1
|
Yu X, Wu H, Su J, Liu X, Liang K, Li Q, Yu R, Shao X, Wang H, Wang YL, Shyh-Chang N. Acetyl-CoA metabolism maintains histone acetylation for syncytialization of human placental trophoblast stem cells. Cell Stem Cell 2024; 31:1280-1297.e7. [PMID: 39084220 DOI: 10.1016/j.stem.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/15/2023] [Accepted: 07/08/2024] [Indexed: 08/02/2024]
Abstract
During pregnancy, placental-fetal nutrient allocation is crucial for fetal and maternal health. However, the regulatory mechanisms for nutrient metabolism and allocation in placental trophoblasts have remained unclear. Here, we used human first-trimester placenta samples and human trophoblast stem cells (hTSCs) to discover that glucose metabolism is highly active in hTSCs and cytotrophoblasts, but during syncytialization, it decreases to basal levels, remaining necessary for fueling acetyl-CoA and differentiation potential. Acetate supplementation could rescue syncytiotrophoblast fusion from glycolysis deficiency by replenishing acetyl-CoA and maintaining histone acetylation, thus rescuing the activation of syncytialization genes. Even brief glycolysis deficiency could permanently inhibit differentiation potential and promote inflammation, which could also be permanently rescued by brief acetate supplementation in vivo. These results suggest that hTSCs retain only basal glycolytic acetyl-CoA metabolism during syncytialization to regulate cell fates via nutrient-responsive histone acetylation, with implications for our understanding of the balance between placental and fetal nutrition.
Collapse
Affiliation(s)
- Xin Yu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Hao Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jiali Su
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xupeng Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Kun Liang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Qianqian Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Ruoxuan Yu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xuan Shao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Hongmei Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China.
| | - Yan-Ling Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China.
| | - Ng Shyh-Chang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
2
|
KAT8 acetylation-controlled lipolysis affects the invasive and migratory potential of colorectal cancer cells. Cell Death Dis 2023; 14:164. [PMID: 36849520 PMCID: PMC9970984 DOI: 10.1038/s41419-023-05582-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 12/18/2022] [Accepted: 01/11/2023] [Indexed: 03/01/2023]
Abstract
Epigenetic mechanisms involved in gene expression play an essential role in various cellular processes, including lipid metabolism. Lysine acetyltransferase 8 (KAT8), a histone acetyltransferase, has been reported to mediate de novo lipogenesis by acetylating fatty acid synthase. However, the effect of KAT8 on lipolysis is unclear. Here, we report a novel mechanism of KAT8 on lipolysis involving in its acetylation by general control non-repressed protein 5 (GCN5) and its deacetylation by Sirtuin 6 (SIRT6). KAT8 acetylation at K168/175 residues attenuates the binding activity of KAT8 and inhibits the recruitment of RNA pol II to the promoter region of the lipolysis-related genes adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL), subsequently down-regulating lipolysis to affect the invasive and migratory potential of colorectal cancer cells. Our findings uncover a novel mechanism that KAT8 acetylation-controlled lipolysis affects invasive and migratory potential in colorectal cancer cells.
Collapse
|
3
|
Nie Q, Huan X, Kang J, Yin J, Zhao J, Li Y, Zhang Z. MG149 Inhibits MOF-Mediated p53 Acetylation to Attenuate X-Ray Radiation-Induced Apoptosis in H9c2 Cells. Radiat Res 2022; 198:590-598. [PMID: 36481803 DOI: 10.1667/rade-22-00049.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 09/30/2022] [Indexed: 12/13/2022]
Abstract
Cardiomyocyte apoptosis is involved in the pathogenesis of radiation-induced heart disease, but the underlying epigenetic mechanism remains elusive. We evaluated the potential mediating role of males absent on the first (MOF) in the association between epigenetic activation of p53 lysine 120 (p53K120) and X-ray radiation-induced apoptosis in H9c2 cells. H9c2 cells were pretreated for 24 h with the MOF inhibitor MG149 after 4 Gy irradiation, followed by assessment of cell proliferation, injury, and apoptosis. MOF expression was upregulated by X-ray radiation. MG149 suppressed the proliferation inhibition, reduction of mitochondrial membrane potential, ROS production, and cell apoptosis. MG149 may promote the survival of H9c2 cells via inhibition of MOF-mediated p53K120 acetylation in response to X-ray radiation-induced apoptosis. Our data indicates a MOF-associated epigenetic mechanism in H9c2 cells that promotes attenuation of X-ray radiation-induced injury.
Collapse
Affiliation(s)
- Qianwen Nie
- Lanzhou University Second College of Clinical Medicine, Chengguan District, Lanzhou 730030, China.,Department of General Medicine, Second Hospital of Lanzhou University, No.82 Cui Ying Men, Cheng Guan District, Lanzhou 730030, China
| | - Xuan Huan
- Lanzhou University Second College of Clinical Medicine, Chengguan District, Lanzhou 730030, China.,Department of General Medicine, Second Hospital of Lanzhou University, No.82 Cui Ying Men, Cheng Guan District, Lanzhou 730030, China
| | - Jing Kang
- Lanzhou University Second College of Clinical Medicine, Chengguan District, Lanzhou 730030, China.,Department of General Medicine, Second Hospital of Lanzhou University, No.82 Cui Ying Men, Cheng Guan District, Lanzhou 730030, China
| | - Jiangyan Yin
- Lanzhou University Second College of Clinical Medicine, Chengguan District, Lanzhou 730030, China.,Department of General Medicine, Second Hospital of Lanzhou University, No.82 Cui Ying Men, Cheng Guan District, Lanzhou 730030, China
| | - Jiahui Zhao
- Lanzhou University Second College of Clinical Medicine, Chengguan District, Lanzhou 730030, China
| | - Yi Li
- School of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - ZhengYi Zhang
- Lanzhou University Second College of Clinical Medicine, Chengguan District, Lanzhou 730030, China.,Department of General Medicine, Second Hospital of Lanzhou University, No.82 Cui Ying Men, Cheng Guan District, Lanzhou 730030, China
| |
Collapse
|
4
|
Chakraborty S, Singh M, Pandita RK, Singh V, Lo CS, Leonard F, Horikoshi N, Moros EG, Guha D, Hunt CR, Chau E, Ahmed KM, Sethi P, Charaka V, Godin B, Makhijani K, Scherthan H, Deck J, Hausmann M, Mushtaq A, Altaf M, Ramos KS, Bhat KM, Taneja N, Das C, Pandita TK. Heat-induced SIRT1-mediated H4K16ac deacetylation impairs resection and SMARCAD1 recruitment to double strand breaks. iScience 2022; 25:104142. [PMID: 35434547 PMCID: PMC9010620 DOI: 10.1016/j.isci.2022.104142] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/16/2022] [Accepted: 03/21/2022] [Indexed: 12/17/2022] Open
Abstract
Hyperthermia inhibits DNA double-strand break (DSB) repair that utilizes homologous recombination (HR) pathway by a poorly defined mechanism(s); however, the mechanisms for this inhibition remain unclear. Here we report that hyperthermia decreases H4K16 acetylation (H4K16ac), an epigenetic modification essential for genome stability and transcription. Heat-induced reduction in H4K16ac was detected in humans, Drosophila, and yeast, indicating that this is a highly conserved response. The examination of histone deacetylase recruitment to chromatin after heat-shock identified SIRT1 as the major deacetylase subsequently enriched at gene-rich regions. Heat-induced SIRT1 recruitment was antagonized by chromatin remodeler SMARCAD1 depletion and, like hyperthermia, the depletion of the SMARCAD1 or combination of the two impaired DNA end resection and increased replication stress. Altered repair protein recruitment was associated with heat-shock-induced γ-H2AX chromatin changes and DSB repair processing. These results support a novel mechanism whereby hyperthermia impacts chromatin organization owing to H4K16ac deacetylation, negatively affecting the HR-dependent DSB repair.
Collapse
Affiliation(s)
- Sharmistha Chakraborty
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Mayank Singh
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Raj K. Pandita
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
| | - Vipin Singh
- Biophysics & Structural Genomics Division Saha Institute of Nuclear Physics, Bidhan Nagar, Kolkata, West Bengal 700064, India
- Homi Bhaba National Institute, Mumbai, India
| | - Calvin S.C. Lo
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 Rotterdam, CA, the Netherlands
| | - Fransisca Leonard
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Nobuo Horikoshi
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
| | - Eduardo G. Moros
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
- Departments of Radiation Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Deblina Guha
- Biophysics & Structural Genomics Division Saha Institute of Nuclear Physics, Bidhan Nagar, Kolkata, West Bengal 700064, India
| | - Clayton R. Hunt
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
| | - Eric Chau
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Kazi M. Ahmed
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Prayas Sethi
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Vijaya Charaka
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Biana Godin
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Kalpana Makhijani
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Harry Scherthan
- Bundeswehr Institute of Radiobiology Affiliated to the University of Ulm, Neuherbergstr. 11, 80937 Munich, Germany
| | - Jeanette Deck
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| | - Michael Hausmann
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| | - Arjamand Mushtaq
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Mohammad Altaf
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Kenneth S. Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, USA
| | - Krishna M. Bhat
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Nitika Taneja
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 Rotterdam, CA, the Netherlands
| | - Chandrima Das
- Biophysics & Structural Genomics Division Saha Institute of Nuclear Physics, Bidhan Nagar, Kolkata, West Bengal 700064, India
- Homi Bhaba National Institute, Mumbai, India
| | - Tej K. Pandita
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Radiation Oncology, Washington University, St Louis, MO, USA
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, USA
| |
Collapse
|
5
|
Effect of ACY-1215 on cytoskeletal remodeling and histone acetylation in bovine somatic cell nuclear transfer embryos. Theriogenology 2022; 183:98-107. [DOI: 10.1016/j.theriogenology.2022.02.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/10/2022] [Accepted: 02/18/2022] [Indexed: 11/23/2022]
|
6
|
Pandita TK, Hunt CR, Singh V, Adhikary S, Pandita S, Roy S, Ramos K, Das C. Role of the Histone Acetyl Transferase MOF and the Histone Deacetylase Sirtuins in Regulation of H4K16ac During DNA Damage Repair and Metabolic Programming: Implications in Cancer and Aging. Subcell Biochem 2022; 100:115-141. [PMID: 36301493 DOI: 10.1007/978-3-031-07634-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The accurate repair of genomic damage mediated by ionizing radiation (IR), chemo- or radiomimetic drugs, or other exogenous agents, is necessary for maintenance of genome integrity, preservation of cellular viability and prevention of oncogenic transformation. Eukaryotes have conserved mechanisms designed to perceive and repair the damaged DNA quite efficiently. Among the different types of DNA damage, double strand breaks (DSB) are the most detrimental. The cellular DNA DSB response is a hierarchical signaling network that integrates damage sensing and repair with chromatin structural changes that involve a range of pre-existing and induced covalent modifications. Recent studies have revealed that pre-existing histone modifications are important contributors within this signaling/repair network. This chapter discusses the role of a critical histone acetyl transferase (HAT) known as MOF (males absent on the first) and the histone deacetylases (HDACs) Sirtuins on histone H4K16 acetylation (H4K16ac) and DNA damage repair. We also discuss the role of this important histone modification in light of metabolic rewiring and its role in regulating human pathophysiologic states.
Collapse
Affiliation(s)
- Tej K Pandita
- The Houston Methodist Research Institute, Houston, TX, USA.
