1
|
Wang M, Mu G, Qiu B, Wang S, Tao C, Mao Y, Zhao X, Liu J, Chen K, Li Z, Wang W, Yang E, Yang Y. Competitive antagonism of KAT7 crotonylation against acetylation affects procentriole formation and colorectal tumorigenesis. Nat Commun 2025; 16:2379. [PMID: 40064919 PMCID: PMC11893896 DOI: 10.1038/s41467-025-57546-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Accurate procentriole formation is critical for centriole duplication. However, the holistic transcriptional regulatory mechanisms underlying this process remain elusive. Here, we show that KAT7 crotonylation, facilitated by the crotonyltransferase hMOF, competes against its acetylation regulated by the deacetylase HDAC2 at the K432 residue upon DNA damage stimulation. This competition diminishes its histone acetyltransferase activity, leading to the inhibition of procentriole formation in colorectal cancer cells. Mechanistically, the reduction of KAT7 histone acetyltransferase activity by the antagonistic effect of KAT7 crotonylation against its acetylation decreases the gene expression associated with procentriole formation by modulating the enrichment of H3K14ac at their promoters and plays an important role in colorectal tumorigenesis. Furthermore, KAT7 crotonylation and acetylation are associated with the prognosis in colorectal cancer patients. Collectively, our findings uncover a previously unidentified role of KAT7 in the regulation of procentriole formation and colorectal tumorigenesis via competitive antagonism of its crotonylation against acetylation.
Collapse
Affiliation(s)
- Meng Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Guanqun Mu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Bingquan Qiu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Shuo Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Changyu Tao
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yutong Mao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xinhui Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jiansong Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Keyu Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ziyu Li
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Weibin Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ence Yang
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Yang Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China.
| |
Collapse
|
2
|
Su Z, Tian M, Shibata E, Shibata Y, Yang T, Wang Z, Jin F, Zang C, Dutta A. Regulation of epigenetics and chromosome structure by human ORC2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.18.629220. [PMID: 39829907 PMCID: PMC11741241 DOI: 10.1101/2024.12.18.629220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The six subunit Origin Recognition Complex (ORC) is a DNA replication initiator that also promotes heterochromatinization in some species. A multi-omics study in a human cell line with mutations in three subunits of ORC, reveals that the subunits bind to DNA independent of each other rather than as part of a common six-subunit ORC. While DNA-bound ORC2 was seen to compact chromatin and attract repressive histone marks, the activation of chromatin and protection from repressive marks was seen at a large number of sites. The epigenetic changes regulate hundreds of genes, including some epigenetic regulators, adding an indirect mechanism by which ORC2 regulates epigenetics without local binding. DNA-bound ORC2 also prevents the acquisition of CTCF at focal sites in the genome to regulate chromatin loops. Thus, individual ORC subunits are major regulators, in both directions, of epigenetics, gene expression and chromosome structure, independent of the role of ORC in replication.
Collapse
|
3
|
Keller MA, Nakamura M. Acetyltransferase in cardiovascular disease and aging. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:10.20517/jca.2024.21. [PMID: 39958699 PMCID: PMC11827898 DOI: 10.20517/jca.2024.21] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
Acetyltransferases are enzymes that catalyze the transfer of an acetyl group to a substrate, a modification referred to as acetylation. Loss-of-function variants in genes encoding acetyltransferases can lead to congenital disorders, often characterized by intellectual disability and heart and muscle defects. Their activity is influenced by dietary nutrients that alter acetyl coenzyme A levels, a key cofactor. Cardiovascular diseases, including ischemic, hypertensive, and diabetic heart diseases - leading causes of mortality in the elderly - are largely attributed to prolonged lifespan and the growing prevalence of metabolic syndrome. Acetyltransferases thus serve as a crucial link between lifestyle modifications, cardiometabolic disease, and aging through both epigenomic and non-epigenomic mechanisms. In this review, we discuss the roles and relevance of acetyltransferases. While the sirtuin family of deacetylases has been extensively studied in longevity, particularly through fasting-mediated NAD+ metabolism, recent research has brought attention to the essential roles of acetyltransferases in health and aging-related pathways, including cell proliferation, DNA damage response, mitochondrial function, inflammation, and senescence. We begin with an overview of acetyltransferases, classifying them by domain structure, including canonical and non-canonical lysine acetyltransferases, N-terminal acetyltransferases, and sialic acid O-acetyltransferases. We then discuss recent advances in understanding acetyltransferase-related pathologies, particularly focusing on cardiovascular disease and aging, and explore their potential therapeutic applications for promoting health in older individuals.
Collapse
Affiliation(s)
- Mariko Aoyagi Keller
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
4
|
Malysa A, Zhang XM, Bepler G. Minichromosome Maintenance Proteins: From DNA Replication to the DNA Damage Response. Cells 2024; 14:12. [PMID: 39791713 PMCID: PMC11719910 DOI: 10.3390/cells14010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
The DNA replication machinery is highly conserved from bacteria to eukaryotic cells. Faithful DNA replication is vital for cells to transmit accurate genetic information to the next generation. However, both internal and external DNA damages threaten the intricate DNA replication process, leading to the activation of the DNA damage response (DDR) system. Dysfunctional DNA replication and DDR are a source of genomic instability, causing heritable mutations that drive cancer evolutions. The family of minichromosome maintenance (MCM) proteins plays an important role not only in DNA replication but also in DDR. Here, we will review the current strides of MCM proteins in these integrated processes as well as the acetylation/deacetylation of MCM proteins and the value of MCMs as biomarkers in cancer.
Collapse
Affiliation(s)
| | | | - Gerold Bepler
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, 4100 John R Street, Detroit, MI 48201, USA; (A.M.); (X.M.Z.)
| |
Collapse
|
5
|
Su Z, Zhang Y, Tang J, Zhou Y, Long C. Multifunctional acyltransferase HBO1: a key regulatory factor for cellular functions. Cell Mol Biol Lett 2024; 29:141. [PMID: 39543485 PMCID: PMC11566351 DOI: 10.1186/s11658-024-00661-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
HBO1, also known as KAT7 or MYST2, is a crucial histone acetyltransferase with diverse cellular functions. It typically forms complexes with protein subunits or cofactors such as MEAF6, ING4, or ING5, and JADE1/2/3 or BRPF1/2/3, where the BRPF or JADE proteins serve as the scaffold targeting histone H3 or H4, respectively. The histone acetylation mediated by HBO1 plays significant roles in DNA replication and gene expression regulation. Additionally, HBO1 catalyzes the modification of proteins through acylation with propionyl, butyryl, crotonyl, benzoyl, and acetoacetyl groups. HBO1 undergoes ubiquitination and degradation by two types of ubiquitin complexes and can also act as an E3 ubiquitin ligase for the estrogen receptor α (ERα). Moreover, HBO1 participates in the expansion of medullary thymic epithelial cells (mTECs) and regulates the expression of peripheral tissue genes (PTGs) mediated by autoimmune regulator (AIRE), thus inducing immune tolerance. Furthermore, HBO1 influences the renewal of hematopoietic stem cells and the development of neural stem cells significantly. Importantly, the overexpression of HBO1 in various cancers suggests its carcinogenic role and potential as a therapeutic target. This review summarizes recent advancements in understanding HBO1's involvement in acylation modification, DNA replication, ubiquitination, immunity, and stem cell renewal.
Collapse
Affiliation(s)
- Zhanhuan Su
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Yang Zhang
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Jingqiong Tang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410078, Hunan, China.
| | - Chen Long
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
6
|
Yokoyama A, Niida H, Kutateladze TG, Côté J. HBO1, a MYSTerious KAT and its links to cancer. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195045. [PMID: 38851533 PMCID: PMC11330361 DOI: 10.1016/j.bbagrm.2024.195045] [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: 04/01/2024] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
The histone acetyltransferase HBO1, also known as KAT7, is a major chromatin modifying enzyme responsible for H3 and H4 acetylation. It is found within two distinct tetrameric complexes, the JADE subunit-containing complex and BRPF subunit-containing complex. The HBO1-JADE complex acetylates lysine 5, 8 and 12 of histone H4, and the HBO1-BRPF complex acetylates lysine 14 of histone H3. HBO1 regulates gene transcription, DNA replication, DNA damage repair, and centromere function. It is involved in diverse signaling pathways and plays crucial roles in development and stem cell biology. Recent work has established a strong relationship of HBO1 with the histone methyltransferase MLL/KMT2A in acute myeloid leukemia. Here, we discuss functional and pathological links of HBO1 to cancer, highlighting the underlying mechanisms that may pave the way to the development of novel anti-cancer therapies.
Collapse
Affiliation(s)
- Akihiko Yokoyama
- Tsuruoka Metabolomics Laboratory, National Cancer Center, Tsuruoka, Yamagata 997-0052, Japan.
| | - Hiroyuki Niida
- Department of Molecular Biology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Tatiana G Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, United States of America.
| | - Jacques Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division-CHU de Québec-UL Research Center, Laval University Cancer Research Center, Quebec City, QC G1R 3S3, Canada.
| |
Collapse
|
7
|
Yuan Q, Wu Y, Xue C, Zhao D, Wang H, Shen Y. KAT7 serves as an oncogenic gene and regulates CCL3 expression via STAT1 signaling in osteosarcoma. Biochem Biophys Res Commun 2024; 722:150156. [PMID: 38797155 DOI: 10.1016/j.bbrc.2024.150156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Osteosarcoma, considered as the primary cause of malignant bone tumors in children, necessitates novel therapeutic strategies to enhance overall survival rates. KAT7, a histone acetyltransferase, exerts pivotal functions in gene transcription and immune modulation. In light of this, our study identified a significant upregulation of KAT7 in the mRNA and protein levels in human osteosarcoma, boosting cell proliferation in vivo and in vitro. In addition, KAT7-mediated H3K14ac activation induced MMP14 transcription, leading to increased expression and facilitation of osteosarcoma cell metastasis. Subsequent bioinformatics analyses highlighted a correlation between KAT7 and adaptive immune responses, indicating CCL3 as a downstream target of KAT7. Mechanistically, STAT1 was found to transcriptionally upregulate CCL3 expression. Furthermore, overexpression of KAT7 suppressed CCL3 secretions, whereas knockdown of KAT7 enhanced its release. Overall, these findings underscore the oncogenic role of KAT7 in regulating immune responses for osteosarcoma treatment.
Collapse
Affiliation(s)
- Quan Yuan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu Province, People's Republic of China
| | - Yuxuan Wu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Cheng Xue
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Deyong Zhao
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Haibo Wang
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Yixin Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu Province, People's Republic of China.
| |
Collapse
|
8
|
Hong G, Chen W, Gong M, Wu Y, Shu G, Xiao Y, Zhang T, ShuXiong X. KAT7 suppresses tumorigenesis in clear cell renal cell carcinoma (ccRCC) by regulating cell cycle and ferroptosis sensitivity. Exp Cell Res 2024; 441:114149. [PMID: 38960363 DOI: 10.1016/j.yexcr.2024.114149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/23/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most aggressive malignancies in the urological system, known for its high immunogenicity. However, its pathogenesis remains unclear. This study utilized bioinformatics algorithms and in vitro experiments to investigate the role of KAT7 in ccRCC. The results indicate that KAT7 is significantly downregulated in ccRCC tissues and cell lines, which is linked to distant metastasis and unfavorable outcomes in ccRCC patients. Overexpression of KAT7 in vitro notably decreased the proliferation, migration, and invasion of renal cancer cells and inhibited Epithelial-Mesenchymal Transition (EMT). Additionally, Gene Set Enrichment Analysis (GSEA) demonstrated that KAT7-related gene functions are associated with cell cycle and ferroptosis transcription factors. Treatment with a KAT7 acetylation inhibitor in ccRCC cell lines reversed the S phase arrest caused by KAT7 overexpression. Similarly, ferroptosis inhibitors alleviated ferroptosis induced by overexpressed KAT7. In conclusion, the findings suggest that KAT7 acts as a tumor suppressor in ccRCC by modulating the cell cycle and ferroptosis sensitivity, underscoring its potential as a therapeutic target and prognostic biomarker for renal cell carcinoma patients.
