1
|
ter Laak A, Hillig RC, Ferrara SJ, Korr D, Barak N, Lienau P, Herbert S, Fernández-Montalván AE, Neuhaus R, Gorjánácz M, Puetter V, Badock V, Bone W, Strathdee C, Siegel F, Schatz C, Nowak-Reppel K, Doehr O, Gradl S, Hartung IV, Meyerson M, Bouché L. Discovery and Characterization of BAY-184: A New Potent and Selective Acylsulfonamide-Benzofuran In Vivo-Active KAT6AB Inhibitor. J Med Chem 2024; 67:19282-19303. [PMID: 39450890 PMCID: PMC11571114 DOI: 10.1021/acs.jmedchem.4c01709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024]
Abstract
KAT6A and KAT6B genes are two closely related lysine acetyltransferases that transfer an acetyl group from acetyl coenzyme A (AcCoA) to lysine residues of target histone substrates, hence playing a key role in chromatin regulation. KAT6A and KAT6B genes are frequently amplified in various cancer types. In breast cancer, the 8p11-p12 amplicon occurs in 12-15% of cases, resulting in elevated copy numbers and expression levels of chromatin modifiers like KAT6A. Here, we report the discovery of a new acylsulfonamide-benzofuran series as a novel structural class for KAT6A/B inhibition. These compounds were identified through high-throughput screening and subsequently optimized using molecular modeling and cocrystal structure determination. The final tool compound, BAY-184 (29), was successfully validated in an in vivo proof-of-concept study.
Collapse
Affiliation(s)
- Antonius ter Laak
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Roman C. Hillig
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Steven J. Ferrara
- Broad
Institute of MIT and Harvard, Center for the Development of Therapeutics, 415 Main St., Cambridge, Massachusetts 02142, United States
| | - Daniel Korr
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Naomi Barak
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Philip Lienau
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Simon Herbert
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | | | - Roland Neuhaus
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Mátyás Gorjánácz
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Vera Puetter
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Volker Badock
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Wilhelm Bone
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Craig Strathdee
- Broad
Institute of MIT and Harvard, Center for the Development of Therapeutics, 415 Main St., Cambridge, Massachusetts 02142, United States
| | - Franziska Siegel
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Christoph Schatz
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Katrin Nowak-Reppel
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Olaf Doehr
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Stefan Gradl
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Ingo V. Hartung
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Matthew Meyerson
- Broad
Institute of MIT and Harvard, Center for the Development of Therapeutics, 415 Main St., Cambridge, Massachusetts 02142, United States
| | - Léa Bouché
- Bayer
AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| |
Collapse
|
2
|
Hasebe T, Fujimoto K, Ishizuya-Oka A. Stem cell development involves divergent thyroid hormone receptor subtype expression and epigenetic modifications in the amphibian intestine during metamorphosis. VITAMINS AND HORMONES 2023; 122:1-22. [PMID: 36863790 DOI: 10.1016/bs.vh.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In the amphibian intestine during metamorphosis, most of the larval epithelial cells undergo apoptosis, while a small number of the epithelial cells dedifferentiate into stem cells (SCs). The SCs actively proliferate and then newly generate the adult epithelium analogous to the mammalian counterpart, which is continuously renewed from the SCs throughout adulthood. This larval-to-adult intestinal remodeling can be experimentally induced by thyroid hormone (TH) through interacting with the surrounding connective tissue that develops as the stem cell niche. Thus, the amphibian intestine provides us a valuable opportunity to study how the SCs and their niche are formed during development. To clarify the TH-induced and evolutionally conserved mechanism of SC development at the molecular level, numerous TH response genes have been identified in the Xenopus laevis intestine over the last three decades and extensively analyzed for their expression and function by using wild-type and transgenic Xenopus tadpoles. Interestingly, accumulating evidence indicates that thyroid hormone receptor (TR) epigenetically regulates the expression of TH response genes involved in the remodeling. In this review, we highlight recent progress in the understanding of SC development, focusing on epigenetic gene regulation by TH/TR signaling in the X. laevis intestine. We here propose that two subtypes of TRs, TRα and TRβ, play distinct roles in the intestinal SC development via different histone modifications in different cell types.