- Department of Cellular and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, USA.
| | - Clayton R Hunt
- The Houston Methodist Research Institute, Houston, TX, USA
| | - Vipin Singh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhaba National Institute, Mumbai, India
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Shruti Pandita
- Department of Internal Medicine, Division of Hematology, Oncology and Cellular Therapy, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Siddhartha Roy
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Kenneth Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, USA
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
7
|
Epigenetic repression of Wnt receptors in AD: a role for Sirtuin2-induced H4K16ac deacetylation of Frizzled1 and Frizzled7 promoters. Mol Psychiatry 2022; 27:3024-3033. [PMID: 35296808 PMCID: PMC9205772 DOI: 10.1038/s41380-022-01492-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Growing evidence supports a role for deficient Wnt signalling in Alzheimer's disease (AD). First, the Wnt antagonist DKK1 is elevated in AD brains and is required for amyloid-β-induced synapse loss. Second, LRP6 Wnt co-receptor is required for synapse integrity and three variants of this receptor are linked to late-onset AD. However, the expression/role of other Wnt signalling components remain poorly explored in AD. Wnt receptors Frizzled1 (Fzd1), Fzd5, Fzd7 and Fzd9 are of interest due to their role in synapse formation/plasticity. Our analyses showed reduced FZD1 and FZD7 mRNA levels in the hippocampus of human early AD stages and in the hAPPNLGF/NLGF mouse model. This transcriptional downregulation was accompanied by reduced levels of the pro-transcriptional histone mark H4K16ac and a concomitant increase of its deacetylase Sirt2 at Fzd1 and Fzd7 promoters in AD. In vitro and in vivo inhibition of Sirt2 rescued Fzd1 and Fzd7 mRNA expression and H4K16ac levels at their promoters. In addition, we showed that Sirt2 recruitment to Fzd1 and Fzd7 promoters is dependent on FoxO1 activity in AD, thus acting as a co-repressor. Finally, we found reduced levels of SIRT2 inhibitory phosphorylation in nuclear samples from human early AD stages with a concomitant increase in the SIRT2 phosphatase PP2C. This results in hyperactive nuclear Sirt2 and favours Fzd1 and Fzd7 repression in AD. Collectively, our findings define a novel role for nuclear hyperactivated SIRT2 in repressing Fzd1 and Fzd7 expression via H4K16ac deacetylation in AD. We propose SIRT2 as an attractive target to ameliorate AD pathology.
Collapse
|
8
|
Guo X, Cui C, Song J, He Q, Zang N, Hu H, Wang X, Li D, Wang C, Hou X, Li X, Liang K, Yan F, Chen L. Mof acetyltransferase inhibition ameliorates glucose intolerance and islet dysfunction of type 2 diabetes via targeting pancreatic α-cells. Mol Cell Endocrinol 2021; 537:111425. [PMID: 34391847 DOI: 10.1016/j.mce.2021.111425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Previously, we reported that Mof was highly expressed in α-cells, and its knockdown led to ameliorated fasting blood glucose (FBG) and glucose tolerance in non-diabetic mice, attributed by reduced total α-cell but enhanced prohormone convertase (PC)1/3-positive α-cell mass. However, how Mof and histone 4 lysine 16 acetylation (H4K16ac) control α-cell and whether Mof inhibition improves glucose handling in type 2 diabetes (T2DM) mice remain unknown. METHODS Mof overexpression and chromatin immunoprecipitation sequence (ChIP-seq) based on H4K16ac were applied to determine the effect of Mof on α-cell transcriptional factors and underlying mechanism. Then we administrated mg149 to α-TC1-6 cell line, wild type, db/db and diet-induced obesity (DIO) mice to observe the impact of Mof inhibition in vitro and in vivo. In vitro, western blotting and TUNEL staining were used to examine α-cell apoptosis and function. In vivo, glucose tolerance, hormone levels, islet population, α-cell ratio and the co-staining of glucagon and PC1/3 or PC2 were examined. RESULTS Mof activated α-cell-specific transcriptional network. ChIP-seq results indicated that H4K16ac targeted essential genes regulating α-cell differentiation and function. Mof activity inhibition in vitro caused impaired α-cell function and enhanced apoptosis. In vivo, it contributed to ameliorated glucose intolerance and islet dysfunction, characterized by decreased fasting glucagon and elevated post-challenge insulin levels in T2DM mice. CONCLUSION Mof regulates α-cell differentiation and function via acetylating H4K16ac and H4K16ac binding to Pax6 and Foxa2 promoters. Mof inhibition may be a potential interventional target for T2DM, which led to decreased α-cell ratio but increased PC1/3-positive α-cells.
Collapse
Affiliation(s)
- Xinghong Guo
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Chen Cui
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Jia Song
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Qin He
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Nan Zang
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Huiqing Hu
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaojie Wang
- Department of Pharmacology, Basic Medicine School of Shandong University, Jinan, 250012, Shandong, China
| | - Danyang Li
- Department of Rehabilitation, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Chuan Wang
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Xinguo Hou
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Xiangzhi Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Life Science School of Shandong University, Qingdao, 266237, Shandong, China
| | - Kai Liang
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China; Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, 250012, Shandong, China
| | - Fei Yan
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China; Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, 250012, Shandong, China.
| | - Li Chen
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China; Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, 250012, Shandong, China.
| |
Collapse
|
9
|
Vazquez BN, Vaquero A, Schindler K. Sirtuins in female meiosis and in reproductive longevity. Mol Reprod Dev 2020; 87:1175-1187. [PMID: 33184962 PMCID: PMC7775317 DOI: 10.1002/mrd.23437] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/01/2020] [Indexed: 01/03/2023]
Abstract
Transmission of genetic material through high-quality gametes to progeny requires accurate homologous chromosome recombination and segregation during meiosis. A failure to accomplish these processes can have major consequences in reproductive health, including infertility, and development disorders in offspring. Sirtuins, a family of NAD+ -dependent protein deacetylases and ADP-ribosyltransferases, play key roles in genome maintenance, metabolism, and aging. In recent years, Sirtuins have emerged as regulators of several reproductive processes and interventions aiming to target Sirtuin activity are of great interest in the reproductive biology field. Sirtuins are pivotal to protect germ cells against oxidative stress, a major determinant influencing ovarian aging and the quality of gametes. Sirtuins also safeguard the integrity of the genome through epigenetic programs required for regulating gene repression, DNA repair, and chromosome segregation, among others. Although these functions are relatively well characterized in many somatic tissues, how they contribute to reproductive functions is not well understood. This review summarizes our current knowledge on the role of Sirtuins in female reproductive systems and discusses the underlying molecular pathways. In addition, we highlight the importance of Sirtuins as antiaging factors in the ovary and summarize current preclinical efforts to identify treatments to extend female reproductive longevity.
Collapse
Affiliation(s)
- Berta N. Vazquez
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Catalonia, Spain
- Department de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Alejandro Vaquero
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Catalonia, Spain
| | - Karen Schindler
- Human Genetics Institute of New Jersey (HGINJ), Department of Genetics, Rutgers University, 145 Bevier Rd., Piscataway, NJ, 08854, USA
| |
Collapse
|
10
|
Wang G, Song L, Bai T, Liang W. BcSas2-Mediated Histone H4K16 Acetylation Is Critical for Virulence and Oxidative Stress Response of Botrytis cinerea. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2020; 33:1242-1251. [PMID: 32689887 DOI: 10.1094/mpmi-06-20-0149-r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Histone acetyltransferase plays a critical role in transcriptional regulation by increasing accessibility of target genes to transcriptional activators. Botrytis cinerea is an important necrotrophic fungal pathogen with worldwide distribution and a very wide host range, but little is known of how the fungus regulates the transition from saprophytic growth to infectious growth. Here, the function of BcSas2, a histone acetyltransferase of B. cinerea, was investigated. Deletion of the BcSAS2 gene resulted in significantly reduced acetylation levels of histone H4, particularly of H4K16ac. The deletion mutant ΔBcSas2.1 was not only less pathogenic but also more sensitive to oxidative stress than the wild-type strain. RNA-Seq analysis revealed that a total of 13 B. cinerea genes associated with pathogenicity were down-regulated in the ΔBcSas2.1 mutant. Independent knockouts of two of these genes, BcXYGA (xyloglucanase) and BcCAT (catalase), led to dramatically decreased virulence and hypersensitivity to oxidative stress, respectively. Chromatin immunoprecipitation followed by quantitative PCR confirmed that BcSas2 bound directly to the promoter regions of both these pathogenicity-related genes. These observations indicated that BcSas2 regulated the transcription of pathogenicity-related genes by controlling the acetylation level of H4K16, thereby affecting the virulence and oxidative sensitivity of B. cinerea.
Collapse
Affiliation(s)
- Guangyuan Wang
- College of Life Sciences, Shandong Province Key Laboratory of Applied Mycology, Qingdao Agricultural University, Qingdao 266109, China
| | - Limin Song
- College of Plant Health and Medicine, the Key Laboratory of Integrated Crop Pest Management of Shandong Province, Qingdao Agricultural University, Qingdao 266109, China
| | - Tingting Bai
- College of Plant Health and Medicine, the Key Laboratory of Integrated Crop Pest Management of Shandong Province, Qingdao Agricultural University, Qingdao 266109, China
| | - Wenxing Liang
- College of Plant Health and Medicine, the Key Laboratory of Integrated Crop Pest Management of Shandong Province, Qingdao Agricultural University, Qingdao 266109, China
| |
Collapse
|
11
|
Singh M, Bacolla A, Chaudhary S, Hunt CR, Pandita S, Chauhan R, Gupta A, Tainer JA, Pandita TK. Histone Acetyltransferase MOF Orchestrates Outcomes at the Crossroad of Oncogenesis, DNA Damage Response, Proliferation, and Stem Cell Development. Mol Cell Biol 2020; 40:e00232-20. [PMID: 32661120 PMCID: PMC7459263 DOI: 10.1128/mcb.00232-20] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The DNA and protein complex known as chromatin is subject to posttranslational modifications (PTMs) that regulate cellular functions such that PTM dysregulation can lead to disease, including cancer. One critical PTM is acetylation/deacetylation, which is being investigated as a means to develop targeted cancer therapies. The histone acetyltransferase (HAT) family of proteins performs histone acetylation. In humans, MOF (hMOF), a member of the MYST family of HATs, acetylates histone H4 at lysine 16 (H4K16ac). MOF-mediated acetylation plays a critical role in the DNA damage response (DDR) and embryonic stem cell development. Functionally, MOF is found in two distinct complexes: NSL (nonspecific lethal) in humans and MSL (male-specific lethal) in flies. The NSL complex is also able to acetylate additional histone H4 sites. Dysregulation of MOF activity occurs in multiple cancers, including ovarian cancer, medulloblastoma, breast cancer, colorectal cancer, and lung cancer. Bioinformatics analysis of KAT8, the gene encoding hMOF, indicated that it is highly overexpressed in kidney tumors as part of a concerted gene coexpression program that can support high levels of chromosome segregation and cell proliferation. The linkage between MOF and tumor proliferation suggests that there are additional functions of MOF that remain to be discovered.