Collapse
Affiliation(s)
- GuangYi Hong
- Guizhou University Medicine College, Guiyang, 550025, Guizhou Province, China
| | - Wei Chen
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
| | - MaoDi Gong
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
| | - YiKun Wu
- Guizhou University Medicine College, Guiyang, 550025, Guizhou Province, China
| | - GuoFeng Shu
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
| | - Yu Xiao
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
| | - Tao Zhang
- Guizhou University Medicine College, Guiyang, 550025, Guizhou Province, China
| | - Xu ShuXiong
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China.
| |
Collapse
|
9
|
Kamel H, Shete V, Gadamsetty S, Graves D, Bachus S, Akkerman N, Pelka P, Thimmapaya B. HBO1/KAT7/MYST2 HAT complex regulates human adenovirus replicative cycle. Heliyon 2024; 10:e28827. [PMID: 38601626 PMCID: PMC11004756 DOI: 10.1016/j.heliyon.2024.e28827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/12/2024] Open
Abstract
Human adenoviruses (HAdV) belong to a small DNA tumor virus family that continues as valuable models in understanding the viral strategies of usurping cell growth regulation. A number of HAdV type 2/5 early viral gene products interact with a variety of cellular proteins to build a conducive environment that promotes viral replication. Here we show that HBO1 (Histone Acetyltransferase Binding to ORC1), a member of the MYST histone acetyltransferase (HAT) complex (also known as KAT7 and MYST2) that acetylates most of the histone H3 lysine 14, is essential for HAdV5 growth. HBO1/MYST2/KAT7 HAT complexes are critical for a variety of cellular processes including control of cell proliferation. In HBO1 downregulated human cells, HAdV5 infection results in reduced expression of E1A and other viral early genes, virus growth is also reduced significantly. Importantly, HBO1 downregulation reduced H3 lysine 14 acetylation at viral promoters during productive infection, likely driving reduced viral gene expression. HBO1 was also associated with viral promoters during infection and co-localized with viral replication centers in the nuclei of infected cells. In transiently transfected cells, overexpression of E1A along with HBO1 stimulated histone acetyltransferase activity of HBO1. E1A also co-immunoprecipitated with HBO1 in transiently transfected cells. In summary, our results demonstrate that HAdV recruits the HBO1 HAT complex to aid in viral replication.
Collapse
Affiliation(s)
- Heba Kamel
- Microbiology and Immunology Department, Fienberg School of Medicine, Northwestern University, Chicago, USA
| | - Varsha Shete
- Microbiology and Immunology Department, Fienberg School of Medicine, Northwestern University, Chicago, USA
| | - Sayikrushna Gadamsetty
- Microbiology and Immunology Department, Fienberg School of Medicine, Northwestern University, Chicago, USA
| | - Drayson Graves
- Department of Microbiology, and Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Scott Bachus
- Department of Microbiology, and Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Nikolas Akkerman
- Department of Microbiology, and Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Peter Pelka
- Department of Microbiology, and Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Bayar Thimmapaya
- Microbiology and Immunology Department, Fienberg School of Medicine, Northwestern University, Chicago, USA
| |
Collapse
|
10
|
Yang Z, Mogre S, He R, Berdan EL, Ho Sui S, Hill S. The ORFIUS complex regulates ORC2 localization at replication origins. NAR Cancer 2024; 6:zcae003. [PMID: 38288445 PMCID: PMC10823580 DOI: 10.1093/narcan/zcae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 12/18/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024] Open
Abstract
High-grade serous ovarian cancer (HGSC) is a lethal malignancy with elevated replication stress (RS) levels and defective RS and RS-associated DNA damage responses. Here we demonstrate that the bromodomain-containing protein BRD1 is a RS suppressing protein that forms a replication origin regulatory complex with the histone acetyltransferase HBO1, the BRCA1 tumor suppressor, and BARD1, ORigin FIring Under Stress (ORFIUS). BRD1 and HBO1 promote eventual origin firing by supporting localization of the origin licensing protein ORC2 at origins. In the absence of BRD1 and/or HBO1, both origin firing and nuclei with ORC2 foci are reduced. BRCA1 regulates BRD1, HBO1, and ORC2 localization at replication origins. In the absence of BRCA1, both origin firing and nuclei with BRD1, HBO1, and ORC2 foci are increased. In normal and non-HGSC ovarian cancer cells, the ORFIUS complex responds to ATR and CDC7 origin regulatory signaling and disengages from origins during RS. In BRCA1-mutant and sporadic HGSC cells, BRD1, HBO1, and ORC2 remain associated with replication origins, and unresponsive to RS, DNA damage, or origin regulatory kinase inhibition. ORFIUS complex dysregulation may promote HGSC cell survival by allowing for upregulated origin firing and cell cycle progression despite accumulating DNA damage, and may be a RS target.
Collapse
Affiliation(s)
- Zelei Yang
- Department of Medical Oncology and Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Saie Mogre
- Department of Medical Oncology and Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ruiyang He
- Department of Medical Oncology and Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Emma L Berdan
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Shannan J Ho Sui
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Sarah J Hill
- Department of Medical Oncology and Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
11
|
Ahmad H, Chetlangia N, Prasanth SG. Chromatin's Influence on Pre-Replication Complex Assembly and Function. BIOLOGY 2024; 13:152. [PMID: 38534422 PMCID: PMC10968542 DOI: 10.3390/biology13030152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 03/28/2024]
Abstract
In all eukaryotes, the initiation of DNA replication requires a stepwise assembly of factors onto the origins of DNA replication. This is pioneered by the Origin Recognition Complex, which recruits Cdc6. Together, they bring Cdt1, which shepherds MCM2-7 to form the OCCM complex. Sequentially, a second Cdt1-bound hexamer of MCM2-7 is recruited by ORC-Cdc6 to form an MCM double hexamer, which forms a part of the pre-RC. Although the mechanism of ORC binding to DNA varies across eukaryotes, how ORC is recruited to replication origins in human cells remains an area of intense investigation. This review discusses how the chromatin environment influences pre-RC assembly, function, and, eventually, origin activity.
Collapse
Affiliation(s)
- Hina Ahmad
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, 601S Goodwin Avenue, Urbana, IL 61801, USA; (H.A.); (N.C.)
| | - Neha Chetlangia
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, 601S Goodwin Avenue, Urbana, IL 61801, USA; (H.A.); (N.C.)
| | - Supriya G. Prasanth
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, 601S Goodwin Avenue, Urbana, IL 61801, USA; (H.A.); (N.C.)
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
12
|
Gao Y, Sheng X, Tan D, Kim S, Choi S, Paudel S, Lee T, Yan C, Tan M, Kim KM, Cho SS, Ki SH, Huang H, Zhao Y, Lee S. Identification of Histone Lysine Acetoacetylation as a Dynamic Post-Translational Modification Regulated by HBO1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300032. [PMID: 37382194 PMCID: PMC10477889 DOI: 10.1002/advs.202300032] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/29/2023] [Indexed: 06/30/2023]
Abstract
Ketone bodies have long been known as a group of lipid-derived alternative energy sources during glucose shortages. Nevertheless, the molecular mechanisms underlying their non-metabolic functions remain largely elusive. This study identified acetoacetate as the precursor for lysine acetoacetylation (Kacac), a previously uncharacterized and evolutionarily conserved histone post-translational modification. This protein modification is comprehensively validated using chemical and biochemical approaches, including HPLC co-elution and MS/MS analysis using synthetic peptides, Western blot, and isotopic labeling. Histone Kacac can be dynamically regulated by acetoacetate concentration, possibly via acetoacetyl-CoA. Biochemical studies show that HBO1, traditionally known as an acetyltransferase, can also serve as an acetoacetyltransferase. In addition, 33 Kacac sites are identified on mammalian histones, depicting the landscape of histone Kacac marks across species and organs. In summary, this study thus discovers a physiologically relevant and enzymatically regulated histone mark that sheds light on the non-metabolic functions of ketone bodies.
Collapse
Affiliation(s)
- Yan Gao
- College of PharmacyKyungpook National UniversityDaegu41566Republic of Korea
| | - Xinlei Sheng
- Ben May Department for Cancer ResearchThe University of ChicagoChicagoIL60637USA
| | - Doudou Tan
- Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - SunJoo Kim
- College of PharmacyKyungpook National UniversityDaegu41566Republic of Korea
- Ben May Department for Cancer ResearchThe University of ChicagoChicagoIL60637USA
| | - Soyoung Choi
- College of PharmacyKyungpook National UniversityDaegu41566Republic of Korea
| | - Sanjita Paudel
- College of PharmacyKyungpook National UniversityDaegu41566Republic of Korea
| | - Taeho Lee
- College of PharmacyKyungpook National UniversityDaegu41566Republic of Korea
| | - Cong Yan
- Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Minjia Tan
- Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Kyu Min Kim
- Department of Biomedical Science, College of Natural ScienceChosun UniversityGwangju61452South Korea
| | - Sam Seok Cho
- College of PharmacyChosun UniversityGwangju61452South Korea
| | - Sung Hwan Ki
- College of PharmacyChosun UniversityGwangju61452South Korea
| | - He Huang
- Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yingming Zhao
- Ben May Department for Cancer ResearchThe University of ChicagoChicagoIL60637USA
| | - Sangkyu Lee
- School of PharmacySungkyunkwan UniversitySuwon16419South Korea
| |
Collapse
|
13
|
Hu Y, Stillman B. Origins of DNA replication in eukaryotes. Mol Cell 2023; 83:352-372. [PMID: 36640769 PMCID: PMC9898300 DOI: 10.1016/j.molcel.2022.12.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023]
Abstract
Errors occurring during DNA replication can result in inaccurate replication, incomplete replication, or re-replication, resulting in genome instability that can lead to diseases such as cancer or disorders such as autism. A great deal of progress has been made toward understanding the entire process of DNA replication in eukaryotes, including the mechanism of initiation and its control. This review focuses on the current understanding of how the origin recognition complex (ORC) contributes to determining the location of replication initiation in the multiple chromosomes within eukaryotic cells, as well as methods for mapping the location and temporal patterning of DNA replication. Origin specification and configuration vary substantially between eukaryotic species and in some cases co-evolved with gene-silencing mechanisms. We discuss the possibility that centromeres and origins of DNA replication were originally derived from a common element and later separated during evolution.
