions of SETD2 in the central nervous system. J Cell Sci 2023; 136:jcs261406. [PMID: 37921122 DOI: 10.1242/jcs.261406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023] Open
Abstract
The covalent modification of histones is critical for many biological functions in mammals, including gene regulation and chromatin structure. Posttranslational histone modifications are added and removed by specialised 'writer' and 'eraser' enzymes, respectively. One such writer protein implicated in a wide range of cellular processes is SET domain-containing 2 (SETD2), a histone methyltransferase that catalyses the trimethylation of lysine 36 on histone H3 (H3K36me3). Recently, SETD2 has also been found to modify proteins other than histones, including actin and tubulin. The emerging roles of SETD2 in the development and function of the mammalian central nervous system (CNS) are of particular interest as several SETD2 variants have been implicated in neurodevelopmental disorders, such as autism spectrum disorder and the overgrowth disorder Luscan-Lumish syndrome. Here, we summarise the numerous roles of SETD2 in mammalian cellular functions and development, with a focus on the CNS. We also provide an overview of the consequences of SETD2 variants in human disease and discuss future directions for understanding essential cellular functions of SETD2.
Collapse
Affiliation(s)
- Benjamin Mitchell
- The School of Biomedical Sciences, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Stefan Thor
- The School of Biomedical Sciences, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Michael Piper
- The School of Biomedical Sciences, University of Queensland, Brisbane, Queensland 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
40
|
Kovatcheva M, Melendez E, Chondronasiou D, Pietrocola F, Bernad R, Caballe A, Junza A, Capellades J, Holguín-Horcajo A, Prats N, Durand S, Rovira M, Yanes O, Stephan-Otto Attolini C, Kroemer G, Serrano M. Vitamin B 12 is a limiting factor for induced cellular plasticity and tissue repair. Nat Metab 2023; 5:1911-1930. [PMID: 37973897 PMCID: PMC10663163 DOI: 10.1038/s42255-023-00916-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 09/27/2023] [Indexed: 11/19/2023]
Abstract
Transient reprogramming by the expression of OCT4, SOX2, KLF4 and MYC (OSKM) is a therapeutic strategy for tissue regeneration and rejuvenation, but little is known about its metabolic requirements. Here we show that OSKM reprogramming in mice causes a global depletion of vitamin B12 and molecular hallmarks of methionine starvation. Supplementation with vitamin B12 increases the efficiency of reprogramming both in mice and in cultured cells, the latter indicating a cell-intrinsic effect. We show that the epigenetic mark H3K36me3, which prevents illegitimate initiation of transcription outside promoters (cryptic transcription), is sensitive to vitamin B12 levels, providing evidence for a link between B12 levels, H3K36 methylation, transcriptional fidelity and efficient reprogramming. Vitamin B12 supplementation also accelerates tissue repair in a model of ulcerative colitis. We conclude that vitamin B12, through its key role in one-carbon metabolism and epigenetic dynamics, improves the efficiency of in vivo reprogramming and tissue repair.
Collapse
Affiliation(s)
- Marta Kovatcheva
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Elena Melendez
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Dafni Chondronasiou
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Federico Pietrocola
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Raquel Bernad
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Adrià Caballe
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Alexandra Junza
- Universitat Rovira i Virgili, Department of Electronic Engineering, IISPV, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Capellades
- Universitat Rovira i Virgili, Department of Electronic Engineering, IISPV, Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Metabolomics Platform, Reus, Spain
| | - Adrián Holguín-Horcajo
- Department of Physiological Science, School of Medicine, Universitat de Barcelona (UB), L'Hospitalet de Llobregat, Spain
- Pancreas Regeneration: Pancreatic Progenitors and Their Niche Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Neus Prats
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Sylvere Durand
- Metabolomics and Cell Biology Platforms UMS AMMICa/UMR 1138, Institut Gustave Roussy, Villejuif, France
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Institut Universitaire de France, Paris, France
| | - Meritxell Rovira
- Department of Physiological Science, School of Medicine, Universitat de Barcelona (UB), L'Hospitalet de Llobregat, Spain
- Pancreas Regeneration: Pancreatic Progenitors and Their Niche Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Oscar Yanes
- Universitat Rovira i Virgili, Department of Electronic Engineering, IISPV, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms UMS AMMICa/UMR 1138, Institut Gustave Roussy, Villejuif, France
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Institut Universitaire de France, Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
- Altos Labs, Cambridge Institute of Science, Cambridge, UK.