Collapse
Affiliation(s)
- Mayank Singh
- Department of Medical Oncology, BRAIRCH, All India Institute of Medical Sciences Delhi, New Delhi, India
| | - Albino Bacolla
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Shilpi Chaudhary
- Department of Medical Oncology, BRAIRCH, All India Institute of Medical Sciences Delhi, New Delhi, India
| | - Clayton R Hunt
- The Houston Methodist Research Institute, Houston, Texas, USA
| | - Shruti Pandita
- Department of Internal Medicine, Division of Hematology, Oncology and Cellular Therapy, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Ravi Chauhan
- Department of Medical Oncology, BRAIRCH, All India Institute of Medical Sciences Delhi, New Delhi, India
| | - Ashna Gupta
- Department of Medical Oncology, BRAIRCH, All India Institute of Medical Sciences Delhi, New Delhi, India
| | - John A Tainer
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Tej K Pandita
- The Houston Methodist Research Institute, Houston, Texas, USA
- Department of Cellular and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
12
|
Ray A, Khan P, Nag Chaudhuri R. Deacetylation of H4 lysine16 affects acetylation of lysine residues in histone H3 and H4 and promotes transcription of constitutive genes. Epigenetics 2020; 16:597-617. [PMID: 32795161 DOI: 10.1080/15592294.2020.1809896] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Histone modification map of H4 N-terminal tail residues in Saccharomyces cerevisiae reveals the prominence of lysine acetylation. Previous reports have indicated the importance of lysine acetylation in maintaining chromatin structure and function. H4K16, a residue with highly regulated acetylation dynamics has unique functions not overlapping with the other H4 N- terminal acetylable residues. The present work unravels the role of H4K16 acetylation in regulating expression of constitutive genes. H4K16 gets distinctly deacetylated over the coding region of constitutively expressed genes. Deacetylation of H4K16 reduces H3K9 acetylation at the cellular and gene level. Reduced H3K9 acetylation however did not negatively correlate with active gene transcription. Significantly, H4K16 deacetylation was found to be associated with hypoacetylated H4K12 throughout the locus of constitutive genes. H4K16 and K12 deacetylation is known to favour active transcription. Sas2, the HAT mutant showed similar patterns of hypoacetylated H3K9 and H4K12 at the active loci, clearly implying that the modifications were associated with deacetylation state of H4K16. Deacetylation of H4K16 was also concurrent with increased H3K56 acetylation in the promoter region and ORF of the constitutive genes. Combination of all these histone modifications significantly reduced H3 occupancy, increased promoter accessibility and enhanced RNAPII recruitment at the constitutively active loci. Consequently, we found that expression of active genes was higher in H4K16R mutant which mimic deacetylated state, but not in H4K16Q mimicking constitutive acetylation. To summarize, H4K16 deacetylation linked with H4K12 and H3K9 hypoacetylation along with H3K56 hyperacetylation generate a chromatin landscape that is conducive for transcription of constitutive genes.
Collapse
Affiliation(s)
- Anagh Ray
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| | - Preeti Khan
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| | | |
Collapse
|
13
|
Banirazi Motlagh N, Mohammadpour Esfahani B, Ashrafi B, Zare-Mirakabad F. The assessment of histone acetylation marks in the vicinity of transcription factor binding sites in human CD4 + T cells using information theory methods. Comput Biol Chem 2020; 86:107232. [PMID: 32142982 DOI: 10.1016/j.compbiolchem.2020.107232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 01/29/2019] [Accepted: 02/08/2020] [Indexed: 11/24/2022]
Abstract
The genetic information encoded in structural genes is decoded by an intracellular process called gene expression. This mechanism is regulated by epigenetic processes such as histone acetylation. Histone acetylation, which happens in nucleosomes, exposes DNA (genome) to transcription factors. Therefore, the correlation between histone acetylation and gene expression has been assessed as a fundamental issue in many previous studies. In the proposed research, we investigate which marks of histone acetylation are informative and which ones are redundant in the vicinity of SP1 transcription factor binding sites, in human CD4 + T cell. To achieve this, we use information theory methods. Subsequently, we apply a multilayer perceptron neural network to show that the selected histone acetylation marks by information theory methods are sufficiently informative. Finally, we use the neural network to predict binding sites of 17 other transcription factors on chromosomes 1 and 2. The results suggest that information conveyed by the selected histone acetylation marks are equivalent to that of all 18 marks associated with SP1 transcription factor binding sites on chromosome 1. Furthermore, almost 91.75 % of SP1 binding sites of chromosome 2 are predicted by the selected histone acetylation marks while all 18 marks predict 90.56 % correctly. Moreover, the selected histone acetylation marks are efficient at predicting 17 other types of transcription factor binding sites.
Collapse
Affiliation(s)
- Nafiseh Banirazi Motlagh
- Department of Mathematics and Computer Science, Amirkabir University of Technology, Tehran, Iran
| | | | - Behnoosh Ashrafi
- Department of Mathematics and Computer Science, Amirkabir University of Technology, Tehran, Iran
| | - Fatemeh Zare-Mirakabad
- Department of Mathematics and Computer Science, Amirkabir University of Technology, Tehran, Iran.
| |
Collapse
|
14
|
Xu SY, Lv HQ, Li WQ, Hong H, Peng YJ, Zhu BM. Electroacupuncture Alleviates Cerebral Ischemia/Reperfusion Injury in Rats by Histone H4 Lysine 16 Acetylation-Mediated Autophagy. Front Psychiatry 2020; 11:576539. [PMID: 33391046 PMCID: PMC7775364 DOI: 10.3389/fpsyt.2020.576539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/19/2020] [Indexed: 02/05/2023] Open
Abstract
Background: Electroacupuncture (EA) treatment in ischemic stroke has been highlighted recently; however, the specific mechanism is still elusive. Autophagy is considered a new target for cerebral ischemia/reperfusion (I/R), but whether it plays a role of protecting or causing rapid cell apoptosis remains unclear. Studies have reported that the reduction in lysine 16 of histone H4 acetylation coheres with autophagy induction. The primary purpose of the study was to explore whether EA could alleviate I/R via autophagy-mediated histone H4 lysine 16 acetylation in the middle cerebral artery occlusion (MCAO) rat model. Methods: One hundred and twenty male Sprague-Dawley rats were divided into five groups: control group, MCAO group, MCAO+EA group, MCAO+EA+hMOF siRNA group, and MCAO+EA+Sirt1 inhibitor group. EA was applied to "Baihui" (Du20) and "Renzhong" (Du26) at 5 min after modeling and 16 h after the first EA intervention. The structure and molecular markers of the rat brain were evaluated. Results: EA significantly alleviated I/R injury by upregulating the expressions of Sirt1, Beclin1, and LC3-II and downregulating the expressions of hMOF and H4K16ac. In contrast, the Sirt1 inhibitor lowered the increase in Sirt1, Beclin1, and LC3-II and enhanced the level of hMOF and H4K16ac expressions associated with EA treatment. Besides, ChIP assay revealed that the binding of H4K16ac in the Beclin1 promoter region of the autophagy target gene was significantly raised in the MCAO+EA group and MCAO+EA+hMOF siRNA group. Conclusions: EA treatment inhibited the H4K16ac process, facilitated autophagy, and alleviated I/R injury. These findings suggested that regulating histone H4 lysine 16 acetylation-mediated autophagy may be a key mechanism of EA at Du20 and Du26 to treat I/R.
Collapse
Affiliation(s)
- Shu-Ying Xu
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - He-Qun Lv
- Department of Acupuncture and Encephalopathy, Yancheng Hospital of Traditional Chinese Medicine, Yancheng, China
| | - Wen-Qian Li
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Hong
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong-Jun Peng
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Bing-Mei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Urdinguio RG, Lopez V, Bayón GF, Diaz de la Guardia R, Sierra MI, García-Toraño E, Perez RF, García MG, Carella A, Pruneda PC, Prieto C, Dmitrijeva M, Santamarina P, Belmonte T, Mangas C, Diaconu E, Ferrero C, Tejedor JR, Fernandez-Morera JL, Bravo C, Bueno C, Sanjuan-Pla A, Rodriguez RM, Suarez-Alvarez B, López-Larrea C, Bernal T, Colado E, Balbín M, García-Suarez O, Chiara MD, Sáenz-de-Santa-María I, Rodríguez F, Pando-Sandoval A, Rodrigo L, Santos L, Salas A, Vallejo-Díaz J, C Carrera A, Rico D, Hernández-López I, Vayá A, Ricart JM, Seto E, Sima-Teruel N, Vaquero A, Valledor L, Cañal MJ, Pisano D, Graña-Castro O, Thomas T, Voss AK, Menéndez P, Villar-Garea A, Deutzmann R, Fernandez AF, Fraga MF. Chromatin regulation by Histone H4 acetylation at Lysine 16 during cell death and differentiation in the myeloid compartment. Nucleic Acids Res 2019; 47:5016-5037. [PMID: 30923829 PMCID: PMC6547425 DOI: 10.1093/nar/gkz195] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 02/26/2019] [Accepted: 03/15/2019] [Indexed: 12/14/2022] Open
Abstract
Histone H4 acetylation at Lysine 16 (H4K16ac) is a key epigenetic mark involved in gene regulation, DNA repair and chromatin remodeling, and though it is known to be essential for embryonic development, its role during adult life is still poorly understood. Here we show that this lysine is massively hyperacetylated in peripheral neutrophils. Genome-wide mapping of H4K16ac in terminally differentiated blood cells, along with functional experiments, supported a role for this histone post-translational modification in the regulation of cell differentiation and apoptosis in the hematopoietic system. Furthermore, in neutrophils, H4K16ac was enriched at specific DNA repeats. These DNA regions presented an accessible chromatin conformation and were associated with the cleavage sites that generate the 50 kb DNA fragments during the first stages of programmed cell death. Our results thus suggest that H4K16ac plays a dual role in myeloid cells as it not only regulates differentiation and apoptosis, but it also exhibits a non-canonical structural role in poising chromatin for cleavage at an early stage of neutrophil cell death.