Collapse
Affiliation(s)
- Yixin Hu
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA; Program in Molecular and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Bruce Stillman
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
14
|
The CMG helicase and cancer: a tumor "engine" and weakness with missing mutations. Oncogene 2023; 42:473-490. [PMID: 36522488 PMCID: PMC9948756 DOI: 10.1038/s41388-022-02572-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
The replicative Cdc45-MCM-GINS (CMG) helicase is a large protein complex that functions in the DNA melting and unwinding steps as a component of replisomes during DNA replication in mammalian cells. Although the CMG performs this important role in cell growth, the CMG is not a simple bystander in cell cycle events. Components of the CMG, specifically the MCM precursors, are also involved in maintaining genomic stability by regulating DNA replication fork speeds, facilitating recovery from replicative stresses, and preventing consequential DNA damage. Given these important functions, MCM/CMG complexes are highly regulated by growth factors such as TGF-ß1 and by signaling factors such as Myc, Cyclin E, and the retinoblastoma protein. Mismanagement of MCM/CMG complexes when these signaling mediators are deregulated, and in the absence of the tumor suppressor protein p53, leads to increased genomic instability and is a contributor to tumorigenic transformation and tumor heterogeneity. The goal of this review is to provide insight into the mechanisms and dynamics by which the CMG is regulated during its assembly and activation in mammalian genomes, and how errors in CMG regulation due to oncogenic changes promote tumorigenesis. Finally, and most importantly, we highlight the emerging understanding of the CMG helicase as an exploitable vulnerability and novel target for therapeutic intervention in cancer.
Collapse
|
15
|
Stem cell plasticity, acetylation of H3K14, and de novo gene activation rely on KAT7. Cell Rep 2023; 42:111980. [PMID: 36641753 DOI: 10.1016/j.celrep.2022.111980] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 09/30/2022] [Accepted: 12/23/2022] [Indexed: 01/16/2023] Open
Abstract
In the conventional model of transcriptional activation, transcription factors bind to response elements and recruit co-factors, including histone acetyltransferases. Contrary to this model, we show that the histone acetyltransferase KAT7 (HBO1/MYST2) is required genome wide for histone H3 lysine 14 acetylation (H3K14ac). Examining neural stem cells, we find that KAT7 and H3K14ac are present not only at transcribed genes but also at inactive genes, intergenic regions, and in heterochromatin. KAT7 and H3K14ac were not required for the continued transcription of genes that were actively transcribed at the time of loss of KAT7 but indispensable for the activation of repressed genes. The absence of KAT7 abrogates neural stem cell plasticity, diverse differentiation pathways, and cerebral cortex development. Re-expression of KAT7 restored stem cell developmental potential. Overexpression of KAT7 enhanced neuron and oligodendrocyte differentiation. Our data suggest that KAT7 prepares chromatin for transcriptional activation and is a prerequisite for gene activation.
Collapse
|
16
|
Mi YY, Ji Y, Zhang L, Sun CY, Wei BB, Yang DJ, Wan HY, Qi XW, Wu S, Zhu LJ. A first-in-class HBO1 inhibitor WM-3835 inhibits castration-resistant prostate cancer cell growth in vitro and in vivo. Cell Death Dis 2023; 14:67. [PMID: 36709328 PMCID: PMC9884225 DOI: 10.1038/s41419-023-05606-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/30/2023]
Abstract
The prognosis and overall survival of castration-resistant prostate cancer (CRPC) patients are poor. The search for novel and efficient anti-CRPC agents is therefore extremely important. WM-3835 is a cell-permeable, potent and first-in-class HBO1 (KAT7 or MYST2) inhibitor. Here in primary human prostate cancer cells-derived from CRPC patients, WM-3835 potently inhibited cell viability, proliferation, cell cycle progression and in vitro cell migration. The HBO1 inhibitor provoked apoptosis in the prostate cancer cells. It failed to induce significant cytotoxicity and apoptosis in primary human prostate epithelial cells. shRNA-induced silencing of HBO1 resulted in robust anti-prostate cancer cell activity as well, and adding WM-3835 failed to induce further cytotoxicity in the primary prostate cancer cells. Conversely, ectopic overexpression of HBO1 further augmented primary prostate cancer cell proliferation and migration. WM-3835 inhibited H3-H4 acetylation and downregulated several pro-cancerous genes (CCR2, MYLK, VEGFR2, and OCIAD2) in primary CRPC cells. Importantly, HBO1 mRNA and protein levels are significantly elevated in CRPC tissues and cells. In vivo, daily intraperitoneal injection of WM-3835 potently inhibited pPC-1 xenograft growth in nude mice, and no apparent toxicities detected. Moreover, intratumoral injection of HBO1 shRNA adeno-associated virus (AAV) suppressed the growth of primary prostate cancer xenografts in nude mice. H3-H4 histone acetylation and HBO1-dependent genes (CCR2, MYLK, VEGFR2, and OCIAD2) were remarkably decreased in WM-3835-treated or HBO1-silenced xenograft tissues. Together, targeting HBO1 by WM-3835 robustly inhibits CRPC cell growth.
Collapse
Affiliation(s)
- Yuan-Yuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yu Ji
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Lifeng Zhang
- Department of Urology, Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Chuan-Yu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bing-Bing Wei
- Department of Urology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Dong-Jie Yang
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Hong-Yuan Wan
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiao-Wei Qi
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Sheng Wu
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Li-Jie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| |
Collapse
|
17
|
Employing non-targeted interactomics approach and subcellular fractionation to increase our understanding of the ghost proteome. iScience 2023; 26:105943. [PMID: 36866041 PMCID: PMC9971881 DOI: 10.1016/j.isci.2023.105943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/07/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Eukaryotic mRNA has long been considered monocistronic, but nowadays, alternative proteins (AltProts) challenge this tenet. The alternative or ghost proteome has largely been neglected and the involvement of AltProts in biological processes. Here, we used subcellular fractionation to increase the information about AltProts and facilitate the detection of protein-protein interactions by the identification of crosslinked peptides. In total, 112 unique AltProts were identified, and we were able to identify 220 crosslinks without peptide enrichment. Among these, 16 crosslinks between AltProts and Referenced Proteins (RefProts) were identified. We further focused on specific examples such as the interaction between IP_2292176 (AltFAM227B) and HLA-B, in which this protein could be a potential new immunopeptide, and the interactions between HIST1H4F and several AltProts which can play a role in mRNA transcription. Thanks to the study of the interactome and the localization of AltProts, we can reveal more of the importance of the ghost proteome.
Collapse
|
18
|
Role of PfMYST in DNA replication in Plasmodium falciparum. Exp Parasitol 2022; 242:108396. [DOI: 10.1016/j.exppara.2022.108396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/23/2022]
|
19
|
Fang Y, Zhao J, Guo X, Dai Y, Zhang H, Yin F, Zhang X, Sun C, Han Z, Wang H, Han Y. Establishment, immunological analysis, and drug prediction of a prognostic signature of ovarian cancer related to histone acetylation. Front Pharmacol 2022; 13:947252. [PMID: 36172179 PMCID: PMC9510621 DOI: 10.3389/fphar.2022.947252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, epigenetic modifications have been increasingly regarded as an important hallmark of cancer. Histone acetylation, as an important part of epigenetic modification, plays a key role in the progress, treatment, and prognosis of many cancers. In this study, based on the TCGA database, we performed LASSO regression and the Cox algorithm to establish a prognostic signature of ovarian cancer associated with histone acetylation modulator genes and verified it externally in the GEO database. Subsequently, we performed an immunological bioinformatics analysis of the model from multiple perspectives using the CIBERSORT algorithm, ESTIMATE algorithm, and TIDE algorithm to verify the accuracy of the model. Based on the prognostic model, we divided ovarian cancer patients into high-risk and low-risk groups, and assessed survival and the efficacy of accepting immunosuppressive therapy. In addition, based on the analysis of characteristics of the model, we also screened targeted drugs for high-risk patients and predicted potential drugs that inhibit platinum resistance through the connectivity map method. We ultimately constructed a histone acetylation modulator-related signature containing 10 histone acetylation modulators, among which HDAC1, HDAC10, and KAT7 can act as independent prognostic factors for ovarian cancer and are related to poor prognosis. In the analysis of the tumor microenvironment, the proportion of the B-infiltrating cells and the macrophages was significantly different between the high- and low-risk groups. Also, the samples with high-risk scores had higher tumor purity and lower immune scores. In terms of treatment, patients in the high-risk group who received immunotherapy had a higher likelihood of immune escape or rejection and were less likely to respond to platinum/paclitaxel therapy. Finally, we screened 20 potential drugs that could target the model for reference.
Collapse
Affiliation(s)
- Yujie Fang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, China
| | - Jing Zhao
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, China
| | - Xu Guo
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, China
| | - Yunfeng Dai
- Department of Radiotherapy, Yingkou Central Hospital, Yingkou, China
| | - Hao Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, China
| | - Fanxin Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, China
| | - Xiaoxu Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, China
| | - Chenxi Sun
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, China
| | - Zequan Han
- Department of Pathology, Yingkou Fangda Hospital, Yingkou, China
| | - Hecheng Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, China
- *Correspondence: Yanshuo Han, ;, Hecheng Wang,
| | - Yanshuo Han
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, China
- *Correspondence: Yanshuo Han, ;, Hecheng Wang,
| |
Collapse
|
20
|
Zu G, Liu Y, Cao J, Zhao B, Zhang H, You L. BRPF1-KAT6A/KAT6B Complex: Molecular Structure, Biological Function and Human Disease. Cancers (Basel) 2022; 14:4068. [PMID: 36077605 PMCID: PMC9454415 DOI: 10.3390/cancers14174068] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
The bromodomain and PHD finger-containing protein1 (BRPF1) is a member of family IV of the bromodomain-containing proteins that participate in the post-translational modification of histones. It functions in the form of a tetrameric complex with a monocytic leukemia zinc finger protein (MOZ or KAT6A), MOZ-related factor (MORF or KAT6B) or HAT bound to ORC1 (HBO1 or KAT7) and two small non-catalytic proteins, the inhibitor of growth 5 (ING5) or the paralog ING4 and MYST/Esa1-associated factor 6 (MEAF6). Mounting studies have demonstrated that all the four core subunits play crucial roles in different biological processes across diverse species, such as embryonic development, forebrain development, skeletal patterning and hematopoiesis. BRPF1, KAT6A and KAT6B mutations were identified as the cause of neurodevelopmental disorders, leukemia, medulloblastoma and other types of cancer, with germline mutations associated with neurodevelopmental disorders displaying intellectual disability, and somatic variants associated with leukemia, medulloblastoma and other cancers. In this paper, we depict the molecular structures and biological functions of the BRPF1-KAT6A/KAT6B complex, summarize the variants of the complex related to neurodevelopmental disorders and cancers and discuss future research directions and therapeutic potentials.
Collapse
Affiliation(s)
- Gaoyu Zu
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ying Liu
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jingli Cao
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Baicheng Zhao
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Hang Zhang
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Linya You
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Fudan University, Shanghai 200040, China
| |
Collapse
|
21
|
Cenik BK, Sze CC, Ryan CA, Das S, Cao K, Douillet D, Rendleman EJ, Zha D, Khan NH, Bartom E, Shilatifard A. A synthetic lethality screen reveals ING5 as a genetic dependency of catalytically dead Set1A/COMPASS in mouse embryonic stem cells. Proc Natl Acad Sci U S A 2022; 119:e2118385119. [PMID: 35500115 PMCID: PMC9171609 DOI: 10.1073/pnas.2118385119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/21/2022] [Indexed: 11/18/2022] Open
Abstract
Embryonic stem cells (ESCs) are defined by their ability to self-renew and the potential to differentiate into all tissues of the developing organism. We previously demonstrated that deleting the catalytic SET domain of the Set1A/complex of proteins associated with SET1 histone methyltransferase (Set1A/COMPASS) in mouse ESCs does not impair their viability or ability to self-renew; however, it leads to defects in differentiation. The precise mechanisms by which Set1A executes these functions remain to be elucidated. In this study, we demonstrate that mice lacking the SET domain of Set1A are embryonic lethal at a stage that is unique from null alleles. To gain insight into Set1A function in regulating pluripotency, we conducted a CRISPR/Cas9-mediated dropout screen and identified the MOZ/MORF (monocytic leukaemia zinc finger protein/monocytic leukaemia zinc finger protein-related factor) and HBO1 (HAT bound to ORC1) acetyltransferase complex member ING5 as a synthetic perturbation to Set1A. The loss of Ing5 in Set1AΔSET mouse ESCs decreases the fitness of these cells, and the simultaneous loss of ING5 and in Set1AΔSET leads to up-regulation of differentiation-associated genes. Taken together, our results point toward Set1A/COMPASS and ING5 as potential coregulators of the self-renewal and differentiation status of ESCs.