Collapse
Affiliation(s)
- Takashi Hasebe
- Department of Biology, Nippon Medical School, Tokyo, Japan.
| | - Kenta Fujimoto
- Department of Biology, Nippon Medical School, Tokyo, Japan
| | | |
Collapse
|
3
|
Shvedunova M, Akhtar A. Modulation of cellular processes by histone and non-histone protein acetylation. Nat Rev Mol Cell Biol 2022; 23:329-349. [PMID: 35042977 DOI: 10.1038/s41580-021-00441-y] [Citation(s) in RCA: 368] [Impact Index Per Article: 122.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2021] [Indexed: 12/12/2022]
Abstract
Lysine acetylation is a widespread and versatile protein post-translational modification. Lysine acetyltransferases and lysine deacetylases catalyse the addition or removal, respectively, of acetyl groups at both histone and non-histone targets. In this Review, we discuss several features of acetylation and deacetylation, including their diversity of targets, rapid turnover, exquisite sensitivity to the concentrations of the cofactors acetyl-CoA, acyl-CoA and NAD+, and tight interplay with metabolism. Histone acetylation and non-histone protein acetylation influence a myriad of cellular and physiological processes, including transcription, phase separation, autophagy, mitosis, differentiation and neural function. The activity of lysine acetyltransferases and lysine deacetylases can, in turn, be regulated by metabolic states, diet and specific small molecules. Histone acetylation has also recently been shown to mediate cellular memory. These features enable acetylation to integrate the cellular state with transcriptional output and cell-fate decisions.
Collapse
Affiliation(s)
- Maria Shvedunova
- Department of Chromatin Regulation, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Asifa Akhtar
- Department of Chromatin Regulation, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany.
| |
Collapse
|
4
|
AKT Isoforms in Macrophage Activation, Polarization, and Survival. Curr Top Microbiol Immunol 2022; 436:165-196. [PMID: 36243844 DOI: 10.1007/978-3-031-06566-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
5
|
Saglam O, Tang Z, Tang G, Medeiros LJ, Toruner GA. KAT6A amplifications are associated with shorter progression-free survival and overall survival in patients with endometrial serous carcinoma. PLoS One 2020; 15:e0238477. [PMID: 32877461 PMCID: PMC7467277 DOI: 10.1371/journal.pone.0238477] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/17/2020] [Indexed: 11/18/2022] Open
Abstract
Somatic copy number alterations (CNA) are common in endometrial serous carcinoma (ESC). We used the Tumor Cancer Genome Atlas Pan Cancer dataset (TCGA Pan Can) to explore the impact of somatic CNA and gene expression levels (mRNA) of cancer-related genes in ESC. Results were correlated with clinico-pathologic parameters such as age of onset, disease stage, progression-free survival (PFS) and overall survival (OS) (n = 108). 1,449 genes with recurrent somatic CNA were identified, observed in 10% or more tumor samples. Somatic CNA and mRNA expression levels were highly correlated (r> = 0.6) for 383 genes. Among these, 45 genes were classified in the Tier 1 category of Cancer Genome Census-Catalogue of Somatic Mutations in Cancer. Eighteen of 45 Tier 1 genes had highly correlated somatic CNA and mRNA expression levels including ARNT, PIK3CA, TBLXR1, ASXL1, EIF4A2, HOOK3, IKBKB, KAT6A, TCEA1, KAT6B, ERBB2, BRD4, KEAP1, PRKACA, DNM2, SMARCA4, AKT2, SS18L1. Our results are in agreement with previously reported somatic CNA for ERBB2, BRD4 and PIK3C in ESC. In addition, AKT2 (p = 0.002) and KAT6A (p = 0.015) amplifications were more frequent in tumor samples from younger patients (<60), and CEBPA (p = 0.028) and MYC (p = 0.023) amplifications were more common with advanced (stage III and IV) disease stage. Patients with tumors carrying KAT6A and MYC amplifications had shorter PFS and OS. The hazard ratio (HR) of KAT6A was 2.82 [95 CI 1.12-7.07] for PFS and 3.87 [95 CI 1.28-11.68] for OS. The HR of MYC was 2.25 [95 CI 1.05-4.81] and 2.62[95 CI 1.07-6.41] for PFS and OS, respectively.