Collapse
Affiliation(s)
- Rocio G Urdinguio
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Spain.,Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Virginia Lopez
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Spain
| | - Gustavo F Bayón
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Rafael Diaz de la Guardia
- Josep Carreras Leukemia Research Institute and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Cáncer (CIBER-ONC), Barcelona, Spain
| | - Marta I Sierra
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Estela García-Toraño
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Raúl F Perez
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Spain.,Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - María G García
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Spain.,Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Antonella Carella
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Spain.,Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Patricia C Pruneda
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Cristina Prieto
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Marija Dmitrijeva
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Pablo Santamarina
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Spain.,Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Thalía Belmonte
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Spain.,Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Cristina Mangas
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Elena Diaconu
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Cecilia Ferrero
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Juan Ramón Tejedor
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Juan Luis Fernandez-Morera
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Cristina Bravo
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Clara Bueno
- Josep Carreras Leukemia Research Institute and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Cáncer (CIBER-ONC), Barcelona, Spain
| | - Alejandra Sanjuan-Pla
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, 46026, Spain
| | - Ramon M Rodriguez
- Translational Immunology Laboratory, Instituto de Investigación Sanitarias del Principado de Asturias (ISPA), Immunology Department, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Beatriz Suarez-Alvarez
- Translational Immunology Laboratory, Instituto de Investigación Sanitarias del Principado de Asturias (ISPA), Immunology Department, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Carlos López-Larrea
- Translational Immunology Laboratory, Instituto de Investigación Sanitarias del Principado de Asturias (ISPA), Immunology Department, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Teresa Bernal
- Servicio de Hematología, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Enrique Colado
- Servicio de Hematología, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Milagros Balbín
- Service of Molecular Oncology, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - Olivia García-Suarez
- Department of Morphology and Cellular Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
| | - María Dolores Chiara
- Otorhinolaryngology Service, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, CIBERONC, Oviedo, Spain
| | - Inés Sáenz-de-Santa-María
- Otorhinolaryngology Service, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, CIBERONC, Oviedo, Spain
| | - Francisco Rodríguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Ana Pando-Sandoval
- Hospital Universitario Central de Asturias (HUCA), Instituto Nacional de Silicosis (INS), Área del Pulmón, Facultad de Medicina, Universidad de Oviedo, Avenida Roma s/n, Oviedo, Asturias 33011, Spain
| | - Luis Rodrigo
- Hospital Universitario Central de Asturias (HUCA), Gastroenterology Service, Facultad de Medicina, Universidad de Oviedo, Avenida de Roma s/n, Oviedo, Asturias 33011, Spain
| | - Laura Santos
- Fundación para la Investigación Biosanitaria de Asturias (FINBA). Instituto de Investigación Sanitaria del Principado de Asturias (ISPA). Avenida de Roma s/n, 33011 Oviedo. Asturias. España
| | - Ana Salas
- Cytometry Service, Servicios Científico-Técnicos (SCTs). Universidad de Oviedo, Oviedo, Spain
| | - Jesús Vallejo-Díaz
- Department of Immunology and Oncology, National Center for Biotechnology, CNB-CSIC, Cantoblanco, 28049 Madrid, Spain
| | - Ana C Carrera
- Department of Immunology and Oncology, National Center for Biotechnology, CNB-CSIC, Cantoblanco, 28049 Madrid, Spain
| | - Daniel Rico
- Institute of Cellular Medicine, Newcastle University, UK
| | | | - Amparo Vayá
- Hemorheology and Haemostasis Unit, Service of Clinical Pathology, La Fe University Hospital, Valencia, Spain
| | | | - Edward Seto
- George Washington University Cancer Center, Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC 20037, USA
| | - Núria Sima-Teruel
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l'Hospitalet, 199-203, 08907- L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alejandro Vaquero
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l'Hospitalet, 199-203, 08907- L'Hospitalet de Llobregat, Barcelona, Spain
| | - Luis Valledor
- Plant Physiology Lab, Department of Organisms and Systems Biology, Faculty of Biology, University of Oviedo, Oviedo, Asturias, Spain
| | - Maria Jesus Cañal
- Plant Physiology Lab, Department of Organisms and Systems Biology, Faculty of Biology, University of Oviedo, Oviedo, Asturias, Spain
| | - David Pisano
- Bioinformatics Unit, Structural Biology and Biocomputing Program, Spanish National Cancer Research Center (CNIO), C/ Melchor Fernández Almagro, 3. 28029 Madrid, Spain
| | - Osvaldo Graña-Castro
- Bioinformatics Unit, Structural Biology and Biocomputing Program, Spanish National Cancer Research Center (CNIO), C/ Melchor Fernández Almagro, 3. 28029 Madrid, Spain
| | - Tim Thomas
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Anne K Voss
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Cáncer (CIBER-ONC), Barcelona, Spain.,Instituciò Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Ana Villar-Garea
- Institute of Biochemistry, Genetics and Microbiology, University of Regensburg, 93053 Regensburg, Germany
| | - Rainer Deutzmann
- Institute of Biochemistry, Genetics and Microbiology, University of Regensburg, 93053 Regensburg, Germany
| | - Agustín F Fernandez
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), ISPA-Hospital Universitario Central de Asturias HUCA, Universidad de Oviedo, Oviedo, Spain
| | - Mario F Fraga
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo-Principado de Asturias, Spain
| |
Collapse
|
16
|
Hunt CR, Pandita TK. "What's Past is Prologue": Pre-Existing Epigenetic Transcriptional Marks May Also Influence DNA Repair Pathway Choice. Radiat Res 2019; 192:577-578. [PMID: 31634055 DOI: 10.1667/rr15541.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Clayton R Hunt
- The Houston Methodist Research Institute, Houston, Texas 77030
| | - Tej K Pandita
- The Houston Methodist Research Institute, Houston, Texas 77030
| |
Collapse
|
17
|
Majora M, Sondenheimer K, Knechten M, Uthe I, Esser C, Schiavi A, Ventura N, Krutmann J. HDAC inhibition improves autophagic and lysosomal function to prevent loss of subcutaneous fat in a mouse model of Cockayne syndrome. Sci Transl Med 2019; 10:10/456/eaam7510. [PMID: 30158153 DOI: 10.1126/scitranslmed.aam7510] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 11/25/2017] [Accepted: 07/31/2018] [Indexed: 01/02/2023]
Abstract
Cockayne syndrome (CS), a hereditary form of premature aging predominantly caused by mutations in the csb gene, affects multiple organs including skin where it manifests with hypersensitivity toward ultraviolet (UV) radiation and loss of subcutaneous fat. There is no curative treatment for CS, and its pathogenesis is only partially understood. Originally considered for its role in DNA repair, Cockayne syndrome group B (CSB) protein most likely serves additional functions. Using CSB-deficient human fibroblasts, Caenorhabditiselegans, and mice, we show that CSB promotes acetylation of α-tubulin and thereby regulates autophagy. At the organ level, chronic exposure of csbm/m mice to UVA radiation caused a severe skin phenotype with loss of subcutaneous fat, inflammation, and fibrosis. These changes in skin tissue were associated with an accumulation of autophagic/lysosomal proteins and reduced amounts of acetylated α-tubulin. At the cellular level, we found that CSB directly interacts with the histone deacetylase 6 (HDAC6) and the α-tubulin acetyltransferase MEC-17. Upon UVA irradiation, CSB is recruited to the centrosome where it colocalizes with dynein and HDAC6. Administration of the pan-HDAC inhibitor SAHA (suberoylanilide hydroxamic acid) enhanced α-tubulin acetylation, improved autophagic function in CSB-deficient models from all three species, and rescued the skin phenotype in csbm/m mice. HDAC inhibition may thus represent a therapeutic option for CS.
Collapse
Affiliation(s)
- Marc Majora
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Kevin Sondenheimer
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Maren Knechten
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Ingo Uthe
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Charlotte Esser
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Alfonso Schiavi
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Natascia Ventura
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany.,Institute of Clinical Chemistry and Laboratory Diagnostics, University of Düsseldorf, Medical Faculty, 40225 Düsseldorf, Germany
| | - Jean Krutmann
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany. .,Medical Faculty, University of Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
18
|
Wang JQ, Yan FQ, Wang LH, Yin WJ, Chang TY, Liu JP, Wu KJ. Identification of new hypoxia-regulated epithelial-mesenchymal transition marker genes labeled by H3K4 acetylation. Genes Chromosomes Cancer 2019; 59:73-83. [PMID: 31408253 DOI: 10.1002/gcc.22802] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/05/2019] [Accepted: 08/09/2019] [Indexed: 12/19/2022] Open
Abstract
Hypoxia-induced epithelial-mesenchymal transition (EMT) involves the interplay between chromatin modifiers histone deacetylase 3 (HDAC3) and WDR5. The histone mark histone 3 lysine 4 acetylation (H3K4Ac) is observed in the promoter regions of various EMT marker genes (eg, CDH1 and VIM). To further define the genome-wide location of H3K4Ac, a chromatin immunoprecipitation followed by massively parallel DNA sequencing (ChIP-seq) analysis was performed using a head and neck squamous cell carcinoma (HNSCC) FaDu cell line under normoxia and hypoxia. H3K4Ac was found to be located mainly around the transcription start site. Coupled with analysis of gene expression by RNA sequencing and using a HDAC3 knockdown cell line, 10 new genes (BMI1, GLI1, SMO, FOXF1, SIRT2, etc) that were labeled by H3K4Ac and regulated by HDAC3 were identified. Overexpression or knockdown of GLI1/SMO increased or repressed the in vitro migration and invasion activity in OECM-1/FaDu cells, respectively. In HNSCC patients, coexpression of GLI1 and SMO in primary tumors correlated with metastasis. Our results identify new EMT marker genes that may play a significant role in hypoxia-induced EMT and metastasis and further provide diagnostic and prognostic implications.
Collapse
Affiliation(s)
- Jian-Qiu Wang
- Institute of Ageing Research, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Feng-Qin Yan
- Department of Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Li-Hui Wang
- Institute of Ageing Research, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Wen-Juan Yin
- Department of Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ting-Yu Chang
- Institute of Microbiology & Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Jun-Ping Liu
- Institute of Ageing Research, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Kou-Juey Wu
- Cancer Genome Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Research Center for Tumor Medical Science, Graduate Institute of New Drug Development, China Medical University, Taichung, Taiwan
| |
Collapse
|
19
|
Horikoshi N, Sharma D, Leonard F, Pandita RK, Charaka VK, Hambarde S, Horikoshi NT, Gaur Khaitan P, Chakraborty S, Cote J, Godin B, Hunt CR, Pandita TK. Pre-existing H4K16ac levels in euchromatin drive DNA repair by homologous recombination in S-phase. Commun Biol 2019; 2:253. [PMID: 31286070 PMCID: PMC6611875 DOI: 10.1038/s42003-019-0498-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 05/29/2019] [Indexed: 12/22/2022] Open
Abstract
The homologous recombination (HR) repair pathway maintains genetic integrity after DNA double-strand break (DSB) damage and is particularly crucial for maintaining fidelity of expressed genes. Histone H4 acetylation on lysine 16 (H4K16ac) is associated with transcription, but how pre-existing H4K16ac directly affects DSB repair is not known. To answer this question, we used CRISPR/Cas9 technology to introduce I-SceI sites, or repair pathway reporter cassettes, at defined locations within gene-rich (high H4K16ac/euchromatin) and gene-poor (low H4K16ac/heterochromatin) regions. The frequency of DSB repair by HR is higher in gene-rich regions. Interestingly, artificially targeting H4K16ac at specific locations using gRNA/dCas9-MOF increases HR frequency in euchromatin. Finally, inhibition/depletion of RNA polymerase II or Cockayne syndrome B protein leads to decreased recruitment of HR factors at DSBs. These results indicate that the pre-existing H4K16ac status at specific locations directly influences the repair of local DNA breaks, favoring HR in part through the transcription machinery.
Collapse
Affiliation(s)
- Nobuo Horikoshi
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA
- Present Address: Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Dharmendra Sharma
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA
| | - Fransisca Leonard
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, TX 77030 USA
| | - Raj K. Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA
| | - Vijaya K. Charaka
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA
| | - Shashank Hambarde
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA
| | - Nobuko T. Horikoshi
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA
| | - Puja Gaur Khaitan
- Department of Surgery, The Houston Methodist Research Institute, Houston, TX 77030 USA
- Present Address: Department of Surgery, Medstar Washington Hospital Center, Washington, DC 20010 USA
| | - Sharmistha Chakraborty
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA
| | - Jacques Cote
- St. Patrick Research Group in Basic Oncology, Laval University Cancer Research Center, Quebec City, QC G1V4G2 Canada
| | - Biana Godin
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, TX 77030 USA
| | - Clayton R. Hunt
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA
| | - Tej K. Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA
| |
Collapse
|
20
|
Chakraborty S, Pandita RK, Hambarde S, Mattoo AR, Charaka V, Ahmed KM, Iyer SP, Hunt CR, Pandita TK. SMARCAD1 Phosphorylation and Ubiquitination Are Required for Resection during DNA Double-Strand Break Repair. iScience 2018; 2:123-135. [PMID: 29888761 PMCID: PMC5993204 DOI: 10.1016/j.isci.2018.03.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/16/2018] [Accepted: 02/28/2018] [Indexed: 02/08/2023] Open
Abstract
The chromatin remodeling factor SMARCAD1, an SWI/SNF ATPase family member, has a role in 5' end resection at DNA double-strand breaks (DSBs) to produce single-strand DNA (ssDNA), a critical step for subsequent checkpoint and repair factor loading to remove DNA damage. However, the mechanistic details of SMARCAD1 coupling to the DNA damage response and repair pathways remains unknown. Here we report that SMARCAD1 is recruited to DNA DSBs through an ATM-dependent process. Depletion of SMARCAD1 reduces ionizing radiation (IR)-induced repairosome foci formation and DSB repair by homologous recombination (HR). IR induces SMARCAD1 phosphorylation at a conserved T906 by ATM kinase, a modification essential for SMARCAD1 recruitment to DSBs. Interestingly, T906 phosphorylation is also important for SMARCAD1 ubiquitination by RING1 at K905. Both these post-translational modifications are critical for regulating the role of SMARCAD1 in DNA end resection, HR-mediated repair, and cell survival after DNA damage.