Collapse
Affiliation(s)
- Bercin K. Cenik
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Christie C. Sze
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Caila A. Ryan
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Siddhartha Das
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Kaixiang Cao
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Delphine Douillet
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Emily J. Rendleman
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Didi Zha
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Nabiha Haleema Khan
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Elizabeth Bartom
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Robert H. Lurie NCI Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Ali Shilatifard
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Robert H. Lurie NCI Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
22
|
Retusone A, a Guaiane-Type Sesquiterpene Dimer from Wikstroemia retusa and Its Inhibitory Effects on Histone Acetyltransferase HBO1 Expression. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092909. [PMID: 35566260 PMCID: PMC9105026 DOI: 10.3390/molecules27092909] [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] [Received: 03/30/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 12/24/2022]
Abstract
Retusone A (1), a new sesquiterpene dimer consisting of two guaiane-type sesquiterpenoids, and oleodaphnal (2) were isolated from heartwood of Wikstroemia retusa (Thymelaeaceae). The planar structure of 1 was elucidated on the basis of HRESIMS and NMR spectroscopic data, and the relative stereochemistry was established by X-ray diffraction analysis. The absolute configuration of 1 was determined by electronic circular dichroism. Compound 1 suppressed luciferase reporter gene expression driven by the HBO1 (histone acetyltransferase binding to ORC1) gene promoter in human breast cancer MCF7 cells. Compound 1 also decreased the expression of endogenous HBO1 mRNA and protein, and inhibited proliferation of the cells. These results suggest that retusone A (1), which has a unique dimeric sesquiterpenoid structure with inhibitory activity against HBO1 expression, may contribute to the development of a novel therapeutic candidate for the treatment of breast cancer.
Collapse
|
23
|
Fang Z, Sun X, Wang X, Ma J, Palaia T, Rana U, Miao B, Ragolia L, Hu W, Miao QR. NOGOB receptor deficiency increases cerebrovascular permeability and hemorrhage via impairing histone acetylation-mediated CCM1/2 expression. J Clin Invest 2022; 132:e151382. [PMID: 35316220 PMCID: PMC9057619 DOI: 10.1172/jci151382] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 03/16/2022] [Indexed: 11/17/2022] Open
Abstract
The loss function of cerebral cavernous malformation (CCM) genes leads to most CCM lesions characterized by enlarged leaking vascular lesions in the brain. Although we previously showed that NOGOB receptor (NGBR) knockout in endothelial cells (ECs) results in cerebrovascular lesions in the mouse embryo, the molecular mechanism by which NGBR regulates CCM1/2 expression has not been elucidated. Here, we show that genetic depletion of Ngbr in ECs at both postnatal and adult stages results in CCM1/2 expression deficiency and cerebrovascular lesions such as enlarged vessels, blood-brain-barrier hyperpermeability, and cerebral hemorrhage. To reveal the molecular mechanism, we used RNA-sequencing analysis to examine changes in the transcriptome. Surprisingly, we found that the acetyltransferase HBO1 and histone acetylation were downregulated in NGBR-deficient ECs. The mechanistic studies elucidated that NGBR is required for maintaining the expression of CCM1/2 in ECs via HBO1-mediated histone acetylation. ChIP-qPCR data further demonstrated that loss of NGBR impairs the binding of HBO1 and acetylated histone H4K5 and H4K12 on the promotor of the CCM1 and CCM2 genes. Our findings on epigenetic regulation of CCM1 and CCM2 that is modulated by NGBR and HBO1-mediated histone H4 acetylation provide a perspective on the pathogenesis of sporadic CCMs.
Collapse
Affiliation(s)
- Zhi Fang
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, USA
- Department of Surgery and Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Xiaoran Sun
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, USA
| | - Xiang Wang
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, USA
- Department of Surgery and Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ji Ma
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, USA
| | - Thomas Palaia
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, USA
| | - Ujala Rana
- Department of Surgery and Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Benjamin Miao
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, USA
| | - Louis Ragolia
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, USA
| | - Wenquan Hu
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, USA
- Department of Surgery and Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Qing Robert Miao
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, USA
- Department of Surgery and Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
24
|
Abstract
DNA replication in eukaryotic cells initiates from large numbers of sites called replication origins. Initiation of replication from these origins must be tightly controlled to ensure the entire genome is precisely duplicated in each cell cycle. This is accomplished through the regulation of the first two steps in replication: loading and activation of the replicative DNA helicase. Here we describe what is known about the mechanism and regulation of these two reactions from a genetic, biochemical, and structural perspective, focusing on recent progress using proteins from budding yeast. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Alessandro Costa
- Macromolecular Machines Laboratory, The Francis Crick Institute, London, UK;
| | - John F X Diffley
- Chromosome Replication Laboratory, The Francis Crick Institute, London, UK;
| |
Collapse
|
25
|
Yang Y, Kueh AJ, Grant ZL, Abeysekera W, Garnham AL, Wilcox S, Hyland CD, Di Rago L, Metcalf D, Alexander WS, Coultas L, Smyth GK, Voss AK, Thomas T. The histone lysine acetyltransferase HBO1 (KAT7) regulates hematopoietic stem cell quiescence and self-renewal. Blood 2022; 139:845-858. [PMID: 34724565 DOI: 10.1182/blood.2021013954] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/13/2021] [Indexed: 11/20/2022] Open
Abstract
The histone acetyltransferase HBO1 (MYST2, KAT7) is indispensable for postgastrulation development, histone H3 lysine 14 acetylation (H3K14Ac), and the expression of embryonic patterning genes. In this study, we report the role of HBO1 in regulating hematopoietic stem cell function in adult hematopoiesis. We used 2 complementary cre-recombinase transgenes to conditionally delete Hbo1 (Mx1-Cre and Rosa26-CreERT2). Hbo1-null mice became moribund due to hematopoietic failure with pancytopenia in the blood and bone marrow 2 to 6 weeks after Hbo1 deletion. Hbo1-deleted bone marrow cells failed to repopulate hemoablated recipients in competitive transplantation experiments. Hbo1 deletion caused a rapid loss of hematopoietic progenitors. The numbers of lineage-restricted progenitors for the erythroid, myeloid, B-, and T-cell lineages were reduced. Loss of HBO1 resulted in an abnormally high rate of recruitment of quiescent hematopoietic stem cells (HSCs) into the cell cycle. Cycling HSCs produced progenitors at the expense of self-renewal, which led to the exhaustion of the HSC pool. Mechanistically, genes important for HSC functions were downregulated in HSC-enriched cell populations after Hbo1 deletion, including genes essential for HSC quiescence and self-renewal, such as Mpl, Tek(Tie-2), Gfi1b, Egr1, Tal1(Scl), Gata2, Erg, Pbx1, Meis1, and Hox9, as well as genes important for multipotent progenitor cells and lineage-specific progenitor cells, such as Gata1. HBO1 was required for H3K14Ac through the genome and particularly at gene loci required for HSC quiescence and self-renewal. Our data indicate that HBO1 promotes the expression of a transcription factor network essential for HSC maintenance and self-renewal in adult hematopoiesis.
Collapse
Affiliation(s)
- Yuqing Yang
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| | - Andrew J Kueh
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| | - Zoe L Grant
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| | - Waruni Abeysekera
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| | - Alexandra L Garnham
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| | - Stephen Wilcox
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
| | - Craig D Hyland
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
| | - Ladina Di Rago
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
| | - Don Metcalf
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| | - Warren S Alexander
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| | - Leigh Coultas
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| | - Gordon K Smyth
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, Australia
| | - Anne K Voss
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| | - Tim Thomas
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; and
- Department of Medical Biology and
| |
Collapse
|
26
|
Jaiswal B, Agarwal A, Gupta A. Lysine Acetyltransferases and Their Role in AR Signaling and Prostate Cancer. Front Endocrinol (Lausanne) 2022; 13:886594. [PMID: 36060957 PMCID: PMC9428678 DOI: 10.3389/fendo.2022.886594] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/17/2022] [Indexed: 11/18/2022] Open
Abstract
The development and growth of a normal prostate gland, as well as its physiological functions, are regulated by the actions of androgens through androgen receptor (AR) signaling which drives multiple cellular processes including transcription, cellular proliferation, and apoptosis in prostate cells. Post-translational regulation of AR plays a vital role in directing its cellular activities via modulating its stability, nuclear localization, and transcriptional activity. Among various post-translational modifications (PTMs), acetylation is an essential PTM recognized in AR and is governed by the regulated actions of acetyltransferases and deacetyltransferases. Acetylation of AR has been identified as a critical step for its activation and depending on the site of acetylation, the intracellular dynamics and activity of the AR can be modulated. Various acetyltransferases such as CBP, p300, PCAF, TIP60, and ARD1 that are known to acetylate AR, may directly coactivate the AR transcriptional function or help to recruit additional coactivators to functionally regulate the transcriptional activity of the AR. Aberrant expression of acetyltransferases and their deregulated activities have been found to interfere with AR signaling and play a key role in development and progression of prostatic diseases, including prostate cancer (PCa). In this review, we summarized recent research advances aimed at understanding the role of various lysine acetyltransferases (KATs) in the regulation of AR activity at the level of post-translational modifications in normal prostate physiology, as well as in development and progression of PCa. Considering the critical importance of KATs in modulating AR activity in physiological and patho-physiological context, we further discussed the potential of targeting these enzymes as a therapeutic option to treat AR-related pathology in combination with hormonal therapy.