Collapse
Affiliation(s)
- Ozlen Saglam
- Department of Surgical Pathology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Zhenya Tang
- Department of Hematopathology, Section of Clinical Cytogenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Guilin Tang
- Department of Hematopathology, Section of Clinical Cytogenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - L. Jeffrey Medeiros
- Department of Hematopathology, Section of Clinical Cytogenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Gokce A. Toruner
- Department of Hematopathology, Section of Clinical Cytogenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
6
|
Yin Q, Shen L, Qi Y, Song D, Ye L, Peng Y, Wang Y, Jin Z, Ning G, Wang W, Lin D, Wang S. Decreased SIRT1 expression in the peripheral blood of patients with Graves' disease. J Endocrinol 2020; 246:161-173. [PMID: 32485674 PMCID: PMC7354706 DOI: 10.1530/joe-19-0501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/02/2020] [Indexed: 11/08/2022]
Abstract
SIRT1, a class III histone/protein deacetylase (HDAC), has been associated with autoimmune diseases. There is a paucity of data about the role of SIRT1 in Graves' disease. The aim of this study was to investigate the role of SIRT1 in the pathogenesis of GD. Here, we showed that SIRT1 expression and activity were significantly decreased in GD patients compared with healthy controls. The NF-κB pathway was activated in the peripheral blood of GD patients. The reduced SIRT1 levels correlated strongly with clinical parameters. In euthyroid patients, SIRT1 expression was markedly upregulated and NF-κB downstream target gene expression was significantly reduced. SIRT1 inhibited the NF-κB pathway activity by deacetylating P65. These results demonstrate that reduced SIRT1 expression and activity contribute to the activation of the NF-κB pathway and may be involved in the pathogenesis of GD.
Collapse
Affiliation(s)
- Qinglei Yin
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Liyun Shen
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Yicheng Qi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, Shanghai Jiao-Tong University School of Medicine, Pudong, Shanghai, China
| | - Dalong Song
- Reproductive Medicine Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Lei Ye
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Ying Peng
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Yanqiu Wang
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Zhou Jin
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Guang Ning
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Weiqing Wang
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Dongping Lin
- Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Affiliated Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- Correspondence should be addressed to D Lin and S Wang: or
| | - Shu Wang
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
- Correspondence should be addressed to D Lin and S Wang: or
| |
Collapse
|
7
|
Hasebe T, Fujimoto K, Buchholz DR, Ishizuya-Oka A. Stem cell development involves divergent thyroid hormone receptor subtype expression and epigenetic modifications in the Xenopus metamorphosing intestine. Gen Comp Endocrinol 2020; 292:113441. [PMID: 32084349 DOI: 10.1016/j.ygcen.2020.113441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/22/2020] [Accepted: 02/16/2020] [Indexed: 12/13/2022]
Abstract
In the intestine during metamorphosis of the frog Xenopus laevis, most of the larval epithelial cells are induced to undergo apoptosis by thyroid hormone (TH), and under continued TH action, the remaining epithelial cells dedifferentiate into stem cells (SCs), which then newly generate an adult epithelium analogous to the mammalian intestinal epithelium. Previously, we have shown that the precursors of the SCs that exist in the larval epithelium as differentiated absorptive cells specifically express receptor tyrosine kinase-like orphan receptor 2 (Ror2). By using Ror2 as a marker, we have immunohistochemically shown here that these SC precursors, but not the larval epithelial cells destined to die by apoptosis, express TH receptor α (TRα). Upon initiation of TH-dependent remodeling, TRα expression remains restricted to the SCs as well as proliferating adult epithelial primordia derived from them. As intestinal folds form, TRα expression becomes localized in the trough of the folds where the SCs reside. In contrast, TRβ expression is transiently up-regulated in the entire intestine concomitantly with the increase of endogenous TH levels and is most highly expressed in the developing adult epithelial primordia. Moreover, we have shown here that global histone H4 acetylation is enhanced in the SC precursors and adult primordia including the SCs, while tri-methylation of histone H3 lysine 27 is lacking in those cells during metamorphosis. Our results strongly suggest distinct roles of TRα and TRβ in the intestinal larval-to-adult remodeling, involving distinctive epigenetic modifications in the SC lineage.