Collapse
Affiliation(s)
- Sharmistha Chakraborty
- Department of Radiation Oncology, Houston Methodist Cancer Center, The Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA.
| | - Raj K Pandita
- Department of Radiation Oncology, Houston Methodist Cancer Center, The Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | - Shashank Hambarde
- Department of Radiation Oncology, Houston Methodist Cancer Center, The Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | - Abid R Mattoo
- Department of Radiation Oncology, Houston Methodist Cancer Center, The Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | - Vijaya Charaka
- Department of Radiation Oncology, Houston Methodist Cancer Center, The Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | - Kazi M Ahmed
- Department of Radiation Oncology, Houston Methodist Cancer Center, The Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | - Swaminathan P Iyer
- Department of Hematology, Houston Methodist Cancer Center, The Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | - Clayton R Hunt
- Department of Radiation Oncology, Houston Methodist Cancer Center, The Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | - Tej K Pandita
- Department of Radiation Oncology, Houston Methodist Cancer Center, The Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA.
| |
Collapse
|
21
|
Epigenetic Modifications as Biomarkers of Tumor Development, Therapy Response, and Recurrence across the Cancer Care Continuum. Cancers (Basel) 2018; 10:cancers10040101. [PMID: 29614786 PMCID: PMC5923356 DOI: 10.3390/cancers10040101] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 02/06/2023] Open
Abstract
Aberrant epigenetic modifications are an early event in carcinogenesis, with the epigenetic landscape continuing to change during tumor progression and metastasis—these observations suggest that specific epigenetic modifications could be used as diagnostic and prognostic biomarkers for many cancer types. DNA methylation, post-translational histone modifications, and non-coding RNAs are all dysregulated in cancer and are detectable to various degrees in liquid biopsies such as sputum, urine, stool, and blood. Here, we will focus on the application of liquid biopsies, as opposed to tissue biopsies, because of their potential as non-invasive diagnostic tools and possible use in monitoring therapy response and progression to metastatic disease. This includes a discussion of septin-9 (SEPT9) DNA hypermethylation for detecting colorectal cancer, which is by far the most developed epigenetic biomarker assay. Despite their potential as prognostic and diagnostic biomarkers, technical issues such as inconsistent methodology between studies, overall low yield of epigenetic material in samples, and the need for improved histone and non-coding RNA purification methods are limiting the use of epigenetic biomarkers. Once these technical limitations are overcome, epigenetic biomarkers could be used to monitor cancer development, disease progression, therapeutic response, and recurrence across the entire cancer care continuum.
Collapse
|
22
|
MOF Suppresses Replication Stress and Contributes to Resolution of Stalled Replication Forks. Mol Cell Biol 2018; 38:MCB.00484-17. [PMID: 29298824 DOI: 10.1128/mcb.00484-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/05/2017] [Indexed: 01/13/2023] Open
Abstract
The human MOF (hMOF) protein belongs to the MYST family of histone acetyltransferases and plays a critical role in transcription and the DNA damage response. MOF is essential for cell proliferation; however, its role during replication and replicative stress is unknown. Here we demonstrate that cells depleted of MOF and under replicative stress induced by cisplatin, hydroxyurea, or camptothecin have reduced survival, a higher frequency of S-phase-specific chromosome damage, and increased R-loop formation. MOF depletion decreased replication fork speed and, when combined with replicative stress, also increased stalled replication forks as well as new origin firing. MOF interacted with PCNA, a key coordinator of replication and repair machinery at replication forks, and affected its ubiquitination and recruitment to the DNA damage site. Depletion of MOF, therefore, compromised the DNA damage repair response as evidenced by decreased Mre11, RPA70, Rad51, and PCNA focus formation, reduced DNA end resection, and decreased CHK1 phosphorylation in cells after exposure to hydroxyurea or cisplatin. These results support the argument that MOF plays an important role in suppressing replication stress induced by genotoxic agents at several stages during the DNA damage response.
Collapse
|
23
|
Carr SM, Munro S, Sagum CA, Fedorov O, Bedford MT, La Thangue NB. Tudor-domain protein PHF20L1 reads lysine methylated retinoblastoma tumour suppressor protein. Cell Death Differ 2017; 24:2139-2149. [PMID: 28841214 PMCID: PMC5686351 DOI: 10.1038/cdd.2017.135] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/12/2017] [Accepted: 07/14/2017] [Indexed: 12/30/2022] Open
Abstract
The retinoblastoma tumour suppressor protein (pRb) classically functions to regulate early cell cycle progression where it acts to enforce a number of checkpoints in response to cellular stress and DNA damage. Methylation at lysine (K) 810, which occurs within a critical CDK phosphorylation site and antagonises a CDK-dependent phosphorylation event at the neighbouring S807 residue, acts to hold pRb in the hypo-phosphorylated growth-suppressing state. This is mediated in part by the recruitment of the reader protein 53BP1 to di-methylated K810, which allows pRb activity to be effectively integrated with the DNA damage response. Here, we report the surprising observation that an additional methylation-dependent interaction occurs at K810, but rather than the di-methyl mark, it is selective for the mono-methyl K810 mark. Binding of the mono-methyl PHF20L1 reader to methylated pRb occurs on E2F target genes, where it acts to mediate an additional level of control by recruiting the MOF acetyltransferase complex to E2F target genes. Significantly, we find that the interplay between PHF20L1 and mono-methyl pRb is important for maintaining the integrity of a pRb-dependent G1-S-phase checkpoint. Our results highlight the distinct roles that methyl-lysine readers have in regulating the biological activity of pRb.
Collapse
Affiliation(s)
- Simon M Carr
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Shonagh Munro
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Cari A Sagum
- Department of Molecular Carcinogenesis, The University of Texas, MD Anderson Cancer Center, Smithville, TX 77030, USA
| | - Oleg Fedorov
- Nuffield Department of Clinical Medicine, Structural Genomics Consortium Oxford, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Mark T Bedford
- Department of Molecular Carcinogenesis, The University of Texas, MD Anderson Cancer Center, Smithville, TX 77030, USA
| | - Nicholas B La Thangue
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
24
|
Marais TLD, Kluz T, Xu D, Zhang X, Gesumaria L, Matsui MS, Costa M, Sun H. Transcription factors and stress response gene alterations in human keratinocytes following Solar Simulated Ultra Violet Radiation. Sci Rep 2017; 7:13622. [PMID: 29051608 PMCID: PMC5648893 DOI: 10.1038/s41598-017-13765-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/27/2017] [Indexed: 12/11/2022] Open
Abstract
Ultraviolet radiation (UVR) from sunlight is the major effector for skin aging and carcinogenesis. However, genes and pathways altered by solar-simulated UVR (ssUVR), a mixture of UVA and UVB, are not well characterized. Here we report global changes in gene expression as well as associated pathways and upstream transcription factors in human keratinocytes exposed to ssUVR. Human HaCaT keratinocytes were exposed to either a single dose or 5 repetitive doses of ssUVR. Comprehensive analyses of gene expression profiles as well as functional annotation were performed at 24 hours post irradiation. Our results revealed that ssUVR modulated genes with diverse cellular functions changed in a dose-dependent manner. Gene expression in cells exposed to a single dose of ssUVR differed significantly from those that underwent repetitive exposures. While single ssUVR caused a significant inhibition in genes involved in cell cycle progression, especially G2/M checkpoint and mitotic regulation, repetitive ssUVR led to extensive changes in genes related to cell signaling and metabolism. We have also identified a panel of ssUVR target genes that exhibited persistent changes in gene expression even at 1 week after irradiation. These results revealed a complex network of transcriptional regulators and pathways that orchestrate the cellular response to ssUVR.
Collapse
Affiliation(s)
- Thomas L Des Marais
- New York University, Department of Environmental Medicine, Tuxedo, New York, United States of America
| | - Thomas Kluz
- New York University, Department of Environmental Medicine, Tuxedo, New York, United States of America
| | - Dazhong Xu
- New York Medical College School of Medicine, Department of Pathology, Valhalla, New York, United States of America
| | - Xiaoru Zhang
- New York University, Department of Environmental Medicine, Tuxedo, New York, United States of America
| | - Lisa Gesumaria
- New York University, Department of Environmental Medicine, Tuxedo, New York, United States of America
| | - Mary S Matsui
- Estee Lauder Companies, Inc., Melville, New York, United States of America
| | - Max Costa
- New York University, Department of Environmental Medicine, Tuxedo, New York, United States of America.
| | - Hong Sun
- New York University, Department of Environmental Medicine, Tuxedo, New York, United States of America.
| |
Collapse
|
25
|
Saidi D, Cheray M, Osman AM, Stratoulias V, Lindberg OR, Shen X, Blomgren K, Joseph B. Glioma-induced SIRT1-dependent activation of hMOF histone H4 lysine 16 acetyltransferase in microglia promotes a tumor supporting phenotype. Oncoimmunology 2017; 7:e1382790. [PMID: 29308302 PMCID: PMC5749650 DOI: 10.1080/2162402x.2017.1382790] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/14/2017] [Accepted: 09/16/2017] [Indexed: 11/06/2022] Open
Abstract
High-grade gliomas are malignant aggressive primary brain tumors with limited therapeutic options, and dismal prognosis for patients. Microglia, the resident immune cells of the brain, are recruited and reprogrammed into tumor-supporting cells by glioma cells, which in turn positively influence tumor expansion and infiltration into surrounding brain tissues. Here, we report that glioma-induced microglia conversion is coupled to an increase of histone H4 lysine 16 (H4K16) acetylation level in microglia, through increased nuclear localization of the deacetylase SIRT1, which in turn results in deacetylation of the H4K16 acetyltransferase hMOF and its recruitment to the chromatin at promoter regions of microglial target genes. Furthermore, we demonstrate that manipulation of the microglial H4K16 acetylation level, taking advantage of the intrinsic H4K16 deacetylase or acetyltransferase activities of SIRT1 and hMOF, respectively, modulated the tumor-supporting function of microglia. This study provides evidence that post-translational modifications of histones and the histone-modifying enzymes controlling them, such as H4K16 acetylation regulated by hMOF and SIRT1, are part of the microglial pro-tumoral activation pathway initiated by glioma cancer cells and represent potentially novel therapeutic targets.