Collapse
Affiliation(s)
- Bharti Jaiswal
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, India
- *Correspondence: Ashish Gupta, ; Bharti Jaiswal,
| | - Akanksha Agarwal
- Epigenetics and Human Disease Laboratory, Centre of Excellence in Epigenetics (CoEE) Department of Life Sciences, Shiv Nadar University, Delhi, UP, India
| | - Ashish Gupta
- Epigenetics and Human Disease Laboratory, Centre of Excellence in Epigenetics (CoEE) Department of Life Sciences, Shiv Nadar University, Delhi, UP, India
- *Correspondence: Ashish Gupta, ; Bharti Jaiswal,
| |
Collapse
|
27
|
Grant ZL, Hickey PF, Abeysekera W, Whitehead L, Lewis SM, Symons RCA, Baldwin TM, Amann-Zalcenstein D, Garnham AL, Smyth GK, Thomas T, Voss AK, Coultas L. The histone acetyltransferase HBO1 promotes efficient tip cell sprouting during angiogenesis. Development 2021; 148:272249. [PMID: 34550360 DOI: 10.1242/dev.199581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 09/15/2021] [Indexed: 12/14/2022]
Abstract
Blood vessel growth and remodelling are essential during embryonic development and disease pathogenesis. The diversity of endothelial cells (ECs) is transcriptionally evident and ECs undergo dynamic changes in gene expression during vessel growth and remodelling. Here, we investigated the role of the histone acetyltransferase HBO1 (KAT7), which is important for activating genes during development and for histone H3 lysine 14 acetylation (H3K14ac). Loss of HBO1 and H3K14ac impaired developmental sprouting angiogenesis and reduced pathological EC overgrowth in the retinal endothelium. Single-cell RNA sequencing of retinal ECs revealed an increased abundance of tip cells in Hbo1-deficient retinas, which led to EC overcrowding in the retinal sprouting front and prevented efficient tip cell migration. We found that H3K14ac was highly abundant in the endothelial genome in both intra- and intergenic regions, suggesting that HBO1 acts as a genome organiser that promotes efficient tip cell behaviour necessary for sprouting angiogenesis. This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Zoe L Grant
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Peter F Hickey
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Waruni Abeysekera
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Sabrina M Lewis
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Robert C A Symons
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Australia.,Department of Surgery, University of Melbourne, Parkville, 3010, Australia.,Department of Ophthalmology, Royal Melbourne Hospital, Parkville, 3050, Australia
| | - Tracey M Baldwin
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
| | - Daniela Amann-Zalcenstein
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Alexandra L Garnham
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Tim Thomas
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Anne K Voss
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Leigh Coultas
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
28
|
Xiao Y, Li W, Yang H, Pan L, Zhang L, Lu L, Chen J, Wei W, Ye J, Li J, Li G, Zhang Y, Tan M, Ding J, Wong J. HBO1 is a versatile histone acyltransferase critical for promoter histone acylations. Nucleic Acids Res 2021; 49:8037-8059. [PMID: 34259319 PMCID: PMC8661427 DOI: 10.1093/nar/gkab607] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022] Open
Abstract
Recent studies demonstrate that histones are subjected to a series of short-chain fatty acid modifications that is known as histone acylations. However, the enzymes responsible for histone acylations in vivo are not well characterized. Here, we report that HBO1 is a versatile histone acyltransferase that catalyzes not only histone acetylation but also propionylation, butyrylation and crotonylation both in vivo and in vitro and does so in a JADE or BRPF family scaffold protein-dependent manner. We show that the minimal HBO1/BRPF2 complex can accommodate acetyl-CoA, propionyl-CoA, butyryl-CoA and crotonyl-CoA. Comparison of CBP and HBO1 reveals that they catalyze histone acylations at overlapping as well as distinct sites, with HBO1 being the key enzyme for H3K14 acylations. Genome-wide chromatin immunoprecipitation assay demonstrates that HBO1 is highly enriched at and contributes to bulk histone acylations on the transcriptional start sites of active transcribed genes. HBO1 promoter intensity highly correlates with the level of promoter histone acylation, but has no significant correlation with level of transcription. We also show that HBO1 is associated with a subset of DNA replication origins. Collectively our study establishes HBO1 as a versatile histone acyltransferase that links histone acylations to promoter acylations and selection of DNA replication origins.
Collapse
Affiliation(s)
- Yanhui Xiao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.,Joint Center for Translational Medicine, School of Life Sciences, East China Normal University and Fengxian District Central Hospital, Shanghai 201499, China
| | - Wenjing Li
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Yang
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Science and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai 200092, China
| | - Lulu Pan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Liwei Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Lu Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiwei Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Wei Wei
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jie Ye
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiwen Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Guohong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yong Zhang
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Science and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Tongji University, Shanghai 200092, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianping Ding
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.,Joint Center for Translational Medicine, School of Life Sciences, East China Normal University and Fengxian District Central Hospital, Shanghai 201499, China
| |
Collapse
|
29
|
Gao YY, Ling ZY, Zhu YR, Shi C, Wang Y, Zhang XY, Zhang ZQ, Jiang Q, Chen MB, Yang S, Cao C. The histone acetyltransferase HBO1 functions as a novel oncogenic gene in osteosarcoma. Am J Cancer Res 2021; 11:4599-4615. [PMID: 33754016 PMCID: PMC7978299 DOI: 10.7150/thno.55655] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/06/2021] [Indexed: 02/06/2023] Open
Abstract
HBO1 (KAT7 or MYST2) is a histone acetyltransferase that acetylates H3 and H4 histones. Methods: HBO1 expression was tested in human OS tissues and cells. Genetic strategies, including shRNA, CRISPR/Cas9 and overexpression constructs, were applied to exogenously alter HBO1 expression in OS cells. The HBO1 inhibitor WM-3835 was utilized to block HBO1 activation. Results:HBO1 mRNA and protein expression is significantly elevated in OS tissues and cells. In established (MG63/U2OS lines) and primary human OS cells, shRNA-mediated HBO1 silencing and CRISPR/Cas9-induced HBO1 knockout were able to potently inhibit cell viability, growth, proliferation, as well as cell migration and invasion. Significant increase of apoptosis was detected in HBO1-silenced/knockout OS cells. Conversely, ectopic HBO1 overexpression promoted OS cell proliferation and migration. We identified ZNF384 (zinc finger protein 384) as a potential transcription factor of HBO1. Increased binding between ZNF384 and HBO1 promoter was detected in OS cell and tissues, whereas ZNF384 silencing via shRNA downregulated HBO1 and produced significant anti-OS cell activity. In vivo, intratumoral injection of HBO1 shRNA lentivirus silenced HBO1 and inhibited OS xenograft growth in mice. Furthermore, growth of HBO1-knockout OS xenografts was significantly slower than the control xenografts. WM-3835, a novel and high-specific small molecule HBO1 inhibitor, was able to potently suppressed OS cell proliferation and migration, and led to apoptosis activation. Furthermore, intraperitoneal injection of a single dose of WM-3835 potently inhibited OS xenograft growth in SCID mice. Conclusion: HBO1 overexpression promotes OS cell growth in vitro and in vivo.
Collapse
|
30
|
Wu J, Li L, Jiang G, Zhan H, Zhu X, Yang W. NCAPG2 facilitates glioblastoma cells' malignancy and xenograft tumor growth via HBO1 activation by phosphorylation. Cell Tissue Res 2021; 383:693-706. [PMID: 32897418 DOI: 10.1007/s00441-020-03281-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 08/12/2020] [Indexed: 10/23/2022]
Abstract
NCAPG2 (non-SMC condensin II complex subunit G2), as an important factor in cell mitosis, has been the focus in the study of different cancers. However, the role of NCAPG2 in the malignancy of glioblastoma cells remains unknown. The findings from the present study demonstrated that NCAPG2 was significantly increased in human glioblastoma tissues and was associated with poor clinical outcome. Moreover, NCAPG2 could promote proliferation, migration, and invasion and regulate cell cycle in glioblastoma cells. Investigation of the molecular mechanism indicated that NCAPG2 regulated HBO1 phosphorylation and H4 histone acetylase activation, modulated the activation of Wnt/β-catenin pathway, and the binding of MCM protein to chromatin to exert its role. Furthermore, knockdown of HBO1 was found to reverse the effect of NCAPG2 overexpression on cell proliferation, migration, invasion, and cell cycle. In addition, knockdown of NCAPG2 attenuated glioblastoma tumorigenesis in vivo. Taken together, the findings demonstrated that NCAPG2 facilitates the malignancy of glioblastoma cells and xenograft tumor growth via HBO1 activation by phosphorylation. These results improve our understanding of the mechanism underlying glioblastoma progression and may contribute to the identification of novel biomarkers and therapeutic targets for glioblastoma.
Collapse
Affiliation(s)
- Jianheng Wu
- Department of Neurosurgery, the People's Hospital of GaoZhou, Gaozhou, Maoming, 525200, Guangdong Province, China
| | - Linfan Li
- Department of Neurosurgery, the People's Hospital of GaoZhou, Gaozhou, Maoming, 525200, Guangdong Province, China
| | - Guangyuan Jiang
- Department of Neurosurgery, Nanxishan Hospital, Guilin, 541000, Guangxi Zhuang Autonomous Region, China
| | - Hui Zhan
- Department of Neurosurgery, the People's Hospital of GaoZhou, Gaozhou, Maoming, 525200, Guangdong Province, China
| | - Xiumei Zhu
- Department of Pathology, the People's Hospital of GaoZhou, Gaozhou, Maoming, 525200, Guangdong, China
| | - Wujun Yang
- Department of Neurosurgery, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture(Enshi Clinical College of Wuhan University), Enshi Tujia and Miao Autonomous Prefecture, 445000, Hubei, China.
| |
Collapse
|
31
|
Liang Y, Su Y, Xu C, Zhang N, Liu D, Li G, Tong T, Chen J. Protein kinase D1 phosphorylation of KAT7 enhances its protein stability and promotes replication licensing and cell proliferation. Cell Death Discov 2020; 6:89. [PMID: 33014433 PMCID: PMC7501302 DOI: 10.1038/s41420-020-00323-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/09/2020] [Accepted: 09/02/2020] [Indexed: 01/24/2023] Open
Abstract
The histone acetyltransferase (HAT) KAT7/HBO1/MYST2 plays a crucial role in the pre-replication complex (pre-RC) formation, DNA replication and cell proliferation via acetylation of histone H4 and H3. In a search for protein kinase D1 (PKD1)-interacting proteins, we have identified KAT7 as a potential PKD1 substrate. We show that PKD1 directly interacts and phosphorylates KAT7 at Thr97 and Thr331 in vitro and in vivo. PKD1-mediated phosphorylation of KAT7 enhances its expression levels and stability by reducing its ubiquitination-mediated degradation. Significantly, the phospho-defective mutant KAT7-Thr97/331A attenuates histone H4 acetylation levels, MCM2/6 loading on the chromatin, DNA replication and cell proliferation. Similarly, PKD1 knockdown decreases, whereas the constitutive active mutant PKD1-CA increases histone H4 acetylation levels and MCM2/6 loading on the chromatin. Overall, these results suggest that PKD1-mediated phosphorylation of KAT7 may be required for pre-RC formation and DNA replication.
Collapse
Affiliation(s)
- Yao Liang
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191 China
| | - Yuanyuan Su
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191 China
| | - Chenzhong Xu
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191 China
| | - Na Zhang
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191 China
| | - Doudou Liu
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191 China
| | - Guodong Li
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191 China
| | - Tanjun Tong
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191 China
| | - Jun Chen
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191 China
| |
Collapse
|
32
|
You D, Richardson JR, Aleksunes LM. Epigenetic Regulation of Multidrug Resistance Protein 1 and Breast Cancer Resistance Protein Transporters by Histone Deacetylase Inhibition. Drug Metab Dispos 2020; 48:459-480. [PMID: 32193359 PMCID: PMC7250367 DOI: 10.1124/dmd.119.089953] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
Multidrug resistance protein 1 (MDR1, ABCB1, P-glycoprotein) and breast cancer resistance protein (BCRP, ABCG2) are key efflux transporters that mediate the extrusion of drugs and toxicants in cancer cells and healthy tissues, including the liver, kidneys, and the brain. Altering the expression and activity of MDR1 and BCRP influences the disposition, pharmacodynamics, and toxicity of chemicals, including a number of commonly prescribed medications. Histone acetylation is an epigenetic modification that can regulate gene expression by changing the accessibility of the genome to transcriptional regulators and transcriptional machinery. Recently, studies have suggested that pharmacological inhibition of histone deacetylases (HDACs) modulates the expression and function of MDR1 and BCRP transporters as a result of enhanced histone acetylation. This review addresses the ability of HDAC inhibitors to modulate the expression and the function of MDR1 and BCRP transporters and explores the molecular mechanisms by which HDAC inhibition regulates these transporters. While the majority of studies have focused on histone regulation of MDR1 and BCRP in drug-resistant and drug-sensitive cancer cells, emerging data point to similar responses in nonmalignant cells and tissues. Elucidating epigenetic mechanisms regulating MDR1 and BCRP is important to expand our understanding of the basic biology of these two key transporters and subsequent consequences on chemoresistance as well as tissue exposure and responses to drugs and toxicants. SIGNIFICANCE STATEMENT: Histone deacetylase inhibitors alter the expression of key efflux transporters multidrug resistance protein 1 and breast cancer resistance protein in healthy and malignant cells.