Collapse
Affiliation(s)
- Takashi Hasebe
- Department of Biology, Nippon Medical School, Kyonan-cho, Musashino, Tokyo, Japan
| | - Kenta Fujimoto
- Department of Biology, Nippon Medical School, Kyonan-cho, Musashino, Tokyo, Japan
| | - Daniel R Buchholz
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Atsuko Ishizuya-Oka
- Department of Biology, Nippon Medical School, Kyonan-cho, Musashino, Tokyo, Japan.
| |
Collapse
|
8
|
Jiang Y, Guo X, Liu L, Rode S, Wang R, Liu H, Yang ZQ. Metagenomic characterization of lysine acetyltransferases in human cancer and their association with clinicopathologic features. Cancer Sci 2020; 111:1829-1839. [PMID: 32162442 PMCID: PMC7226209 DOI: 10.1111/cas.14385] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/05/2020] [Accepted: 03/07/2020] [Indexed: 12/19/2022] Open
Abstract
Lysine acetyltransferases (KATs) are a highly diverse group of epigenetic enzymes that play important roles in various cellular processes including transcription, signal transduction, and cellular metabolism. However, our knowledge of the genomic and transcriptomic alterations of KAT genes and their clinical significance in human cancer remains incomplete. We undertook a metagenomic analysis of 37 KATs in more than 10 000 cancer samples across 33 tumor types, focusing on breast cancer. We identified associations among recurrent genetic alteration, gene expression, clinicopathologic features, and patient survival. Loss‐of‐function analysis was carried out to examine which KAT has important roles in growth and viability of breast cancer cells. We identified that a subset of KAT genes, including NAA10, KAT6A, and CREBBP, have high frequencies of genomic amplification or mutation in a spectrum of human cancers. Importantly, we found that 3 KATs, NAA10, ACAT2, and BRD4, were highly expressed in the aggressive basal‐like subtype, and their expression was significantly associated with disease‐free survival. Furthermore, we showed that depletion of NAA10 inhibits basal‐like breast cancer growth in vitro. Our findings provide a strong foundation for further mechanistic research and for developing therapies that target NAA10 or other KATs in human cancer.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xuhui Guo
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Breast Surgery, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Lanxin Liu
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Shomita Rode
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Rui Wang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Diagnostics of Chinese Medicine, Hebei University of Chinese Medicine, Hebei, China
| | - Hui Liu
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Zeng-Quan Yang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
9
|
Decline of p300 contributes to cell senescence and growth inhibition of hUC-MSCs through p53/p21 signaling pathway. Biochem Biophys Res Commun 2019; 515:24-30. [PMID: 31122700 DOI: 10.1016/j.bbrc.2019.05.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 05/07/2019] [Indexed: 02/08/2023]
Abstract
Human umbilical cord-derived mesenchymal stromal cells (hUC-MSCs) in vitro expansion for long term may undergo epigenetic and genetic alterations that subsequently induce cellular senescence and associated growth inhibition. Increasing evidence implicated that aberrant histone acetylation modulates gene expression responsible for MSCs aging. Whether the dysregulation of p300 and its KAT activity is involved in the aging process of MSCs was still unexplored. In this study, we found a significant decrease of p300 but elevated p53/p21 levels in senescent hUC-MSCs at late-passage. Then we used two different approaches: (i) downregulation of p300 by siRNA and (ii) inhibition of the acetyltransferase(KAT) activity by C646 to determine the role of p300 in regulating MSCs senescence. We showed that inhibition of p300 induce premature senescence and decrease proliferation potential in hUC-MSCs. Moreover, upregulations of p53 and p21 expressions were confirmed in p300 knockdown and C646-treated hUC-MSCs. Taken together, these results suggest that p300 plays an important role in aging process of MSCs associated with activation of p53/p21 signaling pathway.