| |
Collapse
|
41
|
Serdyukova K, Swearingen AR, Coradin M, Nevo M, Tran H, Bajric E, Brumbaugh J. Leveraging dominant-negative histone H3 K-to-M mutations to study chromatin during differentiation and development. Development 2023; 150:dev202169. [PMID: 37846748 PMCID: PMC10617616 DOI: 10.1242/dev.202169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Histone modifications are associated with regulation of gene expression that controls a vast array of biological processes. Often, these associations are drawn by correlating the genomic location of a particular histone modification with gene expression or phenotype; however, establishing a causal relationship between histone marks and biological processes remains challenging. Consequently, there is a strong need for experimental approaches to directly manipulate histone modifications. A class of mutations on the N-terminal tail of histone H3, lysine-to-methionine (K-to-M) mutations, was identified as dominant-negative inhibitors of histone methylation at their respective and specific residues. The dominant-negative nature of K-to-M mutants makes them a valuable tool for studying the function of specific methylation marks on histone H3. Here, we review recent applications of K-to-M mutations to understand the role of histone methylation during development and homeostasis. We highlight important advantages and limitations that require consideration when using K-to-M mutants, particularly in a developmental context.
Collapse
Affiliation(s)
- Ksenia Serdyukova
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Alison R. Swearingen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mariel Coradin
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mika Nevo
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Huong Tran
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Emir Bajric
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Justin Brumbaugh
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
42
|
Liu C, Ni L, Li X, Rao H, Feng W, Zhu Y, Zhang W, Ma C, Xu Y, Gui L, Wang Z, Aji R, Xu J, Gao W, Li L. SETD2 deficiency promotes renal fibrosis through the TGF-β/Smad signalling pathway in the absence of VHL. Clin Transl Med 2023; 13:e1468. [PMID: 37933774 PMCID: PMC10629155 DOI: 10.1002/ctm2.1468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Renal fibrosis is the final development pathway and the most common pathological manifestation of chronic kidney disease. Epigenetic alteration is a significant intrinsic factor contributing to the development of renal fibrosis. SET domain-containing 2 (SETD2) is the sole histone H3K36 trimethyltransferase, catalysing H3K36 trimethylation. There is evidence that SETD2-mediated epigenetic alterations are implicated in many diseases. However, it is unclear what role SETD2 plays in the development of renal fibrosis. METHODS Kidney tissues from mice as well as HK2 cells were used as research subjects. Clinical databases of patients with renal fibrosis were analysed to investigate whether SETD2 expression is reduced in the occurrence of renal fibrosis. SETD2 and Von Hippel-Lindau (VHL) double-knockout mice were used to further investigate the role of SETD2 in renal fibrosis. Renal tubular epithelial cells isolated from mice were used for RNA sequencing and chromatin immunoprecipitation sequencing to search for molecular signalling pathways and key molecules leading to renal fibrosis in mice. Molecular and cell biology experiments were conducted to analyse and validate the role of SETD2 in the development of renal fibrosis. Finally, rescue experiments were performed to determine the molecular mechanism of SETD2 deficiency in the development of renal fibrosis. RESULTS SETD2 deficiency leads to severe renal fibrosis in VHL-deficient mice. Mechanically, SETD2 maintains the transcriptional level of Smad7, a negative feedback factor of the transforming growth factor-β (TGF-β)/Smad signalling pathway, thereby preventing the activation of the TGF-β/Smad signalling pathway. Deletion of SETD2 leads to reduced Smad7 expression, which results in activation of the TGF-β/Smad signalling pathway and ultimately renal fibrosis in the absence of VHL. CONCLUSIONS Our findings reveal the role of SETD2-mediated H3K36me3 of Smad7 in regulating the TGF-β/Smad signalling pathway in renal fibrogenesis and provide an innovative insight into SETD2 as a potential therapeutic target for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Changwei Liu
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Li Ni
- Department of NursingShanghai East HospitalTongji UniversityShanghaiChina
| | - Xiaoxue Li
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Hanyu Rao
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Wenxin Feng