Collapse
Affiliation(s)
- Dahea You
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey (D.Y.); Department of Environmental Health Sciences, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, Florida (J.R.R.); Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey (J.R.R., L.M.A.); and Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Ernest Mario School of Pharmacy, Piscataway, New Jersey (L.M.A.)
| | - Jason R Richardson
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey (D.Y.); Department of Environmental Health Sciences, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, Florida (J.R.R.); Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey (J.R.R., L.M.A.); and Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Ernest Mario School of Pharmacy, Piscataway, New Jersey (L.M.A.)
| | - Lauren M Aleksunes
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey (D.Y.); Department of Environmental Health Sciences, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, Florida (J.R.R.); Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey (J.R.R., L.M.A.); and Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Ernest Mario School of Pharmacy, Piscataway, New Jersey (L.M.A.)
| |
Collapse
|
33
|
Lan R, Wang Q. Deciphering structure, function and mechanism of lysine acetyltransferase HBO1 in protein acetylation, transcription regulation, DNA replication and its oncogenic properties in cancer. Cell Mol Life Sci 2020; 77:637-649. [PMID: 31535175 PMCID: PMC11104888 DOI: 10.1007/s00018-019-03296-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/19/2022]
Abstract
HBO1 complexes are major acetyltransferase responsible for histone H4 acetylation in vivo, which belongs to the MYST family. As the core catalytic subunit, HBO1 consists of an N-terminal domain and a C-terminal MYST domain that are in charge of acetyl-CoA binding and acetylation reaction. HBO1 complexes are multimeric and normally consist of two native subunits MEAF6, ING4 or ING5 and two kinds of cofactors as chromatin reader: Jade-1/2/3 and BRPF1/2/3. The choices of subunits to form the HBO1 complexes provide a regulatory switch to potentiate its activity between histone H4 and H3 tails. Thus, HBO1 complexes present multiple functions in histone acetylation, gene transcription, DNA replication, protein ubiquitination, and immune regulation, etc. HBO1 is a co-activator for CDT1 to facilitate chromatin loading of MCM complexes and promotes DNA replication licensing. This process is regulated by mitotic kinases such as CDK1 and PLK1 by phosphorylating HBO1 and modulating its acetyltransferase activity, therefore, connecting histone acetylation to regulations of cell cycle and DNA replication. In addition, both gene amplification and protein overexpression of HBO1 confirmed its oncogenic role in cancers. In this paper, we review the recent advances and discuss our understanding of the multiple functions, activity regulation, and disease relationship of HBO1.
Collapse
Affiliation(s)
- Rongfeng Lan
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| | - Qianqian Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| |
Collapse
|
34
|
Kueh AJ, Eccles S, Tang L, Garnham AL, May RE, Herold MJ, Smyth GK, Voss AK, Thomas T. HBO1 (KAT7) Does Not Have an Essential Role in Cell Proliferation, DNA Replication, or Histone 4 Acetylation in Human Cells. Mol Cell Biol 2020; 40:e00506-19. [PMID: 31767635 PMCID: PMC6996278 DOI: 10.1128/mcb.00506-19] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/11/2019] [Indexed: 01/01/2023] Open
Abstract
HBO1 (MYST2/KAT7) is essential for histone 3 lysine 14 acetylation (H3K14ac) but is dispensable for H4 acetylation and DNA replication in mouse tissues. In contrast, previous studies using small interfering RNA (siRNA) knockdown in human cell lines have suggested that HBO1 is essential for DNA replication. To determine if HBO1 has distinctly different roles in immortalized human cell lines and normal mouse cells, we performed siRNA knockdown of HBO1. In addition, we used CRISPR/Cas9 to generate 293T, MCF7, and HeLa cell lines lacking HBO1. Using both techniques, we show that HBO1 is essential for all H3K14ac in human cells and is unlikely to have a direct effect on H4 acetylation and only has minor effects on cell proliferation. Surprisingly, the loss of HBO1 and H3K14ac in HeLa cells led to the secondary loss of almost all H4 acetylation after 4 weeks. Thus, HBO1 is dispensable for DNA replication and cell proliferation in immortalized human cells. However, while cell proliferation proceeded without HBO1 and H3K14ac, HBO1 gene deletion led to profound changes in cell adhesion, particularly in 293T cells. Consistent with this phenotype, the loss of HBO1 in both 293T and HeLa principally affected genes mediating cell adhesion, with comparatively minor effects on other cellular processes.
Collapse
Affiliation(s)
- Andrew J Kueh
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Samantha Eccles
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Leonie Tang
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Alexandra L Garnham
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Rose E May
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Marco J Herold
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Gordon K Smyth
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Mathematics and Statistics, University of Melbourne, Parkville, Victoria, Australia
| | - Anne K Voss
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Tim Thomas
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
35
|
Chen L, Luo L, Kang N, He X, Li T, Chen Y. The Protective Effect of HBO1 on Cigarette Smoke Extract-Induced Apoptosis in Airway Epithelial Cells. Int J Chron Obstruct Pulmon Dis 2020; 15:15-24. [PMID: 32021140 PMCID: PMC6954103 DOI: 10.2147/copd.s234634] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose Epigenetic modification is one of most important mechanisms underlying the pathogenesis of chronic obstructive pulmonary disease (COPD). The purpose of this study was to determine whether histone acetyltransferase binding to ORC1 (HBO1) can protect against cigarette smoke (CS)-induced cell apoptosis and sustain normal histone acetylation in COPD. Methods Human lung tissue samples were obtained from patients who underwent lung resection. The emphysema mouse model and HBO1 overexpressing mice were each established by intraperitoneal injection with cigarette smoke extract (CSE) or intratracheal lentiviral vectors instillation. TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) assays were used to assess apoptotic ratio in mice. The apoptosis of human bronchial epithelial cells (HBECs) was assayed by flow cytometry. HBO1, B-cell lymphoma-2 (BCL-2), and H3K14ac protein expression were detected by Western blotting. HBO1 mRNA expression was measured by quantitative real-time polymerase chain reaction. Results Protein expression of HBO1 was decreased significantly in lung tissue from COPD patients and CSE-treated emphysema mouse models. Overexpression of HBO1 attenuated CSE-induced emphysematous changes, as well as apoptosis in the lungs of COPD mice. In vitro, the HBO1 protein degraded in a time- and dose-dependent course with CSE treatment. With flow cytometry, we proved that HBO1 could reverse the apoptosis of HBECs induced by CSE. Furthermore, HBO1 overexpression promoted the expression of anti-apoptotic BCL-2 protein and enhanced H3K14 acetylation in airway epithelial cells. Conclusion These findings demonstrate that the key histone modulator HBO1 plays a protective role in COPD pathogenesis that may shed light on potential therapeutic targets to inhibit the progress of COPD.
Collapse
Affiliation(s)
- Lin Chen
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China
| | - Lijuan Luo
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China
| | - Naixin Kang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China
| | - Xue He
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China
| | - Tiao Li
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China
| | - Yan Chen
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, People's Republic of China.,Research Unit of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China.,Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha 410011, Hunan, People's Republic of China
| |
Collapse
|
36
|
Wang Y, Chen S, Tian W, Zhang Q, Jiang C, Qian L, Liu Y. High-Expression HBO1 Predicts Poor Prognosis in Gastric Cancer. Am J Clin Pathol 2019; 152:517-526. [PMID: 31247063 DOI: 10.1093/ajcp/aqz065] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Our goal was to assess the expression of histone acetyltransferase binding to origin recognition complex 1 (HBO1) in gastric cancer and the effect on prognosis for the patients. METHODS We used quantitative reverse transcription polymerase chain reaction, Western blot, and tissue microarray immunohistochemistry to investigate the expressions of HBO1 messenger RNA (mRNA) and protein in gastric cancer tissues. Online resources, including Oncomine and Kaplan-Meier Plotter, were used to further assess the correlation between HBO1 expression and the prognosis of the patients with gastric cancer. RESULTS HBO1 mRNA and protein expressions in gastric cancer tissues were both significantly higher than those in normal tissues. The correlations between high HBO1 expression and differentiation, invasive depth (T), lymph node metastasis (N), distant metastasis (M), TNM staging, and serum carcinoembryonic antigen levels were positive. High HBO1 expression was negatively correlated with survival time in patients with gastric cancer. CONCLUSIONS HBO1 might be a valuable biomarker to evaluate the prognosis of patients with gastric cancer.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Sufang Chen
- Department of Medical Imaging and Laboratory, Xiangnan University, Chenzhou, China
| | - Wei Tian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qing Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Chunyi Jiang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Li Qian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Ying Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
37
|
Heinz KS, Rapp A, Casas-Delucchi CS, Lehmkuhl A, Romero-Fernández I, Sánchez A, Krämer OH, Marchal JA, Cardoso MC. DNA replication dynamics of vole genome and its epigenetic regulation. Epigenetics Chromatin 2019; 12:18. [PMID: 30871586 PMCID: PMC6416958 DOI: 10.1186/s13072-019-0262-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 03/07/2019] [Indexed: 01/19/2023] Open
Abstract
Background The genome of some vole rodents exhibit large blocks of heterochromatin coupled to their sex chromosomes. The DNA composition and transcriptional activity of these heterochromatin blocks have been studied, but little is known about their DNA replication dynamics and epigenetic composition. Results Here, we show prominent epigenetic marks of the heterochromatic blocks in the giant sex chromosomes of female Microtus cabrerae cells. While the X chromosomes are hypoacetylated and cytosine hypomethylated, they are either enriched for macroH2A and H3K27me3 typical for facultative heterochromatin or for H3K9me3 and HP1 beta typical for constitutive heterochromatin. Using pulse-chase replication labeling and time-lapse microscopy, we found that the heterochromatic block enriched for macroH2A/H3K27me3 of the X chromosome is replicated during mid-S-phase, prior to the heterochromatic block enriched for H3K9me3/HP1 beta, which is replicated during late S-phase. To test whether histone acetylation level regulates its replication dynamics, we induced either global hyperacetylation by pharmacological inhibition or by targeting a histone acetyltransferase to the heterochromatic region of the X chromosomes. Our data reveal that histone acetylation level affects DNA replication dynamics of the sex chromosomes’ heterochromatin and leads to a global reduction in replication fork rate genome wide. Conclusions In conclusion, we mapped major epigenetic modifications controlling the structure of the sex chromosome-associated heterochromatin and demonstrated the occurrence of differences in the molecular mechanisms controlling the replication timing of the heterochromatic blocks at the sex chromosomes in female Microtus cabrerae cells. Furthermore, we highlighted a conserved role of histone acetylation level on replication dynamics across mammalian species. Electronic supplementary material The online version of this article (10.1186/s13072-019-0262-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kathrin S Heinz
- Cell Biology and Epigenetics, Department of Biology, Technische Universität Darmstadt, Schnittspahnstrasse 10, 64287, Darmstadt, Germany
| | - Alexander Rapp
- Cell Biology and Epigenetics, Department of Biology, Technische Universität Darmstadt, Schnittspahnstrasse 10, 64287, Darmstadt, Germany
| | - Corella S Casas-Delucchi
- Cell Biology and Epigenetics, Department of Biology, Technische Universität Darmstadt, Schnittspahnstrasse 10, 64287, Darmstadt, Germany.,Chromosome Replication Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Anne Lehmkuhl
- Cell Biology and Epigenetics, Department of Biology, Technische Universität Darmstadt, Schnittspahnstrasse 10, 64287, Darmstadt, Germany
| | | | - Antonio Sánchez
- Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Oliver H Krämer
- Institute of Toxicology, Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Mainz, Germany
| | | | - M Cristina Cardoso
- Cell Biology and Epigenetics, Department of Biology, Technische Universität Darmstadt, Schnittspahnstrasse 10, 64287, Darmstadt, Germany.
| |
Collapse
|
38
|
Simon RP, Rumpf T, Linkuviene V, Matulis D, Akhtar A, Jung M. Cofactor Analogues as Active Site Probes in Lysine Acetyltransferases. J Med Chem 2019; 62:2582-2597. [PMID: 30785747 DOI: 10.1021/acs.jmedchem.8b01887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lysine acetyltransferases (KATs, also termed histone acetyltransferases, HATs) catalyze the acetylation of substrate lysine residues by employing the cofactor acetyl-coenzyme A (AcCoA), thereby providing a dynamic control mechanism of protein function. Because of their major involvement in cell development and homeostasis, small-molecule modulators of KAT activity are urgently needed to assess their therapeutic potential and for probing their underlying biology. Recent advances in the field suggest that targeting the cofactor binding site represents a promising strategy for identifying potent and selective ligands. Here, we present the synthesis of two functional cofactor-based chemical probes and their usage as mechanistic tools in a broadly applicable assay platform. A fluorescence polarization (FP)-based binding assay was combined with biolayer interferometry competition analysis and a FP competition activity immunoassay to enable easy, reliable, and profound evaluation of ligands that target the KAT cofactor binding site.