Collapse
Affiliation(s)
- Dalel Saidi
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Mathilde Cheray
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Ahmed M Osman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Vassilis Stratoulias
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Olle R Lindberg
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Xianli Shen
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Bertrand Joseph
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
Wisnieski F, Leal MF, Calcagno DQ, Santos LC, Gigek CO, Chen ES, Artigiani R, Demachki S, Assumpção PP, Lourenço LG, Burbano RR, Smith MC. BMP8B Is a Tumor Suppressor Gene Regulated by Histone Acetylation in Gastric Cancer. J Cell Biochem 2016; 118:869-877. [PMID: 27748538 DOI: 10.1002/jcb.25766] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/14/2016] [Indexed: 12/13/2022]
Abstract
Different from genetic alterations, the reversible nature of epigenetic modifications provides an interesting opportunity for the development of clinically relevant therapeutics in different tumors. In this study, we aimed to screen and validate candidate genes regulated by the epigenetic marker associated with transcriptional activation, histone acetylation, in gastric cancer (GC). We first compared gene expression profile of trichostatin A-treated and control GC cell lines using microarray assay. Among the 55 differentially expressed genes identified in this analysis, we chose the up-regulated genes BMP8B and BAMBI for further analyses, that included mRNA and histone acetylation quantification in paired GC and nontumor tissue samples. BMP8B expression was reduced in GC compared to nontumor samples (P < 0.01). In addition, reduced BMP8B expression was associated with poorly differentiated GC (P = 0.02). No differences or histopathological associations were identified concerning BAMBI expression. Furthermore, acetylated H3K9 and H4K16 levels at BMP8B were increased in GC compared to nontumors (P < 0.05). However, reduced levels of acetylated H3K9 and H4K16 were associated with poorly differentiated GC (P < 0.05). Reduced levels of acetylated H3K9 was also associated with diffuse-type histological GC (P < 0.05). Notably, reduced BMP8B mRNA and acetylated H4K16 levels were positively correlated in poorly differentiated GC (P < 0.05). Our study demonstrated that BMP8B seems to be a tumor suppressor gene regulated by H4K16 acetylation in poorly differentiated GC. Therefore, BMP8B may be a potential target for TSA-based therapies in this GC sample subset. J. Cell. Biochem. 118: 869-877, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Fernanda Wisnieski
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, Rua Botucatu, 740, São Paulo 04023900, São Paulo, Brazil
| | - Mariana Ferreira Leal
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, Rua Botucatu, 740, São Paulo 04023900, São Paulo, Brazil.,Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, Rua Borges Lagoa, 783, São Paulo 04038032, São Paulo, Brazil
| | - Danielle Queiroz Calcagno
- Núcleo de Pesquisas em Oncologia, Hospital João de Barros Barreto, Universidade Federal do Pará, Avenida Mundurucus, 4487, Belém 66073000, Pará, Brazil
| | - Leonardo Caires Santos
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, Rua Botucatu, 740, São Paulo 04023900, São Paulo, Brazil
| | - Carolina Oliveira Gigek
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, Rua Botucatu, 740, São Paulo 04023900, São Paulo, Brazil.,Disciplina de Gastroenterologia Cirúrgica, Departamento de Cirurgia, Universidade Federal de São Paulo, Rua Napoleão de Barros, 715, São Paulo 04024002, São Paulo, Brazil
| | - Elizabeth Suchi Chen
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, Rua Botucatu, 740, São Paulo 04023900, São Paulo, Brazil
| | - Ricardo Artigiani
- Departamento de Patologia, Universidade Federal de São Paulo, Rua Botucatu, 740, São Paulo 04023000, São Paulo, Brazil
| | - Sâmia Demachki
- Núcleo de Pesquisas em Oncologia, Hospital João de Barros Barreto, Universidade Federal do Pará, Avenida Mundurucus, 4487, Belém 66073000, Pará, Brazil
| | - Paulo Pimentel Assumpção
- Núcleo de Pesquisas em Oncologia, Hospital João de Barros Barreto, Universidade Federal do Pará, Avenida Mundurucus, 4487, Belém 66073000, Pará, Brazil
| | - Laércio Gomes Lourenço
- Disciplina de Gastroenterologia Cirúrgica, Departamento de Cirurgia, Universidade Federal de São Paulo, Rua Napoleão de Barros, 715, São Paulo 04024002, São Paulo, Brazil
| | - Rommel Rodríguez Burbano
- Laboratório de Citogenética Humana, Instituto de Ciências Biológicas, Universidade Federal do Pará, Rua Augusto Correia, 01, Belém 66075110, Pará, Brazil
| | - Marília Cardoso Smith
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, Rua Botucatu, 740, São Paulo 04023900, São Paulo, Brazil
| |
Collapse
|
27
|
Histone acetyltransferase activity of MOF is required for adult but not early fetal hematopoiesis in mice. Blood 2016; 129:48-59. [PMID: 27827827 DOI: 10.1182/blood-2016-05-714568] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/16/2016] [Indexed: 12/26/2022] Open
Abstract
K(lysine) acetyltransferase 8 (KAT8, also known as MOF) mediates the acetylation of histone H4 at lysine 16 (H4K16ac) and is crucial for murine embryogenesis. Lysine acetyltransferases have been shown to regulate various stages of normal hematopoiesis. However, the function of MOF in hematopoietic stem cell (HSC) development has not yet been elucidated. We set out to study the role of MOF in general hematopoiesis by using a Vav1-cre-induced conditional murine Mof knockout system and found that MOF is critical for hematopoietic cell maintenance and HSC engraftment capacity in adult hematopoiesis. Rescue experiments with a MOF histone acetyltransferase domain mutant illustrated the requirement for MOF acetyltransferase activity in the clonogenic capacity of HSCs and progenitors. In stark contrast, fetal steady-state hematopoiesis at embryonic day (E) 14.5 was not affected by homozygous Mof deletion despite dramatic loss of global H4K16ac. Hematopoietic defects start manifesting in late gestation at E17.5. The discovery that MOF and its H4K16ac activity are required for adult but not early and midgestational hematopoiesis supports the notion that multiple chromatin regulators may be crucial for hematopoiesis at varying stages of development. MOF is therefore a developmental-stage-specific chromatin regulator found to be essential for adult but not early fetal hematopoiesis.
Collapse
|
28
|
Lu L, Chen X, Sanders D, Qian S, Zhong X. High-resolution mapping of H4K16 and H3K23 acetylation reveals conserved and unique distribution patterns in Arabidopsis and rice. Epigenetics 2016; 10:1044-53. [PMID: 26646900 DOI: 10.1080/15592294.2015.1104446] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Histone acetylation and deacetylation are key epigenetic gene regulatory mechanisms that play critical roles in eukaryotes. Acetylation of histone 4 lysine 16 (H4K16ac) is implicated in many cellular processes. However, its biological function and relationship with transcription are largely unexplored in plants. We generated first genome-wide high-resolution maps of H4K16ac in Arabidopsis thaliana and Oryza sativa. We showed that H4K16ac is preferentially enriched around the transcription start sites and positively correlates with gene expression levels. Co-existence of H4K16ac and H3K23ac is correlated with high gene expression levels, suggesting a potentially combinatorial effect of H4K16ac and H3K23ac histone 3 lysine 23 acetylation on gene expression. Our data further revealed that while genes enriched with both H4K16ac and H3K23ac are ubiquitously expressed, genes enriched with only H4K16ac or H3K23ac showed significantly distinct expression patterns in association with particular developmental stages. Unexpectedly, and unlike in Arabidopsis, there are significant levels of both H4K16ac and H3K23ac in the lowly expressed genes in rice. Furthermore, we found that H4K16ac-enriched genes are associated with different biological processes in Arabidopsis and rice, suggesting a potentially species-specific role of H4K16ac in plants. Together, our genome-wide profiling reveals the conserved and unique distribution patterns of H4K16ac and H3K23ac in Arabidopsis and rice and provides a foundation for further understanding their function in plants.
Collapse
Affiliation(s)
- Li Lu
- a Wisconsin Institute for Discovery
| | | | - Dean Sanders
- b Laboratory of Genetics; University of Wisconsin-Madison ; Madison , WI , USA
| | | | - Xuehua Zhong
- a Wisconsin Institute for Discovery.,b Laboratory of Genetics; University of Wisconsin-Madison ; Madison , WI , USA
| |
Collapse
|
29
|
Mujoo K, Butler EB, Pandita RK, Hunt CR, Pandita TK. Pluripotent Stem Cells and DNA Damage Response to Ionizing Radiations. Radiat Res 2016; 186:17-26. [PMID: 27332952 PMCID: PMC4963261 DOI: 10.1667/rr14417.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pluripotent stem cells (PSCs) hold great promise in regenerative medicine, disease modeling, functional genomics, toxicological studies and cell-based therapeutics due to their unique characteristics of self-renewal and pluripotency. Novel methods for generation of pluripotent stem cells and their differentiation to the specialized cell types such as neuronal cells, myocardial cells, hepatocytes and beta cells of the pancreas and many other cells of the body are constantly being refined. Pluripotent stem cell derived differentiated cells, including neuronal cells or cardiac cells, are ideal for stem cell transplantation as autologous or allogeneic cells from healthy donors due to their minimal risk of rejection. Radiation-induced DNA damage, ultraviolet light, genotoxic stress and other intrinsic and extrinsic factors triggers a series of biochemical reactions known as DNA damage response. To maintain genomic stability and avoid transmission of mutations into progenitors cells, stem cells have robust DNA damage response signaling, a contrast to somatic cells. Stem cell transplantation may protect against radiation-induced late effects. In particular, this review focuses on differential DNA damage response between stem cells and derived differentiated cells and the possible pathways that determine such differences.
Collapse
Affiliation(s)
- Kalpana Mujoo
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| | - E. Brian Butler
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| | - Raj K. Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| | - Clayton R. Hunt
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| | - Tej K. Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Weill Cornell Medical College, The Houston Methodist Hospital, 6550 Fannin Street, Houston, TX 77030
| |
Collapse
|
30
|
Hexavalent Chromium (Cr(VI)) Down-Regulates Acetylation of Histone H4 at Lysine 16 through Induction of Stressor Protein Nupr1. PLoS One 2016; 11:e0157317. [PMID: 27285315 PMCID: PMC4902237 DOI: 10.1371/journal.pone.0157317] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/29/2016] [Indexed: 12/14/2022] Open
Abstract
The environmental and occupational carcinogen Hexavalent Chromium (Cr(VI)) has been shown to cause lung cancer in humans when inhaled. In spite of a considerable research effort, the mechanisms of Cr(VI)-induced carcinogenesis remain largely unknown. Nupr1 (nuclear protein 1) is a small, highly basic, and unfolded protein with molecular weight of 8,800 daltons and is induced by a variety of stressors. Studies in animal models have suggested that Nupr1 is a key factor in the development of lung and pancreatic cancers, with little known about the underlying molecular mechanisms. Here we report that the level of Nupr1 is significantly increased in human bronchial epithelial BEAS2B cells following exposure to Cr(VI) through epigenetic mechanisms. Interestingly, Cr(VI) exposure also results in the loss of acetylation at histone H4K16, which is considered a ‘hallmark’ of human cancer. Cr(VI)-induced reduction of H4K16 acetylation appears to be caused by the induction of Nupr1, since (a) overexpression of Nupr1 decreased the levels of both H4K16 acetylation and the histone acetyltransferase MOF (male absent on the first; also known as Kat8, Myst 1), which specifically acetylates H4K16; (b) the loss of acetylation of H4K16 upon Cr(VI) exposure is greatly compromised by knockdown of Nupr1. Moreover, Nupr1-induced reduction of H4K16 acetylation correlates with the transcriptional down-regulation at several genomic loci. Notably, overexpression of Nupr1 induces anchorage-independent cell growth and knockdown of Nupr1 expression prevents Cr(VI)-induced cell transformation. We propose that Cr(VI) induces Nupr1 and rapidly perturbs gene expression by downregulating H4K16 acetylation, thereby contributing to Cr(VI)-induced carcinogenesis.