Collapse
|
10
|
Huai W, Liu X, Wang C, Zhang Y, Chen X, Chen X, Xu S, Thomas T, Li N, Cao X. KAT8 selectively inhibits antiviral immunity by acetylating IRF3. J Exp Med 2019; 216:772-785. [PMID: 30842237 PMCID: PMC6446880 DOI: 10.1084/jem.20181773] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/03/2019] [Accepted: 02/01/2019] [Indexed: 12/11/2022] Open
Abstract
Optimal activation of IRF3 is crucial for maintaining immune homeostasis. Huai et al. demonstrate that KAT8 acetylates IRF3 at lysine 359, inhibits IRF3 recruitment to promoters of type I interferon genes, and then decreases type I interferon production to attenuate antiviral innate immune responses. The transcription factor interferon regulatory factor 3 (IRF3) is essential for virus infection–triggered induction of type I interferons (IFN-I) and innate immune responses. IRF3 activity is tightly regulated by conventional posttranslational modifications (PTMs) such as phosphorylation and ubiquitination. Here, we identify an unconventional PTM of IRF3 that directly inhibits its transcriptional activity and attenuates antiviral immune response. We performed an RNA interference screen and found that lysine acetyltransferase 8 (KAT8), which is ubiquitously expressed in immune cells (particularly in macrophages), selectively inhibits RNA and DNA virus–triggered IFN-I production in macrophages and dendritic cells. KAT8 deficiency protects mice from viral challenge by enhancing IFN-I production. Mechanistically, KAT8 directly interacts with IRF3 and mediates IRF3 acetylation at lysine 359 via its MYST domain. KAT8 inhibits IRF3 recruitment to IFN-I gene promoters and decreases the transcriptional activity of IRF3. Our study reveals a critical role for KAT8 and IRF3 lysine acetylation in the suppression of antiviral innate immunity.
Collapse
Affiliation(s)
- Wanwan Huai
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingguang Liu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Chunmei Wang
- Department of Immunology and Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yunkai Zhang
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Xi Chen
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiang Chen
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Sheng Xu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Tim Thomas
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Victoria, Australia
| | - Nan Li
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Xuetao Cao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China .,National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China.,Department of Immunology and Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,College of Life Science, Nankai University, Tianjin, China
| |
Collapse
|
11
|
Kutateladze TG, Gozani O, Bienz M, Ostankovitch M. Histone modifications for chromatin dynamics and cellular plasticity. J Mol Biol 2019. [PMID: 28623961 DOI: 10.1016/j.jmb.2017.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
12
|
Lin H, Wang B, Yu J, Wang J, Li Q, Cao B. Protein arginine methyltransferase 8 gene enhances the colon cancer stem cell (CSC) function by upregulating the pluripotency transcription factor. J Cancer 2018; 9:1394-1402. [PMID: 29721049 PMCID: PMC5929084 DOI: 10.7150/jca.23835] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/25/2018] [Indexed: 12/12/2022] Open
Abstract
Objective: Cancer stem cells play a crucial role in tumor multidrug resistance and metastasis, which can produce heterogeneous tumor cells and have self-renewal ability. The related literature reported that PRMT8 was overexpressed in tumor stem cells and pluripotent stem cells. However, it's unclear how PRMT8 acts on the stemness of colon tumor cells. This study is designed to detect functions by transfecting with PRMT8 plasmid to colon cancer cells. Methods: In this study we investigated colon cancer cell sphere and its differential expression of PRMT8 compared with colon cancer cells grown by static adherence. RKO Sphere formation assay was used to identify CSCs and verified PRMT8 and pluripotent transcription factors SOX2, OCT4, Nanog expression level in colon cell sphere. Colon cancer cell HCT-8 and RKO up-regulated PRMT8 expression by being transfected with PRMT8 plasmid to evaluate its effect on the stemness of colon tumor cell. Results: In RKO cell sphere, stem cell surface marker CD133 and CD44 were highly expressed. And PRMT8, SOX2, OCT4 and Nanog were also highly expressed in RKO cell sphere. After PRMT8 was up-regulated in HCT-8 and RKO cells, flow cytometry proved that PRMT8 group cells have a significant increase of the side population (SP) cells with cancer stem cell surface markers CD133 and CD44. And overexpression of PRMT8 in HCT-8 and RKO cells facilitated their aggressive traits, which contained proliferation, invasion and migration, as well as leading to their drug resistance. PRMT8 may play a role in colon cancer stem cells (CSC) through its regulation of pluripotent transcription factors, such as Nanog Homeobox (Nanog), octamer-binding transcription factor-4 (Oct4) and SRY-related high-mobility-group(HMG)-box protein-2 (Sox2). Conclusion: PRMT8 may promote the formation of colon cancer stem cells and, thus, be considered a potential therapeutic target for the treatment of malignant colon tumor.