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Yiwen Zhu
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Wei Zhang
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Chunxiao Ma
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Yue Xu
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Liming Gui
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Ziyi Wang
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Rebiguli Aji
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Jin Xu
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Wei‐Qiang Gao
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Li Li
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
43
|
Shipman GA, Padilla R, Horth C, Hu B, Bareke E, Vitorino FN, Gongora JM, Garcia BA, Lu C, Majewski J. Systematic perturbations of SETD2, NSD1, NSD2, NSD3 and ASH1L reveals their distinct contributions to H3K36 methylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559313. [PMID: 37905045 PMCID: PMC10614729 DOI: 10.1101/2023.09.27.559313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Background Methylation of histone 3 lysine 36 (H3K36me) has emerged as an essential epigenetic component for the faithful regulation of gene expression. Despite its importance in development, disease, and cancer, how the molecular agents collectively shape the H3K36me landscape is unclear. Results We use a mouse mesenchymal stem cell model to perturb the H3K36me deposition machinery and infer the activities of the five most prominent players: SETD2, NSD1, NSD2, NSD3, and ASH1L. We find that H3K36me2 is the most abundant of the three methylation states and is predominantly deposited at intergenic regions by NSD1, and partly by NSD2. In contrast, H3K36me1/3 are most abundant within exons and are positively correlated with gene expression. We demonstrate that while SETD2 deposits most H3K36me3, it also deposits H3K36me2 within transcribed genes. Additionally, loss of SETD2 results in an increase of exonic H3K36me1, suggesting other H3K36 methyltransferases (K36MTs) prime gene bodies with lower methylation states ahead of transcription. Through a reductive approach, we uncover the distribution patterns of NSD3- and ASH1L-catalyzed H3K36me2. While NSD1/2 establish broad intergenic H3K36me2 domains, NSD3 deposits H3K36me2 peaks on active promoters and enhancers. Meanwhile, the activity of ASH1L is restricted to the regulatory elements of developmentally relevant genes, and our analyses implicate PBX2 as a potential recruitment factor. Conclusions Within genes, SETD2 deposits both H3K36me2/3, while the other K36MTs are capable of depositing H3K36me1/2 independently of SETD2 activity. For the deposition of H3K36me1/2, we find a hierarchy of K36MT activities where NSD1>NSD2>NSD3>ASH1L. While NSD1 and NSD2 are responsible for most genome-wide propagation of H3K36me2, the activities of NSD3 and ASH1L are confined to active regulatory elements.
Collapse
|
44
|
Cameron DP, Grosser J, Ladigan S, Kuzin V, Iliopoulou E, Wiegard A, Benredjem H, Jackson K, Liffers ST, Lueong S, Cheung PF, Vangala D, Pohl M, Viebahn R, Teschendorf C, Wolters H, Usta S, Geng K, Kutter C, Arsenian-Henriksson M, Siveke JT, Tannapfel A, Schmiegel W, Hahn SA, Baranello L. Coinhibition of topoisomerase 1 and BRD4-mediated pause release selectively kills pancreatic cancer via readthrough transcription. SCIENCE ADVANCES 2023; 9:eadg5109. [PMID: 37831776 PMCID: PMC10575591 DOI: 10.1126/sciadv.adg5109] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 09/13/2023] [Indexed: 10/15/2023]
Abstract
Pancreatic carcinoma lacks effective therapeutic strategies resulting in poor prognosis. Transcriptional dysregulation due to alterations in KRAS and MYC affects initiation, development, and survival of this tumor type. Using patient-derived xenografts of KRAS- and MYC-driven pancreatic carcinoma, we show that coinhibition of topoisomerase 1 (TOP1) and bromodomain-containing protein 4 (BRD4) synergistically induces tumor regression by targeting promoter pause release. Comparing the nascent transcriptome with the recruitment of elongation and termination factors, we found that coinhibition of TOP1 and BRD4 disrupts recruitment of transcription termination factors. Thus, RNA polymerases transcribe downstream of genes for hundreds of kilobases leading to readthrough transcription. This occurs during replication, perturbing replisome progression and inducing DNA damage. The synergistic effect of TOP1 + BRD4 inhibition is specific to cancer cells leaving normal cells unaffected, highlighting the tumor's vulnerability to transcriptional defects. This preclinical study provides a mechanistic understanding of the benefit of combining TOP1 and BRD4 inhibitors to treat pancreatic carcinomas addicted to oncogenic drivers of transcription and replication.