Collapse
Affiliation(s)
- Roman P Simon
- Institute of Pharmaceutical Sciences , University of Freiburg , Albertstraße 25 , 79104 Freiburg im Breisgau , Germany
| | - Tobias Rumpf
- Department of Chromatin Regulation , Max-Planck-Institute of Immunobiology and Epigenetics , Stuebeweg 51 , 79108 Freiburg , Germany
| | - Vaida Linkuviene
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center , Vilnius University , Saulėtekio 7 , 10257 Vilnius , Lithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center , Vilnius University , Saulėtekio 7 , 10257 Vilnius , Lithuania
| | - Asifa Akhtar
- Department of Chromatin Regulation , Max-Planck-Institute of Immunobiology and Epigenetics , Stuebeweg 51 , 79108 Freiburg , Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences , University of Freiburg , Albertstraße 25 , 79104 Freiburg im Breisgau , Germany
| |
Collapse
|
39
|
Voss AK, Thomas T. Histone Lysine and Genomic Targets of Histone Acetyltransferases in Mammals. Bioessays 2018; 40:e1800078. [PMID: 30144132 DOI: 10.1002/bies.201800078] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/01/2018] [Indexed: 01/08/2023]
Abstract
Histone acetylation has been recognized as an important post-translational modification of core nucleosomal histones that changes access to the chromatin to allow gene transcription, DNA replication, and repair. Histone acetyltransferases were initially identified as co-activators that link DNA-binding transcription factors to the general transcriptional machinery. Over the years, more chromatin-binding modes have been discovered suggesting direct interaction of histone acetyltransferases and their protein complex partners with histone proteins. While much progress has been made in characterizing histone acetyltransferase complexes biochemically, cell-free activity assay results are often at odds with in-cell histone acetyltransferase activities. In-cell studies suggest specific histone lysine targets, but broad recruitment modes, apparently not relying on specific DNA sequences, but on chromatin of a specific functional state. Here we review the evidence for general versus specific roles of individual nuclear lysine acetyltransferases in light of in vivo and in vitro data in the mammalian system.
Collapse
Affiliation(s)
- Anne K Voss
- Walter and Eliza Hall Institute of Medical Research, 3 1G Royal Parade, Parkville, Melbourne, Victoria, 3052, Australia
| | - Tim Thomas
- Department of Medical Biology, The University of Melbourne, 1G Royal Parade, Parkville, Melbourne, Victoria, 3052, Australia
| |
Collapse
|
40
|
Long C, Lai Y, Li J, Huang J, Zou C. LPS promotes HBO1 stability via USP25 to modulate inflammatory gene transcription in THP-1 cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:773-782. [PMID: 30745998 DOI: 10.1016/j.bbagrm.2018.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The histone acetyltransferase HBO1 (Histone acetyltransferase binding to origin recognition complex 1, Myst2/Kat7) participates in a range of life processes including DNA replication and tumorigenesis. Recent studies revealed that HBO1 is involved in gene transcriptional activation. However, the molecular behavior of HBO1 in inflammation is yet to be studied. Here we report that endotoxin lipopolysaccharide (LPS) elevates HBO1 protein level via up-regulating UPS25 (ubiquitin specific peptidase 25) and alters inflammatory gene transcription in THP-1 monocytes and in human primary macrophages. LPS protects HBO1 from ubiquitin proteasomal degradation without significantly altering its transcription. By immunoprecipitation, we identified that HBO1 associates with a deubiquitinating enzyme USP25 in THP-1 cells. LPS increases protein level of USP25 resulting in accumulation of HBO1 by suppression of HBO1 ubiquitination. Stabilized-HBO1 modulates inflammatory gene transcription in THP-1 cells. These findings indicate that USP25 promotes stability of HBO1 in bacterial infection thereby enhances HBO1-mediated inflammatory gene transcription.
Collapse
Affiliation(s)
- Chen Long
- Department of Minimally Invasive Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China 410011.,Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Yandong Lai
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Jin Li
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| | - Jiangsheng Huang
- Department of Minimally Invasive Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China 410011
| | - Chunbin Zou
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15213
| |
Collapse
|
41
|
Lysine acetyltransferase inhibitors: structure-activity relationships and potential therapeutic implications. Future Med Chem 2018; 10:1067-1091. [PMID: 29676588 DOI: 10.4155/fmc-2017-0244] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Lysine acetylation is a post-translational modification of both histone and nonhistone proteins that is catalyzed by lysine acetyltransferases and plays a key role in numerous biological contexts. The dysregulation of this enzyme activity is implicated in many human pathologies such as cancer, neurological and inflammatory disorders. Many lysine acetyltransferase inhibitors (KATi) have been developed so far, but there is still the need for new, more potent, metabolically stable and selective KATi as chemical tools for studying KAT biology and/or as potential therapeutic agents. This review will examine the features of KAT enzymes and related diseases, with particular emphasis on KATi (bisubstrate analogs, natural compounds and synthetic derivatives), analyzing their mechanism of action, structure-activity relationships, pharmacokinetic/pharmacodynamic properties and potential future applications.
Collapse
|
42
|
Di Martile M, Del Bufalo D, Trisciuoglio D. The multifaceted role of lysine acetylation in cancer: prognostic biomarker and therapeutic target. Oncotarget 2018; 7:55789-55810. [PMID: 27322556 PMCID: PMC5342454 DOI: 10.18632/oncotarget.10048] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/01/2016] [Indexed: 12/28/2022] Open
Abstract
Lysine acetylation is a post-translational modification that regulates gene transcription by targeting histones as well as a variety of transcription factors in the nucleus. Recently, several reports have demonstrated that numerous cytosolic proteins are also acetylated and that this modification, affecting protein activity, localization and stability has profound consequences on their cellular functions. Interestingly, most non-histone proteins targeted by acetylation are relevant for tumorigenesis. In this review, we will analyze the functional implications of lysine acetylation in different cellular compartments, and will examine our current understanding of lysine acetyltransferases family, highlighting the biological role and prognostic value of these enzymes and their substrates in cancer. The latter part of the article will address challenges and current status of molecules targeting lysine acetyltransferase enzymes in cancer therapy.
Collapse
Affiliation(s)
- Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, Research, Advanced Diagnostics and Technological Innovation Department, Regina Elena National Cancer Institute, Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, Research, Advanced Diagnostics and Technological Innovation Department, Regina Elena National Cancer Institute, Rome, Italy
| | - Daniela Trisciuoglio
- Preclinical Models and New Therapeutic Agents Unit, Research, Advanced Diagnostics and Technological Innovation Department, Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
43
|
Guo Z, Zhang Z, Wang Q, Zhang J, Wang L, Zhang Q, Li H, Wu S. Manganese chloride induces histone acetylation changes in neuronal cells: Its role in manganese-induced damage. Neurotoxicology 2017; 65:255-263. [PMID: 29155171 DOI: 10.1016/j.neuro.2017.11.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/03/2017] [Accepted: 11/13/2017] [Indexed: 12/19/2022]
Abstract
Manganese neurotoxicity presents with Parkinson-like symptoms, with degeneration of dopaminergic neurons in the basal ganglia as the principal pathological feature. Manganese neurotoxicity studies may contribute to a better understanding of the mechanism of Parkinson's disease. Here, we examined the effects of manganese on histone acetylation, a major epigenetic change in chromatin that can regulate gene expression, chromatin remodelling, cell cycle progression, DNA repair and apoptosis. In this study, we found that manganese chloride (MnCl2) may significantly suppress the acetylation of histone H3 and H4 in PC12 cells and SHSY5Y cells in a time-dependent manner. Then we tested the role of manganese chloride on histone acetyltransferase (HAT) and histone deacetylase (HDAC). The results showed that MnCl2 increased the activity of HDAC but decreased that of HAT in PC12 cells. Further experiments showed that MnCl2 selectively increased the expression levels of HDAC3 and HDAC4 rather than HDAC1 and HDAC2, but decreased that of HAT in PC12 cells and SHSY5Y cells. Pretreatment with the HAT inhibitor anacardic acid (AA) enhanced manganese-induced decrease in cell viability and apoptosis, but HDAC inhibition by TSA drug had an opposite effect in PC12 cells. Collectively, MnCl2 inhibited the acetylation of core histones in cell culture models of PD, and that inhibition of HDAC activity by TSA protects against manganese-induced cell death, indicating that histone acetylation may represent key epigenetic changes in manganese-induced dopaminergic neurotoxicity.
Collapse
Affiliation(s)
- Zhenkun Guo
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Zhipeng Zhang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Qingqing Wang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Jie Zhang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Lijin Wang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Qunwei Zhang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Environmental and Occupational Health Sciences, University of Louisville, Louisville, KY 40202, USA
| | - Huangyuan Li
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| | - Siying Wu
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
44
|
Zhang Z, Guo Z, Zhan Y, Li H, Wu S. Role of histone acetylation in activation of nuclear factor erythroid 2-related factor 2/heme oxygenase 1 pathway by manganese chloride. Toxicol Appl Pharmacol 2017; 336:94-100. [PMID: 29054681 DOI: 10.1016/j.taap.2017.10.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/10/2017] [Accepted: 10/16/2017] [Indexed: 01/03/2023]
Abstract
Manganese neurotoxicity is characterized by Parkinson-like symptoms with degeneration of dopaminergic neurons in the basal ganglia as the principal pathological feature. Manganese neurotoxicity studies may contribute to a good understanding of the mechanism of Parkinson's disease (PD). In this study, we first confirmed that MnCl2 can promote the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) protein in the nucleus or cytoplasm while increasing the binding activity of Nrf2 and antioxidant response elements, further promoting the expression of downstream target gene heme oxygenase 1 (HO-1) and leading to increase levels of reactive oxygen species (ROS) and reduce the levels of reduced glutathione (GSH). Second, we investigated the role of histone acetylation in the activation of Nrf2/HO-1 pathway by manganese chloride in rat adrenal pheochromocytoma (PC12) cells. Histone acetyltransferase inhibitor (anacardic acid) and histone deacetylase inhibitor (trichostatin A, TSA) were used as pretreatment reagents to adjust the level of histone acetylation. Here, we show that downregulation of histone acetylation can inhibit Mn-induced Nrf2 nuclear translocation and further inhibits the Mn-activated Nrf2/HO-1 pathway. This downregulation also promotes manganese-induced increase of ROS and decrease of GSH in neurons. These results suggest that the downregulation of histone acetylation may play an important role in the neurotoxicity caused by manganese and that TSA may provide new ideas and targets in treating manganese-induced Parkinson's syndrome and PD.