Collapse
Affiliation(s)
- Haishan Lin
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bin Wang
- Department of Medical Administration, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jing Yu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jing Wang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Qin Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bangwei Cao
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
13
|
Han N, Shi L, Guo Q, Sun W, Yu Y, Yang L, Zhang X, Zhang M. HAT1 induces lung cancer cell apoptosis via up regulating Fas. Oncotarget 2017; 8:89970-89977. [PMID: 29163803 PMCID: PMC5685724 DOI: 10.18632/oncotarget.21205] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/17/2017] [Indexed: 12/25/2022] Open
Abstract
The dysfunction of apoptosis is one of the factors contributing to lung cancer (LC) growth. Histone acetyltransferase HAT1 can up regulate cell apoptosis. This study aims to investigate the mechanism by which HAT1 induces LC cell (LCC) apoptosis via up regulating the expression of Fas. In this study, the surgically removed human LC tissues were collected. LCCs were isolated from the LC tissues and analyzed for the expression of HAT1 and Fas by RT-qPCR and Western blotting. We observed that the expression of Fas was negatively correlated with PAR2 in LCCs. Activation of PAR2 suppressed the expression of Fas in normal lung epithelial cells. The expression of HAT1 was lower and positively correlated with Fas expression and negatively correlated with PAR2 expression in LCCs. Activation of PAR2 suppressed Fas expression in lung epithelial cells via inhibiting HAT1. Restoration of HAT1 expression restored Fas expression in LCCs and induced LCC apoptosis. In conclusion, less expression of HAT1 in LCCs was associated with the pathogenesis of LC. Up regulation of HAT1 expression in LCCs can induce LCCs apoptosis, which may be a potential novel therapy for the treatment of LC.
Collapse
Affiliation(s)
- Na Han
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Shi
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyun Guo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Yu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxi Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengxian Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Chromatin remodeling system p300-HDAC2-Sin3A is involved in Arginine Starvation-Induced HIF-1α Degradation at the ASS1 promoter for ASS1 Derepression. Sci Rep 2017; 7:10814. [PMID: 28883660 PMCID: PMC5589935 DOI: 10.1038/s41598-017-11445-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/17/2017] [Indexed: 12/17/2022] Open
Abstract
Argininosuccinate synthetase 1 (ASS1) is the key enzyme that controls biosynthesis of arginine (Arg). ASS1 is silenced in many human malignancies therefore, these tumors require extracellular Arg for growth. The Arg-degrading recombinant protein, pegylated arginine deiminase (ADI-PEG20), has been in clinical trials for targeting Arg auxotrophic tumors by Arg starvation therapy. Resistance to Arg starvation is often developed through reactivation of ASS1 expression. We previously demonstrated that ASS1 silencing is controlled by HIF-1α and Arg starvation-reactivated ASS1 is associated with HIF-1α downregulation. However, mechanisms underlying ASS1 repression and HIF-1α turnover are not known. Here, we demonstrate that interplay of p300-HDAC2-Sin3A in the chromatin remodeling system is involved in HIF-1α degradation at the ASS1 promoter. The histone acetyltransferase p300 is normally associated with the ASS1 promoter to maintain acetylated H3K14ac and H3K27ac for ASS1 silencing. Arg starvation induces p300 dissociation, allowing histone HDAC2 and cofactor Sin3A to deacetylate these histones at the ASS1 promoter, thereby facilitating HIF-1α-proteasomal complex, driven by PHD2, to degrade HIF-1α in situ. Arg starvation induces PHD2 and HDAC2 interaction which is sensitive to antioxidants. This is the first report describing epigenetic regulation of chromosomal HIF-1α turnover in gene activation that bears important implication in cancer therapy.