Collapse
Affiliation(s)
- Donald P. Cameron
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jan Grosser
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Swetlana Ladigan
- Ruhr University Bochum, Faculty of Medicine, Department of Molecular GI Oncology, Bochum, Germany
- Ruhr University Bochum, Knappschaftskrankenhaus, Department of Internal Medicine, Bochum, Germany
| | - Vladislav Kuzin
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Evanthia Iliopoulou
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Anika Wiegard
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hajar Benredjem
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kathryn Jackson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sven T. Liffers
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Smiths Lueong
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Phyllis F. Cheung
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Deepak Vangala
- Ruhr University Bochum, Faculty of Medicine, Department of Molecular GI Oncology, Bochum, Germany
- Ruhr University Bochum, Knappschaftskrankenhaus, Department of Internal Medicine, Bochum, Germany
| | - Michael Pohl
- Ruhr University Bochum, Knappschaftskrankenhaus, Department of Internal Medicine, Bochum, Germany
| | - Richard Viebahn
- Ruhr University Bochum, Knappschaftskrankenhaus, Department of Surgery, Bochum, Germany
| | | | - Heiner Wolters
- Department of Visceral and General Surgery, St. Josef-Hospital, Dortmund, Germany
| | - Selami Usta
- Department of Visceral and General Surgery, St. Josef-Hospital, Dortmund, Germany
| | - Keyi Geng
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Claudia Kutter
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | | | - Jens T. Siveke
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | | | - Wolff Schmiegel
- Ruhr University Bochum, Knappschaftskrankenhaus, Department of Internal Medicine, Bochum, Germany
| | - Stephan A. Hahn
- Ruhr University Bochum, Faculty of Medicine, Department of Molecular GI Oncology, Bochum, Germany
| | - Laura Baranello
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
Velez J, Kaniskan HÜ, Jin J. Recent advances in developing degraders & inhibitors of lysine methyltransferases. Curr Opin Chem Biol 2023; 76:102356. [PMID: 37379717 PMCID: PMC10527319 DOI: 10.1016/j.cbpa.2023.102356] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/30/2023]
Abstract
Over the last several decades, there has been continued interest in developing novel therapeutic approaches targeting protein lysine methyltransferases (PKMTs). Along with PKMT inhibitors, targeted protein degradation (TPD) has emerged as a promising strategy to attenuate aberrant PKMT activity. Particularly, proteolysis targeting chimeras (PROTACs) effectively eliminate PKMTs of interest, suppressing all enzymatic and non-enzymatic functions. PROTACs and other TPD approaches add new depth to PKMT research and novel therapeutics discovery. This review focuses on recent advances in PKMT degrader and inhibitor development over the last several years.