Collapse
|
31
|
Davis MR, Daggett JJ, Pascual AS, Lam JM, Leyva KJ, Cooper KE, Hull EE. Epigenetically maintained SW13+ and SW13- subtypes have different oncogenic potential and convert with HDAC1 inhibition. BMC Cancer 2016; 16:316. [PMID: 27188282 PMCID: PMC4870788 DOI: 10.1186/s12885-016-2353-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 05/11/2016] [Indexed: 12/19/2022] Open
Abstract
Background The BRM and BRG1 tumor suppressor genes are mutually exclusive ATPase subunits of the SWI/SNF chromatin remodeling complex. The human adrenal carcinoma SW13 cell line can switch between a subtype which expresses these subunits, SW13+, and one that expresses neither subunit, SW13-. Loss of BRM expression occurs post-transcriptionally and can be restored via histone deacetylase (HDAC) inhibition. However, most previously used HDAC inhibitors are toxic and broad-spectrum, providing little insight into the mechanism of the switch between subtypes. In this work, we explore the mechanisms of HDAC inhibition in promoting subtype switching and further characterize the oncogenic potential of the two epigenetically distinct SW13 subtypes. Methods SW13 subtype morphology, chemotaxis, growth rates, and gene expression were assessed by standard immunofluorescence, transwell, growth, and qPCR assays. Metastatic potential was measured by anchorage-independent growth and MMP activity. The efficacy of HDAC inhibitors in inducing subtype switching was determined by immunofluorescence and qPCR. Histone modifications were assessed by western blot. Results Treatment of SW13- cells with HDAC1 inhibitors most effectively promotes re-expression of BRM and VIM, characteristic of the SW13+ phenotype. During treatment, hyperacetylation of histone residues and hypertrimethylation of H3K4 is pronounced. Furthermore, histone modification enzymes, including HDACs and KDM5C, are differentially expressed during treatment but several features of this differential expression pattern differs from that seen in the SW13- and SW13+ subtypes. As the SW13- subtype is more proliferative while the SW13+ subtype is more metastatic, treatment with HDACi increases the metastatic potential of SW13 cells while restoring expression of the BRM tumor suppressor. Conclusions When compared to the SW13- subtype, SW13+ cells have restored BRM expression, increased metastatic capacity, and significantly different expression of a variety of chromatin remodeling factors including those involved with histone acetylation and methylation. These data are consistent with a multistep mechanism of SW13- to SW13+ conversion and subtype stabilization: histone hypermodification results in the altered expression of chromatin remodeling factors and chromatin epigenetic enzymes and the re-expression of BRM which results in restoration of SWI/SNF complex function and leads to changes in chromatin structure and gene expression that stabilize the SW13+ phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2353-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- McKale R Davis
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Juliane J Daggett
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Agnes S Pascual
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Jessica M Lam
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Kathryn J Leyva
- Department of Microbiology and Immunology, Midwestern University, Glendale, AZ, USA
| | - Kimbal E Cooper
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Elizabeth E Hull
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA.
| |
Collapse
|
32
|
Mujoo K, Hunt CR, Horikoshi N, Pandita TK. A multifaceted role for MOF histone modifying factor in genome maintenance. Mech Ageing Dev 2016; 161:177-180. [PMID: 27038808 DOI: 10.1016/j.mad.2016.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/24/2016] [Accepted: 03/28/2016] [Indexed: 11/19/2022]
Abstract
MOF (males absent on the first) was initially identified as a dosage compensation factor in Drosophila that acetylates lysine 16 of histone H4 (H4K16ac) and increased gene transcription from the single copy male X-chromosome. In humans, however, the ortholog of Drosophila MOF has been shown to interact with a range of proteins that extend its potential significance well beyond transcription. For example, recent results indicate MOF is an upstream regulator of the ATM (ataxia-telangiectasia mutated) protein, the loss of which is responsible for ataxia telangiectasia (AT). ATM is a key regulatory kinase that interacts with and phosphorylates multiple substrates that influence critical, cell-cycle control and DNA damage repair pathways in addition to other pathways. Thus, directly or indirectly, MOF may be involved in a wide range of cellular functions. This review will focus on the contribution of MOF to cellular DNA repair and new results that are beginning to examine the in vivo physiological role of MOF.
Collapse
Affiliation(s)
- Kalpana Mujoo
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Clayton R Hunt
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Nobuo Horikoshi
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030, United States
| | - Tej K Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030, United States.
| |
Collapse
|
33
|
Abstract
Loss of function or mutation of the ataxia-telangiectasia mutated gene product (ATM) results in inherited genetic disorders characterized by neurodegeneration, immunodeficiency, and cancer. Ataxia-telangiectasia mutated (ATM) gene product belongs to the PI3K-like protein kinase (PIKKs) family and is functionally implicated in mitogenic signal transduction, chromosome condensation, meiotic recombination, cell-cycle control, and telomere maintenance. The ATM protein kinase is primarily activated in response to DNA double strand breaks (DSBs), the most deleterious form of DNA damage produced by ionizing radiation (IR) or radiomimetic drugs. It is detected at DNA damage sites, where ATM autophosphorylation causes dissociation of the inactive homodimeric form to the activated monomeric form. Interestingly, heat shock can activate ATM independent of the presence of DNA strand breaks. ATM is an integral part of the sensory machinery that detects DSBs during meiosis, mitosis, or DNA breaks mediated by free radicals. These DNA lesions can trigger higher order chromatin reorganization fuelled by posttranslational modifications of histones and histone binding proteins. Our group, and others, have shown that ATM activation is tightly regulated by chromatin modifications. This review summarizes the multiple approaches used to discern the role of ATM and other associated proteins in chromatin modification in response to DNA damage.
Collapse
|
34
|
Adhikary S, Sanyal S, Basu M, Sengupta I, Sen S, Srivastava DK, Roy S, Das C. Selective Recognition of H3.1K36 Dimethylation/H4K16 Acetylation Facilitates the Regulation of All-trans-retinoic Acid (ATRA)-responsive Genes by Putative Chromatin Reader ZMYND8. J Biol Chem 2015; 291:2664-81. [PMID: 26655721 DOI: 10.1074/jbc.m115.679985] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Indexed: 01/25/2023] Open
Abstract
ZMYND8 (zinc finger MYND (Myeloid, Nervy and DEAF-1)-type containing 8), a newly identified component of the transcriptional coregulator network, was found to interact with the Nucleosome Remodeling and Deacetylase (NuRD) complex. Previous reports have shown that ZMYND8 is instrumental in recruiting the NuRD complex to damaged chromatin for repressing transcription and promoting double strand break repair by homologous recombination. However, the mode of transcription regulation by ZMYND8 has remained elusive. Here, we report that through its specific key residues present in its conserved chromatin-binding modules, ZMYND8 interacts with the selective epigenetic marks H3.1K36Me2/H4K16Ac. Furthermore, ZMYND8 shows a clear preference for canonical histone H3.1 over variant H3.3. Interestingly, ZMYND8 was found to be recruited to several developmental genes, including the all-trans-retinoic acid (ATRA)-responsive ones, through its modified histone-binding ability. Being itself inducible by ATRA, this zinc finger transcription factor is involved in modulating other ATRA-inducible genes. We found that ZMYND8 interacts with transcription initiation-competent RNA polymerase II phosphorylated at Ser-5 in a DNA template-dependent manner and can alter the global gene transcription. Overall, our study identifies that ZMYND8 has CHD4-independent functions in regulating gene expression through its modified histone-binding ability.
Collapse
Affiliation(s)
- Santanu Adhikary
- From the Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata-700064 and the Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata-700032, India
| | - Sulagna Sanyal
- From the Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata-700064 and
| | - Moitri Basu
- From the Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata-700064 and
| | - Isha Sengupta
- From the Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata-700064 and
| | - Sabyasachi Sen
- From the Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata-700064 and
| | - Dushyant Kumar Srivastava
- the Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata-700032, India
| | - Siddhartha Roy
- the Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata-700032, India
| | - Chandrima Das
- From the Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata-700064 and
| |
Collapse
|
35
|
Janczar K, Janczar S, Pastorczak A, Mycko K, Paige AJW, Zalewska-Szewczyk B, Wagrowska-Danilewicz M, Danilewicz M, Mlynarski W. Preserved global histone H4 acetylation linked to ETV6-RUNX1 fusion and PAX5 deletions is associated with favorable outcome in pediatric B-cell progenitor acute lymphoblastic leukemia. Leuk Res 2015; 39:1455-61. [PMID: 26520622 DOI: 10.1016/j.leukres.2015.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/14/2015] [Indexed: 10/22/2022]
Abstract
Epigenetic dysregulation is a hallmark of cancer executed by a number of complex processes the most important of which converge on DNA methylation and histone protein modifications. Epigenetic marks are potentially reversible and thus promising drug targets. In the setting of acute lymphoblastic leukemia (ALL) they have been associated with clinicopathological features including risk of relapse or molecular subgroups of the disease. Here, using immunocytochemistry of bone marrow smears from diagnosis, we studied global histone H4 acetylation, whose loss was previously linked to treatment failure in adults with ALL, in pediatric patients. We demonstrate that preserved global histone H4 acetylation is significantly associated with favorable outcome (RFS, EFS, OS) in children with B cell progenitor (BCP) ALL, recapitulating the findings from adult populations. Further, for the first time we demonstrate differential histone H4 acetylation in molecular subclasses of BCP-ALL including cases with ETV6-RUNX1 fusion gene or PAX5 deletion or deletions in genes linked to B cell development. We conclude global histone H4 acetylation is a prognostic marker and a potential therapeutic target in ALL.
Collapse
Affiliation(s)
- K Janczar
- Department of Nephropathology, Medical University of Lodz, Poland
| | - S Janczar
- Department of Paediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, Poland.
| | - A Pastorczak
- Department of Paediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, Poland
| | - K Mycko
- Department of Paediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, Poland
| | - A J W Paige
- Department of Life Science, University of Bedfordshire, UK
| | - B Zalewska-Szewczyk
- Department of Paediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, Poland
| | | | - M Danilewicz
- Department of Nephropathology, Medical University of Lodz, Poland
| | - W Mlynarski
- Department of Paediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, Poland
| |
Collapse
|
36
|
More complex transcriptional regulation and stress response by MOF. Oncogene 2015; 35:2681-3. [PMID: 26434593 DOI: 10.1038/onc.2015.373] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/12/2015] [Indexed: 12/11/2022]
Abstract
MOF (males absent on the first) was initially discovered as a dosage compensation factor that regulates the epigenetic acetylation of histone H4 lysine 16. In this issue, Sheikh et al. demonstrate that MOF expression is not required for normal kidney tissue function but is required for maintaining transcriptional regulation under conditions of stress. This work along with results from previous investigators highlights the importance of the cell lineage-chromatin modification interaction in determining transcriptional programs and physiological outcomes under normal and stress conditions.