Collapse
|
15
|
Ackloo S, Brown PJ, Müller S. Chemical probes targeting epigenetic proteins: Applications beyond oncology. Epigenetics 2017; 12:378-400. [PMID: 28080202 PMCID: PMC5453191 DOI: 10.1080/15592294.2017.1279371] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 12/23/2016] [Accepted: 01/02/2017] [Indexed: 12/15/2022] Open
Abstract
Epigenetic chemical probes are potent, cell-active, small molecule inhibitors or antagonists of specific domains in a protein; they have been indispensable for studying bromodomains and protein methyltransferases. The Structural Genomics Consortium (SGC), comprising scientists from academic and pharmaceutical laboratories, has generated most of the current epigenetic chemical probes. Moreover, the SGC has shared about 4 thousand aliquots of these probes, which have been used primarily for phenotypic profiling or to validate targets in cell lines or primary patient samples cultured in vitro. Epigenetic chemical probes have been critical tools in oncology research and have uncovered mechanistic insights into well-established targets, as well as identify new therapeutic starting points. Indeed, the literature primarily links epigenetic proteins to oncology, but applications in inflammation, viral, metabolic and neurodegenerative diseases are now being reported. We summarize the literature of these emerging applications and provide examples where existing probes might be used.
Collapse
Affiliation(s)
- Suzanne Ackloo
- Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Peter J. Brown
- Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Susanne Müller
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Straβe 15, Frankfurt am Main, Germany
| |
Collapse
|
16
|
Rabhi N, Hannou SA, Froguel P, Annicotte JS. Cofactors As Metabolic Sensors Driving Cell Adaptation in Physiology and Disease. Front Endocrinol (Lausanne) 2017; 8:304. [PMID: 29163371 PMCID: PMC5675844 DOI: 10.3389/fendo.2017.00304] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022] Open
Abstract
Chromatin architectures and epigenetic fingerprint regulation are fundamental for genetically determined biological processes. Chemical modifications of the chromatin template sensitize the genome to intracellular metabolism changes to set up diverse functional adaptive states. Accumulated evidence suggests that the action of epigenetic modifiers is sensitive to changes in dietary components and cellular metabolism intermediates, linking nutrition and energy metabolism to gene expression plasticity. Histone posttranslational modifications create a code that acts as a metabolic sensor, translating changes in metabolism into stable gene expression patterns. These observations support the notion that epigenetic reprograming-linked energy input is connected to the etiology of metabolic diseases and cancer. In the present review, we introduce the role of epigenetic cofactors and their relation with nutrient intake and we question the links between epigenetic regulation and the development of metabolic diseases.
Collapse
Affiliation(s)
- Nabil Rabhi
- Lille University, UMR 8199—EGID, Lille, France
- CNRS, UMR 8199, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Sarah Anissa Hannou
- Lille University, UMR 8199—EGID, Lille, France
- CNRS, UMR 8199, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Philippe Froguel
- Lille University, UMR 8199—EGID, Lille, France
- CNRS, UMR 8199, Lille, France
- Institut Pasteur de Lille, Lille, France
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Jean-Sébastien Annicotte
- Lille University, UMR 8199—EGID, Lille, France
- CNRS, UMR 8199, Lille, France
- Institut Pasteur de Lille, Lille, France
- *Correspondence: Jean-Sébastien Annicotte,
| |
Collapse
|