Collapse
Affiliation(s)
- Zhipeng Zhang
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Center for Adverse Drug Reaction Monitoring, Fujian Provincial Food and Drug Administration, Fuzhou 350001, China
| | - Zhenkun Guo
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yanting Zhan
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Deapartment of Mannagement, Fujian Health College, Fuzhou 350101, China
| | - Huangyuan Li
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| | - Siying Wu
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
45
|
Tao Y, Zhong C, Zhu J, Xu S, Ding J. Structural and mechanistic insights into regulation of HBO1 histone acetyltransferase activity by BRPF2. Nucleic Acids Res 2017; 45:5707-5719. [PMID: 28334966 PMCID: PMC5449618 DOI: 10.1093/nar/gkx142] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 02/21/2017] [Indexed: 12/15/2022] Open
Abstract
HBO1, a member of the MYST family of histone acetyltransferases (HATs), is required for global acetylation of histone H3K14 and embryonic development. It functions as a catalytic subunit in multisubunit complexes comprising a BRPF1/2/3 or JADE1/2/3 scaffold protein, and two accessory proteins. BRPF2 has been shown to be important for the HAT activity of HBO1 toward H3K14. Here we demonstrated that BRPF2 can regulate the HAT activity of HBO1 toward free H3 and H4, and nucleosomal H3. Particularly, a short N-terminal region of BRPF2 is sufficient for binding to HBO1 and can potentiate its activity toward H3K14. The crystal structure of the HBO1 MYST domain in complex with this segment of BRPF2 together with the biochemical and cell biological data revealed the key residues responsible for the HBO1–BRPF2 interaction. Our structural and functional data together indicate that the N-terminal region of BRPF2 plays an important role in the binding of HBO1 and a minor role in the binding of nucleosomes, which provide new mechanistic insights into the regulation of the HAT activity of HBO1 by BRPF2.
Collapse
Affiliation(s)
- Ye Tao
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Chen Zhong
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Junjun Zhu
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Shutong Xu
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jianping Ding
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, and University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China.,Shanghai Science Research Center, Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China.,Collaborative Innovation Center for Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| |
Collapse
|
46
|
Pardo M, Yu L, Shen S, Tate P, Bode D, Letney BL, Quelle DE, Skarnes W, Choudhary JS. Myst2/Kat7 histone acetyltransferase interaction proteomics reveals tumour-suppressor Niam as a novel binding partner in embryonic stem cells. Sci Rep 2017; 7:8157. [PMID: 28811661 PMCID: PMC5557939 DOI: 10.1038/s41598-017-08456-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/10/2017] [Indexed: 12/28/2022] Open
Abstract
MYST histone acetyltransferases have crucial functions in transcription, replication and DNA repair and are hence implicated in development and cancer. Here we characterise Myst2/Kat7/Hbo1 protein interactions in mouse embryonic stem cells by affinity purification coupled to mass spectrometry. This study confirms that in embryonic stem cells Myst2 is part of H3 and H4 histone acetylation complexes similar to those described in somatic cells. We identify a novel Myst2-associated protein, the tumour suppressor protein Niam (Nuclear Interactor of ARF and Mdm2). Human NIAM is involved in chromosome segregation, p53 regulation and cell proliferation in somatic cells, but its role in embryonic stem cells is unknown. We describe the first Niam embryonic stem cell interactome, which includes proteins with roles in DNA replication and repair, transcription, splicing and ribosome biogenesis. Many of Myst2 and Niam binding partners are required for correct embryonic development, implicating Myst2 and Niam in the cooperative regulation of this process and suggesting a novel role for Niam in embryonic biology. The data provides a useful resource for exploring Myst2 and Niam essential cellular functions and should contribute to deeper understanding of organism early development and survival as well as cancer. Data are available via ProteomeXchange with identifier PXD005987.
Collapse
Affiliation(s)
- Mercedes Pardo
- Proteomic Mass Spectrometry, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom.
| | - Lu Yu
- Proteomic Mass Spectrometry, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Shihpei Shen
- Stem Cell Engineering, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
- Cold Genesys Inc., Santa Ana, CA, USA
| | - Peri Tate
- Stem Cell Engineering, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Daniel Bode
- Proteomic Mass Spectrometry, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
- Wellcome Trust PhD Program, Cambridge Stem Cell Institute, Cambridge, Cambridgeshire, United Kingdom
| | - Blake L Letney
- Departments of Pharmacology and Pathology, The University of Iowa and Holden Comprehensive Cancer Center, Iowa City, IA, 52242, USA
| | - Dawn E Quelle
- Departments of Pharmacology and Pathology, The University of Iowa and Holden Comprehensive Cancer Center, Iowa City, IA, 52242, USA
| | - William Skarnes
- Stem Cell Engineering, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Jyoti S Choudhary
- Proteomic Mass Spectrometry, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| |
Collapse
|
47
|
Phosphorylated HBO1 at UV irradiated sites is essential for nucleotide excision repair. Nat Commun 2017; 8:16102. [PMID: 28719581 PMCID: PMC5520108 DOI: 10.1038/ncomms16102] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
HBO1, a histone acetyl transferase, is a co-activator of DNA pre-replication complex formation. We recently reported that HBO1 is phosphorylated by ATM and/or ATR and binds to DDB2 after ultraviolet irradiation. Here, we show that phosphorylated HBO1 at cyclobutane pyrimidine dimer (CPD) sites mediates histone acetylation to facilitate recruitment of XPC at the damaged DNA sites. Furthermore, HBO1 facilitates accumulation of SNF2H and ACF1, an ATP-dependent chromatin remodelling complex, to CPD sites. Depletion of HBO1 inhibited repair of CPDs and sensitized cells to ultraviolet irradiation. However, depletion of HBO1 in cells derived from xeroderma pigmentosum patient complementation groups, XPE, XPC and XPA, did not lead to additional sensitivity towards ultraviolet irradiation. Our findings suggest that HBO1 acts in concert with SNF2H-ACF1 to make the chromosome structure more accessible to canonical nucleotide excision repair factors.
Collapse
|
48
|
Abstract
In this review, Prioleau and MacAlpine summarize recent advances in our understanding of how primary sequence, chromatin environment, and nuclear architecture contribute to the dynamic selection and activation of replication origins across diverse cell types and developmental stages. For more than three decades, investigators have sought to identify the precise locations where DNA replication initiates in mammalian genomes. The development of molecular and biochemical approaches to identify start sites of DNA replication (origins) based on the presence of defining and characteristic replication intermediates at specific loci led to the identification of only a handful of mammalian replication origins. The limited number of identified origins prevented a comprehensive and exhaustive search for conserved genomic features that were capable of specifying origins of DNA replication. More recently, the adaptation of origin-mapping assays to genome-wide approaches has led to the identification of tens of thousands of replication origins throughout mammalian genomes, providing an unprecedented opportunity to identify both genetic and epigenetic features that define and regulate their distribution and utilization. Here we summarize recent advances in our understanding of how primary sequence, chromatin environment, and nuclear architecture contribute to the dynamic selection and activation of replication origins across diverse cell types and developmental stages.
Collapse
Affiliation(s)
- Marie-Noëlle Prioleau
- Institut Jacques Monod, UMR7592, Centre National de la Recherche Scientifique, Universite Paris Diderot, Equipe Labellisee Association pour la Recherche sur le Cancer, Paris 75013, France
| | - David M MacAlpine
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710. USA
| |
Collapse
|
49
|
Sugimoto N, Fujita M. Molecular Mechanism for Chromatin Regulation During MCM Loading in Mammalian Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1042:61-78. [PMID: 29357053 DOI: 10.1007/978-981-10-6955-0_3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
DNA replication is a fundamental process required for the accurate and timely duplication of chromosomes. During late mitosis to G1 phase, the MCM2-7 complex is loaded onto chromatin in a manner dependent on ORC, CDC6, and Cdt1, and chromatin becomes licensed for replication. Although every eukaryotic organism shares common features in replication control, there are also some differences among species. For example, in higher eukaryotic cells including human cells, no strict sequence specificity has been observed for replication origins, unlike budding yeast or bacterial replication origins. Therefore, elements other than beyond DNA sequences are important for regulating replication. For example, the stability and precise positioning of nucleosomes affects replication control. However, little is known about how nucleosome structure is regulated when replication licensing occurs. During the last decade, histone acetylation enzyme HBO1, chromatin remodeler SNF2H, and histone chaperone GRWD1 have been identified as chromatin-handling factors involved in the promotion of replication licensing. In this review, we discuss how the rearrangement of nucleosome formation by these factors affects replication licensing.
Collapse
Affiliation(s)
- Nozomi Sugimoto
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| | - Masatoshi Fujita
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
50
|
Pozhitkov AE, Neme R, Domazet-Lošo T, Leroux BG, Soni S, Tautz D, Noble PA. Tracing the dynamics of gene transcripts after organismal death. Open Biol 2017; 7:160267. [PMID: 28123054 PMCID: PMC5303275 DOI: 10.1098/rsob.160267] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/12/2016] [Indexed: 12/13/2022] Open
Abstract
In life, genetic and epigenetic networks precisely coordinate the expression of genes-but in death, it is not known if gene expression diminishes gradually or abruptly stops or if specific genes and pathways are involved. We studied this by identifying mRNA transcripts that apparently increase in relative abundance after death, assessing their functions, and comparing their abundance profiles through postmortem time in two species, mouse and zebrafish. We found mRNA transcript profiles of 1063 genes became significantly more abundant after death of healthy adult animals in a time series spanning up to 96 h postmortem. Ordination plots revealed non-random patterns in the profiles by time. While most of these transcript levels increased within 0.5 h postmortem, some increased only at 24 and 48 h postmortem. Functional characterization of the most abundant transcripts revealed the following categories: stress, immunity, inflammation, apoptosis, transport, development, epigenetic regulation and cancer. The data suggest a step-wise shutdown occurs in organismal death that is manifested by the apparent increase of certain transcripts with various abundance maxima and durations.
Collapse
Affiliation(s)
- Alex E Pozhitkov
- Department of Oral Health Sciences, University of Washington, PO Box 357444, Seattle, WA 98195, USA
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, 24306 Ploen, Germany
| | - Rafik Neme
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, 24306 Ploen, Germany
| | - Tomislav Domazet-Lošo
- Laboratory of Evolutionary Genetics, Division of Molecular Biology, Ruđer Bošković Institute, 10002 Zagreb, Croatia
- Catholic University of Croatia, Ilica 242, Zagreb, Croatia
| | - Brian G Leroux
- Department of Oral Health Sciences, University of Washington, PO Box 357444, Seattle, WA 98195, USA
| | - Shivani Soni
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101-0271, USA
| | - Diethard Tautz
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, 24306 Ploen, Germany
| | - Peter A Noble
- Department of Periodontics, University of Washington, PO Box 357444, Seattle, WA 98195, USA
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101-0271, USA
- PhD Program in Microbiology, Alabama State University, Montgomery, AL 36101-0271, USA
| |
Collapse
|