Collapse
|
37
|
Riffo-Campos ÁL, Castillo J, Tur G, González-Figueroa P, Georgieva EI, Rodríguez JL, López-Rodas G, Rodrigo MI, Franco L. Nucleosome-specific, time-dependent changes in histone modifications during activation of the early growth response 1 (Egr1) gene. J Biol Chem 2014; 290:197-208. [PMID: 25378406 DOI: 10.1074/jbc.m114.579292] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Histone post-translational modifications and nucleosome remodeling are coordinate events involved in eukaryotic transcriptional regulation. There are relatively few data on the time course with which these events occur in individual nucleosomes. As a contribution to fill this gap, we first describe the nature and time course of structural changes in the nucleosomes -2, -1, and +1 of the murine Egr1 gene upon induction. To initiate the transient activation of the gene, we used the stimulation of MLP29 cells with phorbol esters and the in vivo activation after partial hepatectomy. In both models, nucleosomes -1 and +1 are partially evicted, whereas nucleosomes +1 and -2 slide downstream during transcription. The sliding of the latter nucleosome allows the EGR1 protein to bind its site, resulting in the repression of the gene. To decide whether EGR1 is involved in the sliding of nucleosome -2, Egr1 was knocked down. In the absence of detectable EGR1, the nucleosome still slides and remains downstream longer than in control cells, suggesting that the product of the gene may be rather involved in the returning of the nucleosome to the basal position. Moreover, the presence of eight epigenetic histone marks has been determined at a mononucleosomal level in that chromatin region. H3S10phK14ac, H3K4me3, H3K9me3, and H3K27me3 are characteristic of nucleosome +1, and H3K9ac and H4K16ac are mainly found in nucleosome -1, and H3K27ac predominates in nucleosomes -2 and -1. The temporal changes in these marks suggest distinct functions for some of them, although changes in H3K4me3 may result from histone turnover.
Collapse
Affiliation(s)
- Ángela L Riffo-Campos
- From the Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, 46100 Valencia and Institute of Health Research INCLIVA, 46010 Valencia, Spain
| | - Josefa Castillo
- From the Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, 46100 Valencia and Institute of Health Research INCLIVA, 46010 Valencia, Spain
| | - Gema Tur
- From the Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, 46100 Valencia and
| | - Paula González-Figueroa
- From the Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, 46100 Valencia and
| | - Elena I Georgieva
- From the Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, 46100 Valencia and
| | - José L Rodríguez
- From the Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, 46100 Valencia and
| | - Gerardo López-Rodas
- From the Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, 46100 Valencia and Institute of Health Research INCLIVA, 46010 Valencia, Spain
| | - M Isabel Rodrigo
- From the Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, 46100 Valencia and Institute of Health Research INCLIVA, 46010 Valencia, Spain
| | - Luis Franco
- From the Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, 46100 Valencia and Institute of Health Research INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
38
|
Liu Y, Xing ZB, Wang SQ, Chen S, Liu YK, Li YH, Li YF, Wang YQ, Lu Y, Hu WN, Zhang JH. MDM2-MOF-H4K16ac axis contributes to tumorigenesis induced by Notch. FEBS J 2014; 281:3315-24. [PMID: 24898892 DOI: 10.1111/febs.12863] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 05/29/2014] [Accepted: 05/30/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Yan Liu
- College of Life Sciences; Hebei United University; Tangshan China
- Central Laboratory; Cancer Institute; Tangshan People's Hospital; China
| | - Zhao-Bin Xing
- College of Life Sciences; Hebei United University; Tangshan China
| | - Shu-Qing Wang
- Department of Nephrology; Kailuan General Hospital; Tangshan Hebei China
| | - Su Chen
- School of Life Sciences; Tongji University; Shanghai China
| | - Yan-Kun Liu
- Central Laboratory; Cancer Institute; Tangshan People's Hospital; China
| | - Yu-Hui Li
- Central Laboratory; Cancer Institute; Tangshan People's Hospital; China
| | - Yu-Feng Li
- Central Laboratory; Cancer Institute; Tangshan People's Hospital; China
| | - Ya-Qi Wang
- College of Life Sciences; Hebei United University; Tangshan China
- Central Laboratory; Cancer Institute; Tangshan People's Hospital; China
| | - Yang Lu
- First Hospital of Shi-Jia Zhuang City; China
| | - Wan-Ning Hu
- Central Laboratory; Cancer Institute; Tangshan People's Hospital; China
| | - Jing-Hua Zhang
- Central Laboratory; Cancer Institute; Tangshan People's Hospital; China
| |
Collapse
|
39
|
O'Connor CM, DiMaggio PA, Shenk T, Garcia BA. Quantitative proteomic discovery of dynamic epigenome changes that control human cytomegalovirus (HCMV) infection. Mol Cell Proteomics 2014; 13:2399-410. [PMID: 24987098 DOI: 10.1074/mcp.m114.039792] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
This work represents the first comprehensive quantitative analysis of global histone post-translational modifications (PTMs) from a virus infection, namely human cytomegalovirus (HCMV) infection. We used a nanoLC-MS/MS platform to identify and quantify the dynamic histone H3 and H4 PTMs expressed during HCMV replication in primary fibroblasts. Specifically, we examined the changes in histone PTMs over a 96 h time course to sample the immediate early (IE), early (E), and late (L) stages of viral infection. Several changes in histone H3 and H4 PTMs were observed, including a marked increase in H3K79me2 and H3K27me3K36me2, and a decrease in H4K16ac, highlighting likely epigenetic strategies of transcriptional activation and silencing during HCMV lytic infection. Heavy methyl-SILAC (hm-SILAC) was used to further confirm the histone methylation flux (especially for H3K79) during HCMV infection. We evaluated DOT1L (the H3K79 methyltransferase) mRNA levels in mock and HCMV-infected cells over a 96 h time course, and observed a significant increase in this methyltransferase as early as 24 hpi showing that viral infection up-regulates DOT1L expression, which drives H3K79me2. We then used shRNA to create a DOT1L knockdown cell population, and found that HCMV infection of the knockdown cells resulted in a 10-fold growth defect when compared with infected control cells not subjected to knockdown. This work documents multiple histone PTMs that occur in response to HCMV infection of fibroblasts, and provides a framework for evaluation of the role of epigenetic modifications in the virus-host interaction.
Collapse
Affiliation(s)
- Christine M O'Connor
- From the ‡Department of Molecular Genetics, Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio 44195
| | - Peter A DiMaggio
- §Department of Chemical Engineering, Imperial College London, London, UK, SW7 2AZ
| | - Thomas Shenk
- ¶Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | - Benjamin A Garcia
- ‖Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| |
Collapse
|
40
|
The histone acetyltransferase MOF activates hypothalamic polysialylation to prevent diet-induced obesity in mice. Mol Metab 2014; 3:619-29. [PMID: 25161885 PMCID: PMC4142401 DOI: 10.1016/j.molmet.2014.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/27/2014] [Accepted: 05/28/2014] [Indexed: 12/11/2022] Open
Abstract
Overfeeding causes rapid synaptic remodeling in hypothalamus feeding circuits. Polysialylation of cell surface molecules is a key step in this neuronal rewiring and allows normalization of food intake. Here we examined the role of hypothalamic polysialylation in the long-term maintenance of body weight, and deciphered the molecular sequence underlying its nutritional regulation. We found that upon high fat diet (HFD), reduced hypothalamic polysialylation exacerbated the diet-induced obese phenotype in mice. Upon HFD, the histone acetyltransferase MOF was rapidly recruited on the St8sia4 polysialyltransferase-encoding gene. Mof silencing in the mediobasal hypothalamus of adult mice prevented activation of the St8sia4 gene transcription, reduced polysialylation, altered the acute homeostatic feeding response to HFD and increased the body weight gain. These findings indicate that impaired hypothalamic polysialylation contribute to the development of obesity, and establish a role for MOF in the brain control of energy balance.
Collapse
|
41
|
Maroschik B, Gürtler A, Krämer A, Rößler U, Gomolka M, Hornhardt S, Mörtl S, Friedl AA. Radiation-induced alterations of histone post-translational modification levels in lymphoblastoid cell lines. Radiat Oncol 2014; 9:15. [PMID: 24406105 PMCID: PMC3903440 DOI: 10.1186/1748-717x-9-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 12/23/2013] [Indexed: 12/22/2022] Open
Abstract
Background Radiation-induced alterations in posttranslational histone modifications (PTMs) may affect the cellular response to radiation damage in the DNA. If not reverted appropriately, altered PTM patterns may cause long-term alterations in gene expression regulation and thus lead to cancer. It is therefore important to characterize radiation-induced alterations in PTM patterns and the factors affecting them. Methods A lymphoblastoid cell line established from a normal donor was used to screen for alterations in methylation levels at H3K4, H3K9, H3K27, and H4K20, as well as acetylation at H3K9, H3K56, H4K5, and H4K16, by quantitative Western Blot analysis at 15 min, 1 h and 24 h after irradiation with 2 Gy and 10 Gy. The variability of alterations in acetylation marks was in addition investigated in a panel of lymphoblastoid cell lines with differing radiosensitivity established from lung cancer patients. Results The screening procedure demonstrated consistent hypomethylation at H3K4me3 and hypoacetylation at all acetylation marks tested. In the panel of lymphoblastoid cell lines, however, a high degree of inter-individual variability became apparent. Radiosensitive cell lines showed more pronounced and longer lasting H4K16 hypoacetylation than radioresistant lines, which correlates with higher levels of residual γ-H2AX foci after 24 h. Conclusion So far, the factors affecting extent and duration of radiation-induced histone alterations are poorly defined. The present work hints at a high degree of inter-individual variability and a potential correlation of DNA damage repair capacity and alterations in PTM levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anna A Friedl
- Department of Radiation Oncology, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
42
|
Crosstalk between NSL histone acetyltransferase and MLL/SET complexes: NSL complex functions in promoting histone H3K4 di-methylation activity by MLL/SET complexes. PLoS Genet 2013; 9:e1003940. [PMID: 24244196 PMCID: PMC3828133 DOI: 10.1371/journal.pgen.1003940] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 09/16/2013] [Indexed: 12/30/2022] Open
Abstract
hMOF (MYST1), a histone acetyltransferase (HAT), forms at least two distinct multiprotein complexes in human cells. The male specific lethal (MSL) HAT complex plays a key role in dosage compensation in Drosophila and is responsible for histone H4K16ac in vivo. We and others previously described a second hMOF-containing HAT complex, the non-specific lethal (NSL) HAT complex. The NSL complex has a broader substrate specificity, can acetylate H4 on K16, K5, and K8. The WD (tryptophan-aspartate) repeat domain 5 (WDR5) and host cell factor 1 (HCF1) are shared among members of the MLL/SET (mixed-lineage leukemia/set-domain containing) family of histone H3K4 methyltransferase complexes. The presence of these shared subunits raises the possibility that there are functional links between these complexes and the histone modifications they catalyze; however, the degree to which NSL and MLL/SET influence one another's activities remains unclear. Here, we present evidence from biochemical assays and knockdown/overexpression approaches arguing that the NSL HAT promotes histone H3K4me2 by MLL/SET complexes by an acetylation-dependent mechanism. In genomic experiments, we identified a set of genes including ANKRD2, that are affected by knockdown of both NSL and MLL/SET subunits, suggested they are co-regulated by NSL and MLL/SET complexes. In ChIP assays, we observe that depletion of the NSL subunits hMOF or NSL1 resulted in a significant reduction of both H4K16ac and H3K4me2 in the vicinity of the ANKRD2 transcriptional start site proximal region. However, depletion of RbBP5 (a core component of MLL/SET complexes) only reduced H3K4me2 marks, but not H4K16ac in the same region of ANKRD2, consistent with the idea that NSL acts upstream of MLL/SET to regulate H3K4me2 at certain promoters, suggesting coordination between NSL and MLL/SET complexes is involved in transcriptional regulation of certain genes. Taken together, our results suggest a crosstalk between the NSL and MLL/SET complexes in cells.
Collapse
|