Collapse
Affiliation(s)
- Julia Velez
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - 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, New York, NY 10029, United States.
| | - 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, New York, NY 10029, United States.
| |
Collapse
|
46
|
Xue W, Jian W, Meng Y, Wang T, Cai L, Yu Y, Yu Y, Xia Z, Zhang C. Knockdown of SETD2 promotes erastin-induced ferroptosis in ccRCC. Cell Death Dis 2023; 14:539. [PMID: 37604811 PMCID: PMC10442429 DOI: 10.1038/s41419-023-06057-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/15/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancer and is associated with poor prognosis. The histone H3 lysine 36 methyltransferase SET-domain-containing 2 (SETD2) has been reported to be expressed at low levels and frequently mutated in ccRCC. Ferroptosis, a form of death distinct from apoptosis and necrosis, has been reported in recent years in renal cancer. However, the relationship between SETD2 and ferroptosis in renal cancer is not clear. Here, we demonstrated that SETD2 was expressed at low levels in ccRCC and was associated with poor prognosis. Moreover, we found that knockdown of SETD2 increased lipid peroxidation and Fe2+ levels in tumor cells, thereby increasing the sensitivity of erastin, a ferroptosis inducer. Mechanistically, histone H3 lysine 36 trimethylation (H3K36me3) which was catalyzed by SETD2, interacted with the promoter of ferrochelatase (FECH) to regulate its transcription and ferroptosis-related signaling pathways. In conclusion, the presesnt study revealed that knockdown of the epigenetic molecule, SETD2, significantly increases the sensitivity of ferroptosis inducers which promotes tumor cell death, thereby indicating that SETD2 may be a potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Wei Xue
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Wengang Jian
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Yuyang Meng
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Tengda Wang
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Licheng Cai
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Yongchun Yu
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Yipeng Yu
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Zhinan Xia
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Cheng Zhang
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
- Department of Urology, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
| |
Collapse
|
47
|
Zhu JY, Liu C, Huang X, van de Leemput J, Lee H, Han Z. H3K36 Di-Methylation Marks, Mediated by Ash1 in Complex with Caf1-55 and MRG15, Are Required during Drosophila Heart Development. J Cardiovasc Dev Dis 2023; 10:307. [PMID: 37504562 PMCID: PMC10380788 DOI: 10.3390/jcdd10070307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023] Open
Abstract
Methyltransferases regulate transcriptome dynamics during development and aging, as well as in disease. Various methyltransferases have been linked to heart disease, through disrupted expression and activity, and genetic variants associated with congenital heart disease. However, in vivo functional data for many of the methyltransferases in the context of the heart are limited. Here, we used the Drosophila model system to investigate different histone 3 lysine 36 (H3K36) methyltransferases for their role in heart development. The data show that Drosophila Ash1 is the functional homolog of human ASH1L in the heart. Both Ash1 and Set2 H3K36 methyltransferases are required for heart structure and function during development. Furthermore, Ash1-mediated H3K36 methylation (H3K36me2) is essential for healthy heart function, which depends on both Ash1-complex components, Caf1-55 and MRG15, together. These findings provide in vivo functional data for Ash1 and its complex, and Set2, in the context of H3K36 methylation in the heart, and support a role for their mammalian homologs, ASH1L with RBBP4 and MORF4L1, and SETD2, during heart development and disease.
Collapse
Affiliation(s)
- Jun-yi Zhu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chen Liu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xiaohu Huang
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hangnoh Lee
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
48
|
Zhu JY, van de Leemput J, Han Z. The Roles of Histone Lysine Methyltransferases in Heart Development and Disease. J Cardiovasc Dev Dis 2023; 10:305. [PMID: 37504561 PMCID: PMC10380575 DOI: 10.3390/jcdd10070305] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023] Open
Abstract
Epigenetic marks regulate the transcriptomic landscape by facilitating the structural packing and unwinding of the genome, which is tightly folded inside the nucleus. Lysine-specific histone methylation is one such mark. It plays crucial roles during development, including in cell fate decisions, in tissue patterning, and in regulating cellular metabolic processes. It has also been associated with varying human developmental disorders. Heart disease has been linked to deregulated histone lysine methylation, and lysine-specific methyltransferases (KMTs) are overrepresented, i.e., more numerous than expected by chance, among the genes with variants associated with congenital heart disease. This review outlines the available evidence to support a role for individual KMTs in heart development and/or disease, including genetic associations in patients and supporting cell culture and animal model studies. It concludes with new advances in the field and new opportunities for treatment.
Collapse
Affiliation(s)
- Jun-yi Zhu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
49
|
Gong S, Ge Y, Wei Y, Gao Y. Genomic insights into the genetic basis of eagle-beak jaw, large head, and long tail in the big-headed turtle. Ecol Evol 2023; 13:e10361. [PMID: 37502307 PMCID: PMC10368965 DOI: 10.1002/ece3.10361] [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: 02/13/2023] [Revised: 06/15/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
The big-headed turtle (Platysternon megacephalum) is an endemic chelonian species in Asia. Unlike most other turtles in the world, P. megacephalum is characterized with eagle-beak jaw, large head, and long tail. Although these unique characteristics are well recognized, the underlying genetic basis remains largely elusive. Here, we performed comparative genomic analysis between P. megacephalum and other representative species, aiming to reveal the genetic basis of the unique morphological features. Our results revealed that the eagle-beak jaw is most likely enabled by combined effects of expansion of SFRP5, extraction of FGF11, and mutation of both ZFYVE16 and PAX6. Large head is supported by mutations of SETD2 and FGRF2 and copy number variations of six head circumference modulation-related genes (TGFBR2, Twist2, Rdh10, Gas1, Chst11, and SNAP25). The long tail is probably involved in a genetic network comprising Gdf11, Lin 28, and HoxC12, two of which showed a consistent expression pattern with a model organism (mice). These findings suggest that expansion, extraction, and mutation of those genes may have profound effects on unique phenotypes of P. megacephalum.
Collapse
Affiliation(s)
- Shiping Gong
- College of Life Science and TechnologyJinan UniversityGuangzhouChina
| | - Yan Ge
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of ZoologyGuangdong Academy of SciencesGuangzhouChina
| | - Yufeng Wei
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of ZoologyGuangdong Academy of SciencesGuangzhouChina
| | - Yangchun Gao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of ZoologyGuangdong Academy of SciencesGuangzhouChina
| |
Collapse
|
50
|
Yin J, Qi TF, Li L, Wang Y. Targeted Profiling of Epitranscriptomic Reader, Writer, and Eraser Proteins Regulated by H3K36me3. Anal Chem 2023; 95:9672-9679. [PMID: 37296074 PMCID: PMC10372775 DOI: 10.1021/acs.analchem.3c01552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Trimethylation of lysine 36 on histone H3 (H3K36me3), an epigenetic mark associated with actively transcribed genes, plays an important role in multiple cellular processes, including transcription elongation, DNA methylation, DNA repair, etc. Aberrant expression and mutations of the main methyltransferase for H3K36me3, i.e., SET domain-containing 2 (SETD2), were shown to be associated with various cancers. Here, we performed targeted profiling of 154 epitranscriptomic reader, writer, and eraser (RWE) proteins using a scheduled liquid chromatography-parallel-reaction monitoring (LC-PRM) method coupled with the use of stable isotope-labeled (SIL) peptides as internal standards to investigate how H3K36me3 modulates the chromatin occupancies of epitranscriptomic RWE proteins. Our results showed consistent changes in chromatin occupancies of RWE proteins upon losses of H3K36me3 and H4K16ac and a role of H3K36me3 in recruiting METTL3 to chromatin following induction of DNA double-strand breaks. In addition, protein-protein interaction network and Kaplan-Meier survival analyses revealed the importance of METTL14 and TRMT11 in kidney cancer. Taken together, our work unveiled cross-talks between histone epigenetic marks (i.e., H3K36me3 and H4K16ac) and epitranscriptomic RWE proteins and uncovered the potential roles of these RWE proteins in H3K36me3-mediated biological processes.
Collapse
Affiliation(s)
- Jiekai Yin
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521-0403, United States
| | - Tianyu F Qi
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521-0403, United States
| | - Lin Li
- Deparment of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Yinsheng Wang
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521-0403, United States
- Deparment of Chemistry, University of California, Riverside, California 92521-0403, United States
| |
Collapse
|